1
|
Sin MK, Cheng Y, Ahmed A, Roseman JM, Dowling NM, Zamrini E. Cerebral Amyloid Angiopathy, Dementia, and Alzheimer Neuropathologic Changes: Findings From the ACT Autopsy Cohort. Neurology 2024; 103:e210009. [PMID: 39481068 PMCID: PMC11527483 DOI: 10.1212/wnl.0000000000210009] [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: 03/29/2024] [Accepted: 09/04/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral amyloid angiopathy (CAA) is common in older adults and is associated with dementia. Less is known whether this association is mediated by Alzheimer disease (AD) neuropathologic changes, the examination of which was the objective of this study. METHODS This was a retrospective cross-sectional examination of the Kaiser Permanente Washington database of the Adult Changes in Thought (ACT) autopsy cohort with information on CAA, dementia, the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) (amyloid neuritic plaques), and Braak (tau neurofibrillary tangles). CAA was diagnosed by immunohistochemistry and dementia by ACT Consensus Diagnostic Conference. AD neuropathology was categorized by CERAD scores and Braak stages. Multivariable logistic regression models were used to estimate odds ratios (ORs) and 95% CIs of the associations of CAA with dementia, adjusting for age at death and sex, and with additional adjustments separately for CERAD scores (moderate-severe vs mild-absent), Braak stages (V-VI vs 0-IV), APOE ε4, and stroke. Formal mediation analyses were conducted to estimate age-sex-adjusted OR (95% CI) for natural indirect effects (NIEs) of CERAD scores and Braak stages. RESULTS The 848 participants had a mean age of 86.7 ± 4.6 years at death, and 57.6% were female. CAA was present in 322 participants (38.0%), of whom 152, 145, and 25 had mild, moderate, and severe CAA, respectively. Dementia was present in 384 participants (45.3%), of whom 317 had AD. Dementia was more common in those with CAA than without (53.7% vs 40.1%; age-sex-adjusted OR 1.57, 95% CI 1.18-2.10). This association remained significant after separate adjustment for other covariates but lost significance when adjusted for CERAD scores (OR 1.27, 95% CI 0.93-1.71) and Braak stages (OR 0.96, 95% CI 0.69-1.33). Findings from our formal mediation analyses show that ORs (95% CIs) for NIE of CERAD scores and Braak stages were 1.25 (1.13-1.37) and 1.63 (1.38-1.88), respectively, and CERAD scores and Braak stages mediated 53% and 111% of the total association, respectively. DISCUSSION We observed a significant association between CAA and dementia that disappeared when adjusted for CERAD or Braak stages. Findings from our mediation analyses suggest that the CAA-dementia association may be potentially mediated by AD neuropathologic changes. This hypothesis needs to be tested in future mechanistic studies in AD accounting for unmeasured confounders.
Collapse
Affiliation(s)
- Mo-Kyung Sin
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Yan Cheng
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Ali Ahmed
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Jeffrey M Roseman
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - N Maritza Dowling
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| | - Edward Zamrini
- From the Seattle University (M.-K.S.), Washington; George Washington University (Y.C., A.A., N.M.D., E.Z.), Washington, DC; DC VA Medical Center (A.A.), Washington, DC; University of Alabama at Birmingham (J.M.R.); Irvine Clinical Research (E.Z.), California
| |
Collapse
|
2
|
Khan WU, Salman M, Ali M, Majid H, Yar MS, Akhtar M, Parvez S, Najmi AK. Neuroprotective Effects of Sulforaphane in a rat model of Alzheimer's Disease induced by Aβ (1-42) peptides. Neurochem Int 2024; 179:105839. [PMID: 39173832 DOI: 10.1016/j.neuint.2024.105839] [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: 05/31/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
The intricate nature of Alzheimer's disease (AD) has presented significant hurdles in the development of effective interventions. Sulforaphane (SFN) is of interest due to its antioxidative, anti-inflammatory, and neuroprotective properties, which could address various aspects of AD pathology. This study explores the potential of SFN in a rat model of AD induced by Aβ (1-42) peptides. AD symptoms were triggered in rats by injecting Aβ (1-42) peptides directly into their cerebral ventricles. SFN (10 mg/kg and 20 mg/kg), Trigonelline (10 mg/kg), and Pioglitazone (10 mg/kg) were administered in Aβ (1-42) treated animals. Behavioral assessments were performed using the Novel Object Recognition tests. Various biochemical parameters, such as soluble Aβ (1-42), IRS-S312, GSK-3β, TNF-α, acetylcholinesterase, nitrite levels, lipid peroxidation, and reduced glutathione activity, were quantified using ELISA kits and spectrophotometric assays. Histopathological analyses included Hematoxylin and Eosin, Crystal Violet, Congo red, and IRS-1 Immunohistochemistry staining. Quantification was performed to assess neuronal loss and Aβ plaque burden. The novelty of this study lies in its comprehensive evaluation of SFN's impact on multiple AD-related pathways at dual doses. The Novel Object Recognition test revealed that SFN, especially at higher doses, improved memory deficits induced by Aβ (1-42). Biochemically, SFN reduced hippocampal Aβ levels, IRS-S312, GSK-3β, TNF-α, and acetylcholinesterase activity, while increasing glutathione levels, all in a dose-dependent manner. Histopathological analyses further confirmed SFN's protective role against Aβ-induced neuronal damage, amyloidosis, and changes in insulin signaling. These results highlight SFN's potential as a multifaceted therapeutic agent for AD, offering a promising avenue for treatment due to its antioxidative, anti-inflammatory, and neuroprotective properties. The inclusion of combination treatments with Trigonelline and Pioglitazone alongside SFN offers insights into potential synergistic effects, which could pave the way for developing combination therapies for AD.
Collapse
Affiliation(s)
- Wasi Uzzaman Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Mubashshir Ali
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Haya Majid
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
3
|
Goswami P, Akhter J, Mangla A, Suramya S, Jindal G, Ahmad S, Raisuddin S. Downregulation of ATF-4 Attenuates the Endoplasmic Reticulum Stress-Mediated Neuroinflammation and Cognitive Impairment in Experimentally Induced Alzheimer's Disease Model. Mol Neurobiol 2024; 61:5071-5082. [PMID: 38159199 DOI: 10.1007/s12035-023-03861-3] [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: 08/27/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Protein aggregation is invariably associated with the inflammation as a factor in Alzheimer's disease (AD). We investigated the interaction between downstream factors of endoplasmic reticulum (ER) stress pathway and inflammation, with implications in cognitive impairment in AD. Amyloid-β (Aβ)(1-42) was administered by bilateral intracerebroventricular (icv) injection in the brain of adult male Wistar rats to experimentally develop AD. The cognitive impairment was assessed by measuring behavioral parameters such as Morris water maze and novel object recognition tests. Levels of pro-inflammatory cytokines such as interleukin (IL)-1β and tumor necrosis factor (TNF)-α and anti-inflammatory cytokines IL-4 and IL-10 were measured by the enzyme-linked immunosorbent assay (ELISA) in different rat brain regions. Inflammatory marker proteins such as cyclo-oxygenase (COX)-2 and phosphorylation of nuclear factor kappa B (NF-КB) (p65) were measured by the western blotting. Gene expression of ER stress downstream factors such as ATF-4, CHOP, and GADD-34 was analyzed by qRT-PCR. Histological studies were performed to check Aβ accumulation and neuronal degeneration. Integrated stress response inhibitor (ISRIB) was used to confirm the specific role of ER stress-mediated inflammation in cognitive impairment. Administration of Aβ(1-42) resulted in alteration in levels of inflammatory cytokines, inflammatory proteins, and mRNA levels of ER stress downstream factors. ISRIB treatment resulted in attenuation of Aβ(1-42)-induced ER stress, inflammation, neurodegeneration, and cognitive impairment in rats. These results indicate that ER stress-mediated inflammation potentiates the cognitive impairment in AD. An understanding of cascade of events, interaction of ER stress which was a hallmark of the present investigation together with inflammation and modulation of downstream signalling factors could serve as potent biomarkers to study AD progression.
Collapse
Affiliation(s)
- Poonam Goswami
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Juheb Akhter
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Anuradha Mangla
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Suramya Suramya
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Garima Jindal
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Shahzad Ahmad
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Sheikh Raisuddin
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India.
| |
Collapse
|
4
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Unveiling the Therapeutic Potential of Kelulut (Stingless Bee) Honey in Alzheimer's Disease: Findings from a Rat Model Study. Antioxidants (Basel) 2024; 13:926. [PMID: 39199172 PMCID: PMC11351951 DOI: 10.3390/antiox13080926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
Alzheimer's disease (AD) poses a major worldwide health challenge because of its profound impact on cognitive abilities and overall well-being. Despite extensive research and numerous clinical trials, therapeutic options remain limited. Our study aimed to investigate the potential of Kelulut honey (KH) as a novel therapeutic agent for addressing the multifactorial pathology of AD. We tried to evaluate the disease-attenuating and neuroprotective potential of KH in the intrahippocampally induced AD rat model by utilizing histochemistry and enzyme-linked immunosorbent assay (ELISA) studies. A total of 26 male Sprague Dawley rats weighing ~280-380 g were randomly divided into three groups: Control, AD-induced (Aβ), and AD-induced and treated with KH (Aβ+KH). The latter two groups underwent stereotaxic surgery, where 6.25 µg of amyloid β1-42 peptides were injected intrahippocampally. One-week post-surgery, KH was administered to the treatment group at a dose of 1 g/kg body weight for a period of four weeks, after which the rats went through behavior tests. After completion of behavior analysis, the rats were sacrificed, and the brains were processed for histochemistry and ELISA studies. The open field test analysis demonstrated that KH improved the locomotion of Aβ+KH compared to Aβ (p = 0.0013). In comparison, the Morris water maze did not show any nootropic effects on cognition with a paradoxical increase in time spent in the target quadrant by the Aβ group (p = 0.029). Histochemical staining showed markedly increased Congo-red-stained amyloid plaques, which were significantly reduced in dentate gyrus of Aβ+KH compared to Aβ (p < 0.05). Moreover, significantly higher apoptosis was seen in the Aβ group compared to Aβ+KH (p < 0.01) and control groups (p < 0.001). Furthermore, the ELISA studies deduced more phosphorylated tau in the diseased group compared to Aβ+KH (p = 0.038) and controls (p = 0.016). These findings suggest that KH consumption for twenty-eight days has the potential to attenuate the pathological burden of disease while exerting neuroprotective effects in rodent models of AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (A.S.); (F.A.); (S.L.T.)
| |
Collapse
|
5
|
Fang S, Zhang K, Liu D, Yang Y, Xi H, Xie W, Diao K, Rao Z, Wang D, Yang W. Polyphenol-based polymer nanoparticles for inhibiting amyloid protein aggregation: recent advances and perspectives. Front Nutr 2024; 11:1408620. [PMID: 39135555 PMCID: PMC11317421 DOI: 10.3389/fnut.2024.1408620] [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: 04/03/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
Polyphenols are a group of naturally occurring compounds that possess a range of biological properties capable of potentially mitigating or preventing the progression of age-related cognitive decline and Alzheimer's disease (AD). AD is a chronic neurodegenerative disease known as one of the fast-growing diseases, especially in the elderly population. Moreover, as the primary etiology of dementia, it poses challenges for both familial and societal structures, while also imposing a significant economic strain. There is currently no pharmacological intervention that has demonstrated efficacy in treating AD. While polyphenols have exhibited potential in inhibiting the pathological hallmarks of AD, their limited bioavailability poses a significant challenge in their therapeutic application. Furthermore, in order to address the therapeutic constraints, several polymer nanoparticles are being explored as improved therapeutic delivery systems to optimize the pharmacokinetic characteristics of polyphenols. Polymer nanoparticles have demonstrated advantageous characteristics in facilitating the delivery of polyphenols across the blood-brain barrier, resulting in their efficient distribution within the brain. This review focuses on amyloid-related diseases and the role of polyphenols in them, in addition to discussing the anti-amyloid effects and applications of polyphenol-based polymer nanoparticles.
Collapse
Affiliation(s)
- Shuzhen Fang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Kangyi Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, Key Laboratory of Food Nutrition and Safety, School of Tea, Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Danqing Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Yulong Yang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Hu Xi
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Wenting Xie
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Ke Diao
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhihong Rao
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Wenming Yang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| |
Collapse
|
6
|
Yu Z, Blade G, Bouley BS, Dobrucki IT, Dobrucki LW, Mirica LM. Coordination Chemistry of Sulfur-Containing Bifunctional Chelators: Toward in Vivo Stabilization of 64Cu PET Imaging Agents for Alzheimer's Disease. Inorg Chem 2023; 62:20820-20833. [PMID: 38060375 PMCID: PMC11400993 DOI: 10.1021/acs.inorgchem.3c02929] [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] [Indexed: 12/19/2023]
Abstract
The broader utilization of 64Cu positron emission tomography (PET) imaging agents has been hindered by the unproductive demetalation induced by bioreductants. To advance the development of 64Cu-based PET imaging tracers for Alzheimer's Disease (AD), there is a need for novel ligand design strategies. In this study, we developed sulfur-containing dithiapyridinophane (N2S2) bifunctional chelators (BFCs) as well as all nitrogen-based diazapyridinophane (N4) BFCs to compare their abilities to chelate Cu and target Aβ aggregates. Through spectrophotometric titrations and electrochemical measurements, we have demonstrated that the N2S2-based BFCs exhibit >10 orders of magnitude higher binding affinity toward Cu(I) compared to their N4-based counterparts, while both types of BFCs exhibit high stability constants toward Cu(II). Notably, solid state structures for both Cu(II) and Cu(I) complexes supported by the two ligand frameworks were obtained, providing molecular insights into their copper chelating abilities. Aβ binding experiments were conducted to study the structure-affinity relationship, and fluorescence microscopy imaging studies confirmed the selective labeling of the BFCs and their copper complexes. Furthermore, we investigated the potential of these ligands for the 64Cu-based PET imaging of AD through radiolabeling and autoradiography studies. We believe our findings provide molecular insights into the design of bifunctional Cu chelators that can effectively stabilize both Cu(II) and Cu(I) and, thus, can have significant implications for the development of 64Cu PET imaging as a diagnostic tool for AD.
Collapse
Affiliation(s)
- Zhengxin Yu
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Glenn Blade
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Bailey S Bouley
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Iwona T Dobrucki
- Beckman Institute for Advanced Science and Technology, Department of Bioengineering, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Lawrence W Dobrucki
- Beckman Institute for Advanced Science and Technology, Department of Bioengineering, University of Illinois at Urbana─Champaign, Urbana, Illinois 61801, United States
| | - Liviu M Mirica
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana─Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
7
|
Elseweidy MM, Mahrous M, Ali SI, Shaheen MA, Younis NN. Pentoxifylline as Add-On Treatment to Donepezil in Copper Sulphate-Induced Alzheimer's Disease-Like Neurodegeneration in Rats. Neurotox Res 2023; 41:546-558. [PMID: 37821782 PMCID: PMC10682165 DOI: 10.1007/s12640-023-00672-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by behavioral, cognitive, and progressive memory impairments. Extensive neuronal loss, extracellular accumulation of insoluble senile amyloid-β (Aβ) plaques, and intracellular neurofibrillary tangles (NFTs) are the major pathological features. The present study aimed to investigate the therapeutic effect of donepezil (DON) and pentoxifylline (PTX) in combination to combat the neurodegenerative disorders (experimental AD) induced by CuSO4 intake in experimental rats. Thirty adult male Wistar rats (140-160 g) were used in this study. AD was first induced in rats by CuSO4 supplement to drinking water (10 mg/L) for 14 weeks. The AD group received no further treatment. Oral treatment with DON (10 mg/kg/day), PTX (100 mg/kg/day), or DON + PTX for the other three groups was started from the 10th week of CuSO4 intake for 4 weeks. Cortex markers like acetylcholine (ACh), acetylcholinesterase (AChE), total antioxidant capacity (TAC), and malondialdehyde (MDA) and hippocampus markers like β-amyloid precursor protein cleaving enzyme 1 (BACE1), phosphorylated Tau (p-tau), Clusterin (CLU), tumor necrosis factor-α (TNF-α), caspase-9 (CAS-9), Bax, and Bcl-2 were measured. The histopathology studies were done by using hematoxylin and eosin and Congo red stains as well as immunohistochemistry for neurofilament. CuSO4 induced adverse histological and biochemical changes. The histological injury in the hippocampus was inhibited following the administration of the DON and PTX. The brain tissue levels of AChE, MDA, BACE1, p-tau, CLU, CAS-9, Bax, and TNF-α were significantly increased, while brain tissue levels of ACh, TAC, and Bcl-2 were significantly decreased in CuSO4-treated rats as compared with the untreated control group. The effects induced by either DON or PTX on most studied parameters were comparable. Combined treatment of DON and PTX induced remarkable results compared with their individual use. However, more clinical and preclinical studies are still required to further confirm and prove the long-term efficacy of such combination.
Collapse
Affiliation(s)
- Mohamed M Elseweidy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Mohamed Mahrous
- Department of Biochemistry, Faculty of Pharmacy, Port-Said University, Port-Said, 42526, Egypt
| | - Sousou I Ali
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed A Shaheen
- Department of Histology and Cell Biology, Faculty of Human Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Nahla N Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
8
|
Yu Z, Moshood Y, Wozniak MK, Patel S, Terpstra K, Llano DA, Dobrucki LW, Mirica LM. Amphiphilic Molecules Exhibiting Zwitterionic Excited-State Intramolecular Proton Transfer and Near-Infrared Emission for the Detection of Amyloid β Aggregates in Alzheimer's Disease. Chemistry 2023; 29:e202302408. [PMID: 37616059 PMCID: PMC10840928 DOI: 10.1002/chem.202302408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 08/25/2023]
Abstract
Chromophores with zwitterionic excited-state intramolecular proton transfer (ESIPT) have been shown to have larger Stock shifts and red-shifted emission wavelengths compared to the conventional π-delocalized ESIPT molecules. However, there is still a dearth of design strategies to expand the current library of zwitterionic ESIPT compounds. Herein, a novel zwitterionic excited-state intramolecular proton transfer system is reported, enabled by addition of 1,4,7-triazacyclononane (TACN) fragments on a dicyanomethylene-4H-pyran (DCM) scaffold. The solvent-dependent steady-state photophysical studies, pKa measurements, and computational analysis strongly support that the ESIPT process is more efficient with two TACN groups attached to the DCM scaffold and not affected by polar protic solvents. Impressively, compound DCM-OH-2-DT exhibits a near-infrared (NIR) emission at 740 nm along with an uncommonly large Stokes shift. Moreover, DCM-OH-2-DT shows high affinity towards soluble amyloid β (Aβ) oligomers in vitro and in 5xFAD mouse brain sections, and we have successfully applied DCM-OH-2-DT for the in vivo imaging of Aβ aggregates and demonstrated its potential use as an early diagnostic agent for AD. Overall, this study can provide a general molecular design strategy for developing new zwitterionic ESIPT compounds with NIR emission in vivo imaging applications.
Collapse
Affiliation(s)
- Zhengxin Yu
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Yusuff Moshood
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Marcin K. Wozniak
- Beckman Institute for Advanced Science and Technology, Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana IL 61801, United States
| | - Shrey Patel
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Karna Terpstra
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Daniel A. Llano
- Beckman Institute for Advanced Science and Technology, Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana IL 61801, United States
| | - Lawrence W. Dobrucki
- Beckman Institute for Advanced Science and Technology, Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana IL 61801, United States
| | - Liviu M. Mirica
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, United States
| |
Collapse
|
9
|
Cozza M, Amadori L, Boccardi V. Exploring cerebral amyloid angiopathy: Insights into pathogenesis, diagnosis, and treatment. J Neurol Sci 2023; 454:120866. [PMID: 37931443 DOI: 10.1016/j.jns.2023.120866] [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: 07/03/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/08/2023]
Abstract
Cerebral Amyloid Angiopathy (CAA) is a neurological disorder characterized by the deposition of amyloid plaques in the walls of cerebral blood vessels. This condition poses significant challenges in terms of understanding its underlying mechanisms, accurate diagnosis, and effective treatment strategies. This article aims to shed light on the complexities of CAA by providing insights into its pathogenesis, diagnosis, and treatment options. The pathogenesis of CAA involves the accumulation of amyloid beta (Aβ) peptides in cerebral vessels, leading to vessel damage, impaired blood flow, and subsequent cognitive decline. Various genetic and environmental factors contribute to the development and progression of CAA, and understanding these factors is crucial for targeted interventions. Accurate diagnosis of CAA often requires advanced imaging techniques, such as magnetic resonance imaging (MRI) or positron emission tomography (PET) scans, to detect characteristic amyloid deposits in the brain. Early and accurate diagnosis enables appropriate management and intervention strategies. Treatment of CAA focuses on preventing further deposition of amyloid plaques, managing associated symptoms, and reducing the risk of complications such as cerebral hemorrhage. Currently, there are no disease-modifying therapies specifically approved for CAA. However, several experimental treatments targeting Aβ clearance and anti-inflammatory approaches are being investigated in clinical trials, offering hope for future therapeutic advancements.
Collapse
Affiliation(s)
| | - Lucia Amadori
- Department of Integration, Intermediate Care Programme, AUSL Bologna, Italy
| | - Virginia Boccardi
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Italy.
| |
Collapse
|
10
|
Sin MK, Zamrini E, Ahmed A, Nho K, Hajjar I. Anti-Amyloid Therapy, AD, and ARIA: Untangling the Role of CAA. J Clin Med 2023; 12:6792. [PMID: 37959255 PMCID: PMC10647766 DOI: 10.3390/jcm12216792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Anti-amyloid therapies (AATs), such as anti-amyloid monoclonal antibodies, are emerging treatments for people with early Alzheimer's disease (AD). AATs target amyloid β plaques in the brain. Amyloid-related imaging abnormalities (ARIA), abnormal signals seen on magnetic resonance imaging (MRI) of the brain in patients with AD, may occur spontaneously but occur more frequently as side effects of AATs. Cerebral amyloid angiopathy (CAA) is a major risk factor for ARIA. Amyloid β plays a key role in the pathogenesis of AD and of CAA. Amyloid β accumulation in the brain parenchyma as plaques is a pathological hallmark of AD, whereas amyloid β accumulation in cerebral vessels leads to CAA. A better understanding of the pathophysiology of ARIA is necessary for early detection of those at highest risk. This could lead to improved risk stratification and the ultimate reduction of symptomatic ARIA. Histopathological confirmation of CAA by brain biopsy or autopsy is the gold standard but is not clinically feasible. MRI is an available in vivo tool for detecting CAA. Cerebrospinal fluid amyloid β level testing and amyloid PET imaging are available but do not offer specificity for CAA vs amyloid plaques in AD. Thus, developing and testing biomarkers as reliable and sensitive screening tools for the presence and severity of CAA is a priority to minimize ARIA complications.
Collapse
Affiliation(s)
- Mo-Kyung Sin
- College of Nursing, Seattle University, Seattle, WA 98122, USA
| | | | - Ali Ahmed
- VA Medical Center, Washington, DC 20242, USA;
| | - Kwangsik Nho
- School of Medicine, Indianna University, Indianapolis, IN 46202, USA;
| | - Ihab Hajjar
- School of Medicine, University of Texas Southwestern, Dallas, TX 75390, USA;
| |
Collapse
|
11
|
Thapa M, Kumari A, Chin CY, Choby JE, Jin F, Bogati B, Chopyk DM, Koduri N, Pahnke A, Elrod EJ, Burd EM, Weiss DS, Grakoui A. Translocation of gut commensal bacteria to the brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555630. [PMID: 37693595 PMCID: PMC10491268 DOI: 10.1101/2023.08.30.555630] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The gut-brain axis, a bidirectional signaling network between the intestine and the central nervous system, is crucial to the regulation of host physiology and inflammation. Recent advances suggest a strong correlation between gut dysbiosis and neurological diseases, however, relatively little is known about how gut bacteria impact the brain. Here, we reveal that gut commensal bacteria can translocate directly to the brain when mice are fed an altered diet that causes dysbiosis and intestinal permeability, and that this also occurs without diet alteration in distinct murine models of neurological disease. The bacteria were not found in other systemic sites or the blood, but were detected in the vagus nerve. Unilateral cervical vagotomy significantly reduced the number of bacteria in the brain, implicating the vagus nerve as a conduit for translocation. The presence of bacteria in the brain correlated with microglial activation, a marker of neuroinflammation, and with neural protein aggregation, a hallmark of several neurodegenerative diseases. In at least one model, the presence of bacteria in the brain was reversible as a switch from high-fat to standard diet resulted in amelioration of intestinal permeability, led to a gradual loss of detectable bacteria in the brain, and reduced the number of neural protein aggregates. Further, in murine models of Alzheimer's disease, Parkinson's disease, and autism spectrum disorder, we observed gut dysbiosis, gut leakiness, bacterial translocation to the brain, and microglial activation. These data reveal a commensal bacterial translocation axis to the brain in models of diverse neurological diseases.
Collapse
Affiliation(s)
- Manoj Thapa
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Anuradha Kumari
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Chui-Yoke Chin
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Jacob E. Choby
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Fengzhi Jin
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Bikash Bogati
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Daniel M. Chopyk
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Nitya Koduri
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Andrew Pahnke
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Elizabeth J. Elrod
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
| | - Eileen M. Burd
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - David S. Weiss
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
12
|
Khayer N, Motamed N, Marashi SA, Goshadrou F. RT-DOb, a switch gene for the gene pair {Csf1r, Milr1}, can influence the onset of Alzheimer's disease by regulating communication between mast cell and microglia. PLoS One 2023; 18:e0288134. [PMID: 37410787 DOI: 10.1371/journal.pone.0288134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
In biology, homeostasis is a central cellular phenomenon that plays a crucial role in survival. The central nervous system (CNS) is controlled by exquisitely sensitive homeostatic mechanisms when facing inflammatory or pathological insults. Mast cells and microglia play a crucial role in CNS homeostasis by eliminating damaged or unnecessary neurons and synapses. Therefore, decoding molecular circuits that regulate CNS homeostasis may lead to more effective therapeutic strategies that specifically target particular subsets for better therapy of Alzheimer's disease (AD). Based on a computational analysis of a microarray dataset related to AD, the H2-Ob gene was previously identified as a potential modulator of the homeostatic balance between mast cells and microglia. Specifically, it plays such a role in the presence of a three-way gene interaction in which the H2-Ob gene acts as a switch in the co-expression relationship of two genes, Csf1r and Milr1. Therefore, the importance of the H2-Ob gene as a potential therapeutic target for AD has led us to experimentally validate this relationship using the quantitative real-time PCR technique. In the experimental investigation, we confirmed that a change in the expression levels of the RT1-DOb gene (the rat ortholog of murine H2-Ob) can switch the co-expression relationship between Csf1r and Milr1. Furthermore, since the RT1-DOb gene is up-regulated in AD, the mentioned triplets might be related to triggering AD.
Collapse
Affiliation(s)
- Nasibeh Khayer
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nasrin Motamed
- Department of Cellular and Molecular Biology, School of Biology, University of Tehran, Tehran, Iran
| | - Sayed-Amir Marashi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Goshadrou
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Sin MK, Cheng Y, Roseman JM, Zamrini E, Ahmed A. Relationships between Cerebral Vasculopathies and Microinfarcts in a Community-Based Cohort of Older Adults. J Clin Med 2023; 12:3807. [PMID: 37298002 PMCID: PMC10253407 DOI: 10.3390/jcm12113807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Cerebral microinfarcts are associated with cognitive impairment and dementia. Small vessel diseases such as cerebral arteriolosclerosis and cerebral amyloid angiography (CAA) have been found to be associated with microinfarcts. Less is known about the associations of these vasculopathies with the presence, numbers, and location of microinfarcts. These associations were examined in the clinical and autopsy data of 842 participants in the Adult Changes in Thought (ACT) study. Both vasculopathies were categorized by severity (none, mild, moderate, and severe) and region (cortical and subcortical). Odds ratios (OR) and 95% CIs for microinfarcts associated with arteriolosclerosis and CAA adjusted for possible modifying covariates such as age at death, sex, blood pressure, APOE genotype, Braak, and CERAD were estimated. 417 (49.5%) had microinfarcts (cortical, 301; subcortical, 249), 708 (84.1%) had cerebral arteriolosclerosis, 320 (38%) had CAA, and 284 (34%) had both. Ors (95% CI) for any microinfarct were 2.16 (1.46-3.18) and 4.63 (2.90-7.40) for those with moderate (n = 183) and severe (n = 124) arteriolosclerosis, respectively. Respective Ors (95% CI) for the number of microinfarcts were 2.25 (1.54-3.30) and 4.91 (3.18-7.60). Similar associations were observed for cortical and subcortical microinfarcts. Ors (95% Cis) for the number of microinfarcts associated with mild (n = 75), moderate (n = 73), and severe (n = 15) amyloid angiopathy were 0.95 (0.66-1.35), 1.04 (0.71-1.52), and 2.05 (0.94-4.45), respectively. Respective Ors (95% Cis) for cortical microinfarcts were 1.05 (0.71-1.56), 1.50 (0.99-2.27), and 1.69 (0.73-3.91). Respective Ors (95% Cis) for subcortical microinfarcts were 0.84 (0.55-1.28), 0.72 (0.46-1.14), and 0.92 (0.37-2.28). These findings suggest a significant association of cerebral arteriolosclerosis with the presence, number, and location (cortical and subcortical) of microinfarcts, and a weak and non-significant association of CAA with each microinfarct, highlighting the need for future research to better understand the role of small vessel diseases in the pathogenesis of cerebral microinfarcts.
Collapse
Affiliation(s)
- Mo-Kyung Sin
- College of Nursing, Seattle University, Seattle, WA 98122, USA
| | - Yan Cheng
- Biomedical Informatics Center, School of Medicine & Health Sciences, George Washington University, Washington, DC 20052, USA; (Y.C.); (E.Z.); (A.A.)
| | - Jeffrey M. Roseman
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Edward Zamrini
- Biomedical Informatics Center, School of Medicine & Health Sciences, George Washington University, Washington, DC 20052, USA; (Y.C.); (E.Z.); (A.A.)
- Division of Neurology, Irvine Clinical Research, Irvine, CA 92614, USA
- Health and Aging, VA Medical Center, Washington, DC 20060, USA
| | - Ali Ahmed
- Biomedical Informatics Center, School of Medicine & Health Sciences, George Washington University, Washington, DC 20052, USA; (Y.C.); (E.Z.); (A.A.)
- Health and Aging, VA Medical Center, Washington, DC 20060, USA
- School of Medicine, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
14
|
Xu Y, Zhang J, Lee HW, Zhang G, Bai Y, Simons M. High-resolution visualization of pial surface vessels by flattened whole mount staining. iScience 2023; 26:106467. [PMID: 37020957 PMCID: PMC10067958 DOI: 10.1016/j.isci.2023.106467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 12/06/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Understanding development of the cerebral vasculature is essential for the central nervous system (CNS) research and therapeutic developments. Here, we developed a simple, convenient, and fast method-the flattened cortex whole mount (FCWM) technique-for imaging of pial cerebral vessels. FCWM involves dissection of the whole cerebral cortex followed by flattening, sectioning and application of CLARITY technology. Compared to conventional methods, FCWM offers several advantages including (1) high-resolution visualization of the whole cortex pial surface vessel structures and distributions; (2) precise localization of a particular blood vessel, allowing observations of a desired blood vessel during normal development or in disease settings; (3) compatibility with confocal imaging. Application of FCWM for examination of cerebral vasculature during postnatal development or in stroke settings allowed us to demonstrate that cerebral blood vessels manifest type-specific maturation and remodeling which are linked to the rate of endothelial proliferation.
Collapse
Affiliation(s)
- Yanying Xu
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jiasheng Zhang
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Heon-Woo Lee
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yongping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Michael Simons
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Faisal M, Aid J, Nodirov B, Lee B, Hickey MA. Preclinical trials in Alzheimer's disease: Sample size and effect size for behavioural and neuropathological outcomes in 5xFAD mice. PLoS One 2023; 18:e0281003. [PMID: 37036878 PMCID: PMC10085059 DOI: 10.1371/journal.pone.0281003] [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] [Received: 09/08/2022] [Accepted: 01/13/2023] [Indexed: 04/11/2023] Open
Abstract
5xFAD transgenic (TG) mice are used widely in AD preclinical trials; however, data on sample sizes are largely unaddressed. We therefore performed estimates of sample sizes and effect sizes for typical behavioural and neuropathological outcome measures in TG 5xFAD mice, based upon data from single-sex (female) groups. Group-size estimates to detect normalisation of TG body weight to WT littermate levels at 5.5m of age were N = 9-15 depending upon algorithm. However, by 1 year of age, group sizes were small (N = 1 -<6), likely reflecting the large difference between genotypes at this age. To detect normalisation of TG open-field hyperactivity to WT levels at 13-14m, group sizes were also small (N = 6-8). Cued learning in the Morris water maze (MWM) was normal in Young TG mice (5m of age). Mild deficits were noted during MWM spatial learning and memory. MWM reversal learning and memory revealed greater impairment, and groups of up to 22 TG mice were estimated to detect normalisation to WT performance. In contrast, Aged TG mice (tested between 13 and 14m) failed to complete the visual learning (non-spatial) phase of MWM learning, likely due to a failure to recognise the platform as an escape. Estimates of group size to detect normalisation of this severe impairment were small (N = 6-9, depending upon algorithm). Other cognitive tests including spontaneous and forced alternation and novel-object recognition either failed to reveal deficits in TG mice or deficits were negligible. For neuropathological outcomes, plaque load, astrocytosis and microgliosis in frontal cortex and hippocampus were quantified in TG mice aged 2m, 4m and 6m. Sample-size estimates were ≤9 to detect the equivalent of a reduction in plaque load to the level of 2m-old TG mice or the equivalent of normalisation of neuroinflammation outcomes. However, for a smaller effect size of 30%, larger groups of up to 21 mice were estimated. In light of published guidelines on preclinical trial design, these data may be used to provide provisional sample sizes and optimise preclinical trials in 5xFAD TG mice.
Collapse
Affiliation(s)
- Mahvish Faisal
- Department of Pharmacology, Institute of Biomedicine and
Translational Medicine, University of Tartu, Tartu, Estonia
| | - Jana Aid
- Department of Pharmacology, Institute of Biomedicine and
Translational Medicine, University of Tartu, Tartu, Estonia
| | - Bekzod Nodirov
- Department of Pharmacology, Institute of Biomedicine and
Translational Medicine, University of Tartu, Tartu, Estonia
| | - Benjamin Lee
- Department of Pharmacology, Institute of Biomedicine and
Translational Medicine, University of Tartu, Tartu, Estonia
| | - Miriam A. Hickey
- Department of Pharmacology, Institute of Biomedicine and
Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
16
|
Mahmoud AMA, Mantawy EM, Wahdan SA, Ammar RM, El-Demerdash E. Vildagliptin restores cognitive function and mitigates hippocampal neuronal apoptosis in cisplatin-induced chemo-brain: Imperative roles of AMPK/Akt/CREB/ BDNF signaling cascades. Biomed Pharmacother 2023; 159:114238. [PMID: 36640673 DOI: 10.1016/j.biopha.2023.114238] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Cisplatin (CP) is a broad-spectrum antineoplastic agent used to treat many human cancers. Nonetheless, most patients receiving CP suffer from cognitive deficits, a phenomenon termed "chemo-brain". Recently, vildagliptin (Vilda), a DPP-4 inhibitor, has demonstrated promising neuroprotective properties against various neurological diseases. Therefore, the present study aims to investigate the potential neuroprotective properties of Vilda against CP-induced neurotoxicity and elucidate the underlying molecular mechanisms. Chemo-brain was induced in Sprague-Dawley rats by i.p injection of CP at a dose of 5 mg/kg once weekly for four weeks. Vilda was administered daily at a dose (10 mg/kg; P.O) for four weeks. The results revealed that Vilda restored the cognitive function impaired by CP, as assessed by the Morris water maze, Y-maze, and passive avoidance tests. Moreover, Vilda alleviated the CP-induced neurodegeneration, as shown by toluidine blue staining, besides markedly reduced amyloid plaque deposition, as evidenced by Congo red staining. Notably, Vilda boosted cholinergic neurotransmission through the downregulation of the acetylcholinesterase enzyme. In addition, the neuroprotective mechanisms of Vilda include diminishing oxidative stress by reducing MDA levels while raising GSH levels and SOD activity, repressing neuronal apoptosis as shown by elevated Bcl-2 levels together with diminished Bax and caspase-3 expressions, inhibiting neuroinflammation as shown by decreased GFAP expression, and finally boosting hippocampal neurogenesis and survival by upregulating expressions of BDNF and PCNA. These effects were mainly mediated by activating AMPK/Akt/CREB signaling cascades. In summary, Vilda can be considered a promising candidate for guarding against CP-induced chemo-brain and neurodegeneration, thus improving the quality of life of cancer patients.
Collapse
Affiliation(s)
- Abdulla M A Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ramy M Ammar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafr-Elsheikh, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
17
|
Thyroid Hormone Transporters MCT8 and OATP1C1 Are Expressed in Pyramidal Neurons and Interneurons in the Adult Motor Cortex of Human and Macaque Brain. Int J Mol Sci 2023; 24:ijms24043207. [PMID: 36834621 PMCID: PMC9965431 DOI: 10.3390/ijms24043207] [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: 11/10/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Monocarboxylate transporter 8 (MCT8) and organic anion transporter polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters that play an important role in the availability of TH for neural cells, allowing their proper development and function. It is important to define which cortical cellular subpopulations express those transporters to explain why MCT8 and OATP1C1 deficiency in humans leads to dramatic alterations in the motor system. By means of immunohistochemistry and double/multiple labeling immunofluorescence in adult human and monkey motor cortices, we demonstrate the presence of both transporters in long-projection pyramidal neurons and in several types of short-projection GABAergic interneurons in both species, suggesting a critical position of these transporters for modulating the efferent motor system. MCT8 is present at the neurovascular unit, but OATP1C1 is only present in some of the large vessels. Both transporters are expressed in astrocytes. OATP1C1 was unexpectedly found, only in the human motor cortex, inside the Corpora amylacea complexes, aggregates linked to substance evacuation towards the subpial system. On the basis of our findings, we propose an etiopathogenic model that emphasizes these transporters' role in controlling excitatory/inhibitory motor cortex circuits in order to understand some of the severe motor disturbances observed in TH transporter deficiency syndromes.
Collapse
|
18
|
Identification of a Novel Wnt Antagonist Based Therapeutic and Diagnostic Target for Alzheimer's Disease Using a Stem Cell-Derived Model. Bioengineering (Basel) 2023; 10:bioengineering10020192. [PMID: 36829686 PMCID: PMC9952699 DOI: 10.3390/bioengineering10020192] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 02/05/2023] Open
Abstract
Currently, all the existing treatments for Alzheimer's disease (AD) fail to stall progression due to longer duration of time between onset of the symptoms and diagnosis of the disease, raising the necessity of effective diagnostics and novel treatment. Specific molecular regulation of the onset and progression of disease is not yet elucidated. This warranted investigation of the role of Wnt signaling regulators which are thought to be involved in neurogenesis. The AD model was established using amyloid beta (Aβ) in human mesenchymal stem cells derived from amniotic membranes which were differentiated into neuronal cell types. In vivo studies were carried out with Aβ or a Wnt antagonist, AD201, belonging to the sFRP family. We further created an AD201-knockdown in vitro model to determine the role of Wnt antagonism. BACE1 upregulation, ChAT and α7nAChR downregulation with synapse and functionality loss with increases in ROS confirmed the neurodegeneration. Reduced β-catenin and increased AD201 expression indicated Wnt/canonical pathway inhibition. Similar results were exhibited in the in vivo study along with AD-associated behavioural and molecular changes. AD201-knockdown rescued neurons from Aβ-induced toxicity. We demonstrated for the first time a role of AD201 in Alzheimer's disease manifestation, which indicates a promising disease target and biomarker.
Collapse
|
19
|
Zhou G, Xiang T, Xu Y, He B, Wu L, Zhu G, Xie J, Yao L, Xiao Z. Fruquintinib/HMPL-013 ameliorates cognitive impairments and pathology in a mouse model of cerebral amyloid angiopathy (CAA). Eur J Pharmacol 2023; 939:175446. [PMID: 36470443 DOI: 10.1016/j.ejphar.2022.175446] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the cerebrovascular amyloid-β (Aβ) accumulation, and always accompanied by Alzheimer's disease (AD). The mechanisms revealing CAA pathogenesis are still unclear, and it is challenging to develop an efficient therapeutic strategy for its treatment. Vascular endothelial growth factor (VEGF) and its receptors including VEGFR-1,-2,-3 activation are involved in Aβ processing, and modulate numerous cellular events associated with central nervous system (CNS) diseases. In the present study, we attempted to explore the regulatory function of fruquintinib (also named as HMPL-013), a highly selective inhibitor of VEGFR-1,-2,-3 tyrosine kinases, on CAA progression in Tg-SwDI mice. Here, we found that HMPL-013-rich diet consumption for 12 months significantly improved the behavioral performances and cerebral blood flow (CBF) of Tg-SwDI mice compared with the vehicle group. Importantly, HMPL-013 administration considerably reduced Aβ1-40 and Aβ1-42 burden in cortex and hippocampus of Tg-SwDI mice through regulating Aβ metabolism process. Congo red staining confirmed Aβ deposition in vessel walls, reflecting CAA formation, which was, however, strongly ameliorated after HMPL-013 treatment. Neuron death, aberrant glial activation and pro-inflammatory response in brain tissues of Tg-SwDI mice were dramatically alleviated after HMPL-013 consumption. More studies showed that the protective effects of HMPL-013 against CAA might be partially attributed to its regulation on the expression of genes associated with blood vasculature. Intriguingly, VEGF and phosphorylated VEGFR-1,-2 protein expression levels were remarkably decreased by HMPL-013 in cortex and hippocampus of Tg-SwDI mice, which were validated in HMPL-013-treated brain vascular endothelial cells (BVECs) under hypoxia. Finally, we found that VEGF-induced human umbilical vein endothelial cells (HUVEC) proliferation and tube formation were strongly abolished upon HMPL-013 exposure. Collectively, all these findings demonstrated that oral administration of HMPL-013 had therapeutic potential against CAA by reducing Aβ deposition, inflammation and neuron death via suppressing VEGF/VEGFR-1,-2 signaling.
Collapse
Affiliation(s)
- Guijuan Zhou
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China; Department of Rehabilitation Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Tao Xiang
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Yan Xu
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Bing He
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Lin Wu
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Guanghua Zhu
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Juan Xie
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Lan Yao
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Zijian Xiao
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China.
| |
Collapse
|
20
|
Swarup O, Barker JL, Watson R, Davis SM, Campbell BCV, Yassi N. Cerebral amyloid angiopathy: clinical presentations and management challenges in the Australian context. Intern Med J 2022. [PMID: 36565446 DOI: 10.1111/imj.15999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 12/12/2022] [Indexed: 12/25/2022]
Abstract
Cerebral amyloid angiopathy (CAA) is a disease with several clinical manifestations. It is characterised by amyloid-beta deposition in cerebral blood vessels, making them prone to bleeding. The incidence of CAA increases with age and may be associated or co-exist with intraparenchymal neurodegenerative proteinopathies, which makes it an increasingly relevant condition for adult physicians in all areas of medical practice. The vast majority of cases of CAA are sporadic with a small minority of familial cases. CAA is asymptomatic in many older adults but increases the risk of fatal intracerebral or subarachnoid haemorrhage. We review the existing literature on CAA and summarise the key findings. We specifically explore clinical challenges relevant to CAA, particularly in diagnosis, management of intracranial haemorrhage and management of concurrent medical conditions.
Collapse
Affiliation(s)
- Oshi Swarup
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - James L Barker
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Rosie Watson
- Department of Geriatrics, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Stephen M Davis
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Bruce C V Campbell
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Nawaf Yassi
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Fazelinejad H, Zahedi E, Nazarian S, Kaffash Siuki Z, Nasri S, Dadmehr M, Mehrabi M, Khodarahmi R. Neuroprotective effect of Bis(Indolyl)phenylmethane in Alzheimer’s disease rat model through inhibition of hen Lysozyme amyloid fibril-induced neurotoxicity. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2022. [DOI: 10.1007/s13738-022-02692-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
22
|
Yu Z, Guo W, Patel S, Cho HJ, Sun L, Mirica LM. Amphiphilic stilbene derivatives attenuate the neurotoxicity of soluble Aβ 42 oligomers by controlling their interactions with cell membranes. Chem Sci 2022; 13:12818-12830. [PMID: 36519059 PMCID: PMC9645390 DOI: 10.1039/d2sc02654f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/19/2022] [Indexed: 10/14/2023] Open
Abstract
The misfolded proteins or polypeptides commonly observed in neurodegenerative diseases, including Alzheimer's disease (AD), are promising drug targets for developing therapeutic agents. To target the amyloid-β (Aβ) peptide plaques and oligomers, the hallmarks of AD, we have developed twelve amphiphilic small molecules with different hydrophobic and hydrophilic fragments. In vitro fluorescence binding assays demonstrate that these amphiphilic compounds show high binding affinity to both Aβ plaques and oligomers, and six of them exhibit selective binding toward Aβ oligomers. These amphiphilic compounds can also label the Aβ species in the brain sections of transgenic AD mice, as shown by immunostaining with an Aβ antibody. Molecular docking studies were performed to obtain structure-affinity relationships. To our delight, four amphiphilic compounds can alleviate the Cu2+-Aβ induced toxicity in cell viability assays. In addition, confocal fluorescence imaging studies provide evidence that two compounds, ZY-15-MT and ZY-15-OMe, can disrupt the interactions between Aβ oligomers and human neuroblastoma SH-SY5Y cell membranes. Overall, these studies strongly suggest that developing compounds with amphiphilic properties that target Aβ oligomers and modulate the Aβ oligomer-cell membrane interactions can be an effective strategy for the development of small molecule AD therapeutics.
Collapse
Affiliation(s)
- Zhengxin Yu
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign 600 S. Mathews Avenue Urbana Illinois 61801 USA
| | - Weijie Guo
- Department of Biochemistry, University of Illinois at Urbana-Champaign 600 S. Mathews Avenue Urbana Illinois 61801 USA
| | - Shrey Patel
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign 600 S. Mathews Avenue Urbana Illinois 61801 USA
| | - Hong-Jun Cho
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign 600 S. Mathews Avenue Urbana Illinois 61801 USA
| | - Liang Sun
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign 600 S. Mathews Avenue Urbana Illinois 61801 USA
| | - Liviu M Mirica
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign 600 S. Mathews Avenue Urbana Illinois 61801 USA
- Hope Center for Neurological Disorders, Washington University School of Medicine St. Louis MO 63110 USA
| |
Collapse
|
23
|
Vitamin B12 as a Cholinergic System Modulator and Blood Brain Barrier Integrity Restorer in Alzheimer's Disease. Eur J Pharm Sci 2022; 174:106201. [PMID: 35523375 DOI: 10.1016/j.ejps.2022.106201] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 12/22/2022]
Abstract
So far, the cholinergic hypothesis of Alzheimer's disease (AD) remains the fundamental explanation for the complex etiopathology of AD. However, therapeutics raising synaptic acetylcholine (Ach) or having cholinergic receptors agonistic activity had shown limited clinical efficacy, possibly, due to lacking capability to aggregate cholinergic receptors within the degenerated cholinergic neurons. Vitamin-B12 (B12) is an epigenetic modifier. It has a specific CNS transport system via the cubam receptors. The later enclose a cholinergic aggregator; agrin protein, suggesting that B12 administration may cause cholinergic receptors aggregation. Further, B12 involvement in homocysteine (Hcy) metabolism may restore blood brain barrier (BBB) integrity disrupted by elevated Hcy levels in AD. Here in, using a pharmacological model of cholinergic amnesia, three different B12 doses were compared to the standard of care; donepezil (DON) regarding cholinergic system modulation, and Hcy metabolic pathways. Further, AD-associated cerebro-vascular pathology was assessed by morphometric analyses of cerebro-vasculature morphology and ultrastructure using scanning and transmission electron-microscopes, respectively. Consequent effect on key AD-hallmarks and behavioral cognitive tests was also examined. The highest B12-tested dose (B12-HD) showed the greatest hippocampal cholinergic modulation with dose-dependent preferential upregulation of one cholinergic receptor over the other. Altered Hcy metabolism was proved to be a consequence of cholinergic disruption that was variably reversed by different B12 doses. In spite of equipotent effect of DON and B12-HD therapies in decreasing β-amyloid synthesis, B12-HD-treated group revealed the greatest restoration of BBB integrity indicating superior capability of β-amyloid clearance. Therefore, B12-HD therapy may represent a promising AD-modifying agent with extra-ability over conventional cholinergic modulators to aggregate cholinergic receptors.
Collapse
|
24
|
Yu ZY, Yi X, Wang YR, Zeng GH, Tan CR, Cheng Y, Sun PY, Liu ZH, Wang YJ, Liu YH. Inhibiting α1-adrenergic receptor signaling pathway ameliorates AD-type pathologies and behavioral deficits in APPswe/PS1 mouse model. J Neurochem 2022; 161:293-307. [PMID: 35244207 DOI: 10.1111/jnc.15603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/25/2022] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Zhong-Yuan Yu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xu Yi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ye-Ran Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Cheng-Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Pu-Yang Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhi-Hao Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
25
|
Evidence for Enhanced Efficacy of Passive Immunotherapy against Beta-Amyloid in CD33-Negative 5xFAD Mice. Biomolecules 2022; 12:biom12030399. [PMID: 35327591 PMCID: PMC8945487 DOI: 10.3390/biom12030399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 12/10/2022] Open
Abstract
Passive immunotherapy is a very promising approach for the treatment of Alzheimer’s disease (AD). Among the different antibodies under development, those targeting post-translationally modified Aβ peptides might combine efficient reduction in beta-amyloid accompanied by lower sequestration in peripheral compartments and thus anticipated and reduced treatment-related side effects. In that regard, we recently demonstrated that the antibody-mediated targeting of isoD7-modified Aβ peptides leads to the attenuation of AD-like amyloid pathology in 5xFAD mice. In order to assess novel strategies to enhance the efficacy of passive vaccination approaches, we investigated the role of CD33 for Aβ phagocytosis in transgenic mice treated with an isoD7-Aβ antibody. We crossbred 5xFAD transgenic mice with CD33 knock out (CD33KO) mice and compared the amyloid pathology in the different genotypes of the crossbreds. The knockout of CD33 in 5xFAD mice leads to a significant reduction in Aβ plaques and concomitant rescue of behavioral deficits. Passive immunotherapy of 5xFAD/CD33KO showed a significant increase in plaque-surrounding microglia compared to 5xFAD treated with the antibody. Additionally, we observed a stronger lowering of Aβ plaque load after passive immunotherapy in 5xFAD/CD33KO mice. The data suggest an additive effect of passive immunotherapy and CD33KO in terms of lowering Aβ pathology. Hence, a combination of CD33 antagonists and monoclonal antibodies might represent a strategy to enhance efficacy of passive immunotherapy in AD.
Collapse
|
26
|
Gazmeh S, Azhir M, Elyasi L, Jahanshahi M, Nikmahzar E, Jameie SB. Apelin-13 protects against memory impairment and neuronal loss, Induced by Scopolamine in male rats. Metab Brain Dis 2022; 37:701-709. [PMID: 34982353 DOI: 10.1007/s11011-021-00882-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/03/2021] [Indexed: 11/30/2022]
Abstract
The present study aimed to evaluate the effects of Apelin-13 on scopolamine-induced memory impairment in rats. Forty male rats were divided into five groups of eight. The control group received no intervention; the scopolamine group underwent stereotaxic surgery and received 3 mg/kg intraperitoneal scopolamine. The treatment groups additionally received 1.25, 2.5 and 5 µg apelin-13 in right lateral ventricles for 7 days. All rats (except the control group) were tested for the passive avoidance reaction, 24 h after the last drug injection. For histological analysis, hippocampal sections were stained with cresyl violet; synaptogenesis biochemical markers were determined by immunoblotting. Apelin-13 alleviated scopolamine-induced passive avoidance memory impairment and neuronal loss in the rats' hippocampus (P<0.001). The reduction observed in mean concentrations of hippocampal synaptic proteins (including neurexin1, neuroligin, and postsynaptic density protein 95) in scopolamine-treated animals was attenuated by apelin-13 treatment. The results demonstrated that apelin-13 can protect against passive avoidance memory deficiency, and neuronal loss, induced by scopolamine in male rats. Further experimental and clinical studies are required to confirm its therapeutic potential in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Gazmeh
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Azhir
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Leila Elyasi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Km 4 Gorgan-Sari Road (Shastcola), Gorgan, Iran.
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Km 4 Gorgan-Sari Road (Shastcola), Gorgan, Iran
| | - Emsehgol Nikmahzar
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyed Behnamedin Jameie
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Yu Z, Chen D, Tan C, Zeng G, He C, Wang J, Bu X, Wang Y. Physiological clearance of Aβ by spleen and splenectomy aggravates Alzheimer-type pathogenesis. Aging Cell 2022; 21:e13533. [PMID: 34939734 PMCID: PMC8761003 DOI: 10.1111/acel.13533] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/26/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022] Open
Abstract
Background A previous study demonstrated that nearly 40%–60% of brain Aβ flows out into the peripheral system for clearance. However, where and how circulating Aβ is cleared in the periphery remains unclear. The spleen acts as a blood filter and an immune organ. The aim of the present study was to investigate the role of the spleen in the clearance of Aβ in the periphery. Methods We investigated the physiological clearance of Aβ by the spleen and established a mouse model of AD and spleen excision by removing the spleens of APP/PS1 mice to investigate the effect of splenectomy on AD mice. Results We found that Aβ levels in the splenic artery were higher than those in the splenic vein, suggesting that circulating Aβ is cleared when blood flows through the spleen. Next, we found that splenic monocytes/macrophages could take up Aβ directly in vivo and in vitro. Splenectomy aggravated behaviour deficits, brain Aβ burden and AD‐related pathologies in AD mice. Conclusion Our study reveals for the first time that the spleen exerts a physiological function of clearing circulating Aβ in the periphery. Our study also suggests that splenectomy, which is a routine treatment for splenic rupture and hypersplenism, might accelerate the development of AD.
Collapse
Affiliation(s)
- Zhong‐Yuan Yu
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Dong‐Wan Chen
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Cheng‐Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Gui‐Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Chen‐Yang He
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience Daping Hospital Third Military Medical University Chongqing China
- Institute of Brain and Intelligence Third Military Medical University Chongqing China
- Chongqing Key Laboratory of Ageing and Brain Diseases Chongqing China
| |
Collapse
|
28
|
Kulichikhin KY, Fedotov SA, Rubel MS, Zalutskaya NM, Zobnina AE, Malikova OA, Neznanov NG, Chernoff YO, Rubel AA. Development of molecular tools for diagnosis of Alzheimer's disease that are based on detection of amyloidogenic proteins. Prion 2021; 15:56-69. [PMID: 33910450 PMCID: PMC8096329 DOI: 10.1080/19336896.2021.1917289] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia that usually occurs among older people. AD results from neuronal degeneration that leads to the cognitive impairment and death. AD is incurable, typically develops over the course of many years and is accompanied by a loss of functional autonomy, making a patient completely dependent on family members and/or healthcare workers. Critical features of AD are pathological polymerization of Aβ peptide and microtubule-associated protein tau, accompanied by alterations of their conformations and resulting in accumulation of cross-β fibrils (amyloids) in human brains. AD apparently progresses asymptomatically for years or even decades before the appearance of symptoms. Therefore, development of the early AD diagnosis at a pre-symptomatic stage is essential for potential therapies. This review is focused on current and potential molecular tools (including non-invasive methods) that are based on detection of amyloidogenic proteins and can be applicable to early diagnosis of AD.Abbreviations: Aβ - amyloid-β peptide; AβO - amyloid-β oligomers; AD - Alzheimer's disease; ADRDA - Alzheimer's Disease and Related Disorders Association; APH1 - anterior pharynx defective 1; APP - amyloid precursor protein; BACE1 - β-site APP-cleaving enzyme 1; BBB - brain blood barrier; CJD - Creutzfeldt-Jakob disease; CRM - certified reference material; CSF - cerebrospinal fluid; ELISA - enzyme-linked immunosorbent assay; FGD - 18F-fluorodesoxyglucose (2-deoxy-2-[18F]fluoro-D-glucose); IP-MS - immunoprecipitation-mass spectrometry assay; MCI - mild cognitive impairment; MDS - multimer detection system; MRI - magnetic resonance imaging; NIA-AA - National Institute on Ageing and Alzheimer's Association; NINCDS - National Institute of Neurological and Communicative Disorders and Stroke; PEN2 - presenilin enhancer 2; PET - positron emission tomography; PiB - Pittsburgh Compound B; PiB-SUVR - PIB standardized uptake value ratio; PMCA - Protein Misfolding Cycling Amplification; PrP - Prion Protein; P-tau - hyperphosphorylated tau protein; RMP - reference measurement procedure; RT-QuIC - real-time quaking-induced conversion; SiMoA - single-molecule array; ThT - thioflavin T; TSEs - Transmissible Spongiform Encephslopathies; T-tau - total tau protein.
Collapse
Affiliation(s)
| | - Sergei A. Fedotov
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
- I.P Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Maria S. Rubel
- SCAMT Institute, ITMO University, St. Petersburg, Russia
| | - Natalia M. Zalutskaya
- V.M. Bekhterev National Research Medical Center for Psychiatry and Neurology, St. Petersburg, Russia
| | - Anastasia E. Zobnina
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
- Sirius University of Science and Technology, Sochi, Russia
| | - Oksana A. Malikova
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
- Sirius University of Science and Technology, Sochi, Russia
| | - Nikolay G. Neznanov
- V.M. Bekhterev National Research Medical Center for Psychiatry and Neurology, St. Petersburg, Russia
| | - Yury O. Chernoff
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Aleksandr A. Rubel
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
- Sirius University of Science and Technology, Sochi, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
29
|
IKK2/NF-κB Activation in Astrocytes Reduces amyloid β Deposition: A Process Associated with Specific Microglia Polarization. Cells 2021; 10:cells10102669. [PMID: 34685649 PMCID: PMC8534251 DOI: 10.3390/cells10102669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that is accompanied by pronounced neuroinflammatory responses mainly characterized by marked microgliosis and astrogliosis. However, it remains open as to how different aspects of astrocytic and microglial activation affect disease progression. Previously, we found that microglia expansion in the spinal cord, initiated by IKK2/NF-κB activation in astrocytes, exhibits stage-dependent beneficial effects on the progression of amyotrophic lateral sclerosis. Here, we investigated the impact of NF-κB-initiated neuroinflammation on AD pathogenesis using the APP23 mouse model of AD in combination with conditional activation of IKK2/NF-κB signaling in astrocytes. We show that NF-κB activation in astrocytes triggers a distinct neuroinflammatory response characterized by striking astrogliosis as well as prominent microglial reactivity. Immunohistochemistry and Congo red staining revealed an overall reduction in the size and number of amyloid plaques in the cerebral cortex and hippocampus. Interestingly, isolated primary astrocytes and microglia cells exhibit specific marker gene profiles which, in the case of microglia, point to an enhanced plaque clearance capacity. In contrast, direct IKK2/NF-κB activation in microglia results in a pro-inflammatory polarization program. Our findings suggest that IKK2/NF-κB signaling in astrocytes may activate paracrine mechanisms acting on microglia function but also on APP processing in neurons.
Collapse
|
30
|
Morales R, Bravo-Alegria J, Moreno-Gonzalez I, Duran-Aniotz C, Gamez N, Edwards Iii G, Soto C. Transmission of cerebral amyloid pathology by peripheral administration of misfolded Aβ aggregates. Mol Psychiatry 2021; 26:5690-5701. [PMID: 34002023 PMCID: PMC8595465 DOI: 10.1038/s41380-021-01150-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 04/02/2021] [Accepted: 04/26/2021] [Indexed: 02/02/2023]
Abstract
Previous reports showed that brain Aβ amyloidosis can be induced in animal models by exogenous administration of pre-formed aggregates. To date, only intra-peritoneal and intra-venous administrations are described as effective means to peripherally accelerate brain Aβ amyloidosis by seeding. Here, we show that cerebral accumulation of Aβ can be accelerated after exposing mouse models of Alzheimer's disease (AD) to Aβ seeds by different peripheral routes of administration, including intra-peritoneal and intra-muscular. Interestingly, animals receiving drops of brain homogenate laden with Aβ seeds in the eyes were efficiently induced. On the contrary, oral administration of large quantities of brain extracts from aged transgenic mice and AD patients did not have any effect in brain pathology. Importantly, pathological induction by peripheral administration of Aβ seeds generated a large proportion of aggregates in blood vessels, suggesting vascular transport. This information highlights the role of peripheral tissues and body fluids in AD-related pathological changes.
Collapse
Affiliation(s)
- Rodrigo Morales
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Centro integrativo de biología y química aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile.
| | - Javiera Bravo-Alegria
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Universidad de los Andes, Facultad de Medicina, Santiago, Chile
| | - Ines Moreno-Gonzalez
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Centro integrativo de biología y química aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga-Instituto de Investigacion Biomedica-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | - Claudia Duran-Aniotz
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Universidad de los Andes, Facultad de Medicina, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Nazaret Gamez
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga-Instituto de Investigacion Biomedica-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | - George Edwards Iii
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Universidad de los Andes, Facultad de Medicina, Santiago, Chile.
| |
Collapse
|
31
|
Genetic deletion of α7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer's disease-like pathology. Prog Neurobiol 2021; 206:102154. [PMID: 34453977 DOI: 10.1016/j.pneurobio.2021.102154] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/29/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
The accumulation of amyloid-beta peptide (Aβ) and the failure of cholinergic transmission are key players in Alzheimer's disease (AD). However, in the healthy brain, Aβ contributes to synaptic plasticity and memory acting through α7 subtype nicotinic acetylcholine receptors (α7nAChRs). Here, we hypothesized that the α7nAChR deletion blocks Aβ physiological function and promotes a compensatory increase in Aβ levels that, in turn, triggers an AD-like pathology. To validate this hypothesis, we studied the age-dependent phenotype of α7 knock out mice. We found that α7nAChR deletion caused an impairment of hippocampal synaptic plasticity and memory at 12 months of age, paralleled by an increase of Amyloid Precursor Protein expression and Aβ levels. This was accompanied by other classical AD features such as a hyperphosphorylation of tau at residues Ser 199, Ser 396, Thr 205, a decrease of GSK-3β at Ser 9, the presence of paired helical filaments and neurofibrillary tangles, neuronal loss and an increase of GFAP-positive astrocytes. Our findings suggest that α7nAChR malfunction might precede Aβ and tau pathology, offering a different perspective to interpret the failure of anti-Aβ therapies against AD and to find novel therapeutical approaches aimed at restoring α7nAChRs-mediated Aβ function at the synapse.
Collapse
|
32
|
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: 1.0] [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
|
33
|
Lipocalin‐Type Prostaglandin
d
Synthase Conjugates as Magnetic Resonance Imaging Contrast Agents for Detecting Amyloid β‐Rich Regions in the Brain of Live Alzheimer's Disease Mice. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
34
|
Salcedo-Arellano MJ, Wang JY, McLennan YA, Doan M, Cabal-Herrera AM, Jimenez S, Wolf-Ochoa MW, Sanchez D, Juarez P, Tassone F, Durbin-Johnson B, Hagerman RJ, Martínez-Cerdeño V. Cerebral Microbleeds in Fragile X-Associated Tremor/Ataxia Syndrome. Mov Disord 2021; 36:1935-1943. [PMID: 33760253 PMCID: PMC10929604 DOI: 10.1002/mds.28559] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Fragile X-associated tremor/ataxia syndrome is a neurodegenerative disease of late onset developed by carriers of the premutation in the fragile x mental retardation 1 (FMR1) gene. Pathological features of neurodegeneration in fragile X-associated tremor/ataxia syndrome include toxic levels of FMR1 mRNA, ubiquitin-positive intranuclear inclusions, white matter disease, iron accumulation, and a proinflammatory state. OBJECTIVE The objective of this study was to analyze the presence of cerebral microbleeds in the brains of patients with fragile X-associated tremor/ataxia syndrome and investigate plausible causes for cerebral microbleeds in fragile X-associated tremor/ataxia syndrome. METHODS We collected cerebral and cerebellar tissue from 15 fragile X-associated tremor/ataxia syndrome cases and 15 control cases carrying FMR1 normal alleles. We performed hematoxylin and eosin, Perls and Congo red stains, ubiquitin, and amyloid β protein immunostaining. We quantified the number of cerebral microbleeds, amount of iron, presence of amyloid β within the capillaries, and number of endothelial cells containing intranuclear inclusions. We evaluated the relationships between pathological findings using correlation analysis. RESULTS We found intranuclear inclusions in the endothelial cells of capillaries and an increased number of cerebral microbleeds in the brains of those with fragile X-associated tremor/ataxia syndrome, both of which are indicators of cerebrovascular dysfunction. We also found a suggestive association between the amount of capillaries that contain amyloid β in the cerebral cortex and the rate of disease progression. CONCLUSION We propose microangiopathy as a pathologic feature of fragile X-associated tremor/ataxia syndrome. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- María Jimena Salcedo-Arellano
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, USA
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Jun Yi Wang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, USA
- Center for Mind and Brain, University of California Davis, Davis, CA, USA
| | - Yingratana A McLennan
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, USA
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
| | - Mai Doan
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Ana Maria Cabal-Herrera
- Group on Congenital Malformations and Dysmorphology, Faculty of Health, Universidad del Valle (MACOS), Cali, Colombia
| | - Sara Jimenez
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Marisol W Wolf-Ochoa
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Desiree Sanchez
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Pablo Juarez
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Flora Tassone
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, UC Davis School of Medicine, Sacramento, CA, USA
| | - Randi J Hagerman
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, USA
| | - Verónica Martínez-Cerdeño
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, Sacramento, CA, USA
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, Sacramento, CA, USA
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
35
|
Sun L, Cho HJ, Sen S, Arango AS, Huynh TT, Huang Y, Bandara N, Rogers BE, Tajkhorshid E, Mirica LM. Amphiphilic Distyrylbenzene Derivatives as Potential Therapeutic and Imaging Agents for Soluble and Insoluble Amyloid β Aggregates in Alzheimer's Disease. J Am Chem Soc 2021; 143:10462-10476. [PMID: 34213901 PMCID: PMC8762579 DOI: 10.1021/jacs.1c05470] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's Disease (AD) is the most common neurodegenerative disease, and efficient therapeutic and early diagnostic agents for AD are still lacking. Herein, we report the development of a novel amphiphilic compound, LS-4, generated by linking a hydrophobic amyloid-binding distyrylbenzene fragment with a hydrophilic triazamacrocycle, which dramatically increases the binding affinity toward various amyloid β (Aβ) peptide aggregates, especially for soluble Aβ oligomers. Moreover, upon the administration of LS-4 to 5xFAD mice, fluorescence imaging of LS-4-treated brain sections reveals that LS-4 can penetrate the blood-brain barrier and bind to the Aβ oligomers in vivo. In addition, the treatment of 5xFAD mice with LS-4 reduces the amount of both amyloid plaques and associated phosphorylated tau aggregates vs the vehicle-treated 5xFAD mice, while microglia activation is also reduced. Molecular dynamics simulations corroborate the observation that introducing a hydrophilic moiety into the molecular structure of LS-4 can enhance the electrostatic interactions with the polar residues of the Aβ species. Finally, exploiting the Cu2+-chelating property of the triazamacrocycle, we performed a series of imaging and biodistribution studies that show the 64Cu-LS-4 complex binds to the amyloid plaques and can accumulate to a significantly larger extent in the 5xFAD mouse brains vs the wild-type controls. Overall, these results illustrate that the novel strategy, to employ an amphiphilic molecule containing a hydrophilic moiety attached to a hydrophobic amyloid-binding fragment, can increase the binding affinity for both soluble and insoluble Aβ aggregates and can thus be used to detect and regulate various Aβ species in AD.
Collapse
Affiliation(s)
- Liang Sun
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Hong-Jun Cho
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Soumyo Sen
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Andres S Arango
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Truc T Huynh
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri 63108, United States
- Department of Chemistry, Washington University, St. Louis, Missouri 63130, United States
| | - Yiran Huang
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Nilantha Bandara
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri 63108, United States
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri 63108, United States
| | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Center for Biophysics and Quantitative Biology and Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Liviu M Mirica
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
36
|
Mitrano DA, Houle SE, Pearce P, Quintanilla RM, Lockhart BK, Genovese BC, Schendzielos RA, Croushore EE, Dymond EM, Bogenpohl JW, Grau HJ, Webb LS. Olfactory dysfunction in the 3xTg-AD model of Alzheimer's disease. IBRO Neurosci Rep 2021; 10:51-61. [PMID: 33842910 PMCID: PMC8019944 DOI: 10.1016/j.ibneur.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/13/2020] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease in which the risk of development increases with age. People with AD are plagued with deficits in their cognition, memory, and basic social skills. Many of these deficits are believed to be caused by the formation of amyloid-β plaques and neurofibrillary tangles in regions of the brain associated with memory, such as the hippocampus. However, one of the early, preclinical symptoms of AD is the loss of olfactory detection and discrimination. To determine if a mouse model of AD expresses the same olfactory dysfunction seen in human AD, 3xTg-AD mice were given a buried food test and, unlike previous studies, compared to their background and parental strains. Results showed that over 52 weeks, the 3xTg-AD mice took significantly longer to find the buried food than the control strains. The olfactory bulbs of the 3xTg-AD mice were removed, sliced, and stained using Congo red for histological analysis. Amyloid deposits were observed predominantly in the granule layer of the olfactory bulb beginning at 13 weeks of age in 3xTg-AD mice, but not in the control strains of mice. Further examination of the buried food test data revealed that 3xTg-AD females had a significantly longer latency to detect the buried food than males beginning at 26 weeks of age. Overall, this study provides further validation of the 3xTg-AD mouse model of AD and supports the idea that simple olfactory testing could be part of the diagnostic process for human AD.
Collapse
Affiliation(s)
- Darlene A. Mitrano
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Sam E. Houle
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Patrick Pearce
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Ricardo M. Quintanilla
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Blakely K. Lockhart
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Benjamin C. Genovese
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Rachel A. Schendzielos
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Emma E. Croushore
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Ethan M. Dymond
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - James W. Bogenpohl
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Harold J. Grau
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| | - Lisa Smith Webb
- Department of Molecular Biology & Chemistry, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
- Program in Neuroscience, Christopher Newport University, 1 Avenue of the Arts, Newport News, VA 23606, USA
| |
Collapse
|
37
|
Koklu A, Wustoni S, Musteata VE, Ohayon D, Moser M, McCulloch I, Nunes SP, Inal S. Microfluidic Integrated Organic Electrochemical Transistor with a Nanoporous Membrane for Amyloid-β Detection. ACS NANO 2021; 15:8130-8141. [PMID: 33784064 PMCID: PMC8158856 DOI: 10.1021/acsnano.0c09893] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/22/2021] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with a severe loss in thinking, learning, and memory functions of the brain. To date, no specific treatment has been proven to cure AD, with the early diagnosis being vital for mitigating symptoms. A common pathological change found in AD-affected brains is the accumulation of a protein named amyloid-β (Aβ) into plaques. In this work, we developed a micron-scale organic electrochemical transistor (OECT) integrated with a microfluidic platform for the label-free detection of Aβ aggregates in human serum. The OECT channel-electrolyte interface was covered with a nanoporous membrane functionalized with Congo red (CR) molecules showing a strong affinity for Aβ aggregates. Each aggregate binding to the CR-membrane modulated the vertical ion flow toward the channel, changing the transistor characteristics. Thus, the device performance was not limited by the solution ionic strength nor did it rely on Faradaic reactions or conformational changes of bioreceptors. The high transconductance of the OECT, the precise porosity of the membrane, and the compactness endowed by the microfluidic enabled the Aβ aggregate detection over eight orders of magnitude wide concentration range (femtomolar-nanomolar) in 1 μL of human serum samples. We expanded the operation modes of our transistors using different channel materials and found that the accumulation-mode OECTs displayed the lowest power consumption and highest sensitivities. Ultimately, these robust, low-power, sensitive, and miniaturized microfluidic sensors helped to develop point-of-care tools for the early diagnosis of AD.
Collapse
Affiliation(s)
- Anil Koklu
- Biological
and Environmental Science and Engineering (BESE), Organic Bioelectronics
Laboratory, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Shofarul Wustoni
- Biological
and Environmental Science and Engineering (BESE), Organic Bioelectronics
Laboratory, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | | | - David Ohayon
- Biological
and Environmental Science and Engineering (BESE), Organic Bioelectronics
Laboratory, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Maximilian Moser
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
| | - Iain McCulloch
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, United Kingdom
- Physical
Science and Engineering Division, KAUST Solar Center (KSC), KAUST, Thuwal 23955-6900, Saudi Arabia
| | - Suzana P. Nunes
- Advanced
Membranes and Porous Materials Center, KAUST,
BESE, Thuwal 23955-6900, Saudi Arabia
| | - Sahika Inal
- Biological
and Environmental Science and Engineering (BESE), Organic Bioelectronics
Laboratory, King Abdullah University of
Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
38
|
Tajmim A, Cuevas-Ocampo AK, Siddique AB, Qusa MH, King JA, Abdelwahed KS, Sonju JJ, El Sayed KA. (-)-Oleocanthal Nutraceuticals for Alzheimer's Disease Amyloid Pathology: Novel Oral Formulations, Therapeutic, and Molecular Insights in 5xFAD Transgenic Mice Model. Nutrients 2021; 13:nu13051702. [PMID: 34069842 PMCID: PMC8157389 DOI: 10.3390/nu13051702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/03/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex progressive neurodegenerative disorder affecting humans mainly through the deposition of Aβ-amyloid (Aβ) fibrils and accumulation of neurofibrillary tangles in the brain. Currently available AD treatments only exhibit symptomatic relief but do not generally intervene with the amyloid and tau pathologies. The extra-virgin olive oil (EVOO) monophenolic secoiridoid S-(–)-oleocanthal (OC) showed anti-inflammatory activity through COX system inhibition with potency comparable to the standard non-steroidal anti-inflammatory drug (NSAID) like ibuprofen. OC also showed positive in vitro, in vivo, and clinical therapeutic effects against cardiovascular diseases, many malignancies, and AD. Due to its pungent, astringent, and irritant taste, OC should be formulated in acceptable dosage form before its oral use as a potential nutraceutical. The objective of this study is to develop new OC oral formulations, assess whether they maintained OC activity on the attenuation of β-amyloid pathology in a 5xFAD mouse model upon 4-month oral dosing use. Exploration of potential OC formulations underlying molecular mechanism is also within this study scope. OC powder formulation (OC-PF) and OC-solid dispersion formulation with erythritol (OC-SD) were prepared and characterized using FT-IR spectroscopy, powder X-ray diffraction, and scanning electron microscopy (ScEM) analyses. Both formulations showed an improved OC dissolution profile. OC-PF and OC-SD improved memory deficits of 5xFAD mice in behavioral studies. OC-PF and OC-SD exhibited significant attenuation of the accumulation of Aβ plaques and tau phosphorylation in the brain of 5xFAD female mice. Both formulations markedly suppressed C3AR1 (complement component 3a receptor 1) activity by targeting the downstream marker STAT3. Collectively, these results demonstrate the potential for the application of OC-PF as a prospective nutraceutical or dietary supplement to control the progression of amyloid pathogenesis associated with AD.
Collapse
Affiliation(s)
- Afsana Tajmim
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Areli K. Cuevas-Ocampo
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA; (A.K.C.-O.); (J.A.K.)
| | - Abu Bakar Siddique
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Mohammed H. Qusa
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Judy Ann King
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA; (A.K.C.-O.); (J.A.K.)
| | - Khaldoun S. Abdelwahed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
| | - Khalid A. El Sayed
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA; (A.T.); (A.B.S.); (M.H.Q.); (K.S.A.); (J.J.S.)
- Correspondence: ; Tel.: +1-318-342-1725
| |
Collapse
|
39
|
Decker Y, Németh E, Schomburg R, Chemla A, Fülöp L, Menger MD, Liu Y, Fassbender K. Decreased pH in the aging brain and Alzheimer's disease. Neurobiol Aging 2021; 101:40-49. [PMID: 33578193 DOI: 10.1016/j.neurobiolaging.2020.12.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/02/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Using publicly available data sets, we compared pH in the human brain and the cerebrospinal fluid (CSF) of postmortem control and Alzheimer's disease cases. We further investigated the effects of long-term acidosis in vivo in the APP-PS1 mouse model of Alzheimer's disease. We finally examined in vitro whether low pH exposure could modulate the release of proinflammatory cytokines and the uptake of amyloid beta by microglia. In the human brain, pH decreased with aging. Similarly, we observed a reduction of pH in the brain of C57BL/6 mice with age. In addition, independent database analyses revealed that postmortem brain and CSF pH is further reduced in Alzheimer's disease cases compared with controls. Moreover, in vivo experiments showed that low pH CSF infusion increased amyloid beta plaque load in APP-PS1 mice. We further observed that mild acidosis reduced the amyloid beta 42-induced release of tumor necrosis factor-alpha by microglia and their capacity to uptake this peptide. Brain acidosis is associated with aging and might affect pathophysiological processes such as amyloid beta aggregation or inflammation in Alzheimer's disease.
Collapse
Affiliation(s)
- Yann Decker
- Department of Neurology, University of the Saarland, Homburg/Saar, Germany; German Institute for Dementia Prevention (DIDP), University of the Saarland, Homburg/Saar, Germany.
| | - Eszter Németh
- Department of Neurology, University of the Saarland, Homburg/Saar, Germany; German Institute for Dementia Prevention (DIDP), University of the Saarland, Homburg/Saar, Germany
| | - Robert Schomburg
- Rehaklinik Zihlschlacht, Neurologisches Rehabilitationszentrum, Zihlschlacht, Switzerland
| | - Axel Chemla
- Department of Neurology, University of the Saarland, Homburg/Saar, Germany; German Institute for Dementia Prevention (DIDP), University of the Saarland, Homburg/Saar, Germany
| | - Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Yang Liu
- Department of Neurology, University of the Saarland, Homburg/Saar, Germany; German Institute for Dementia Prevention (DIDP), University of the Saarland, Homburg/Saar, Germany
| | - Klaus Fassbender
- Department of Neurology, University of the Saarland, Homburg/Saar, Germany; German Institute for Dementia Prevention (DIDP), University of the Saarland, Homburg/Saar, Germany.
| |
Collapse
|
40
|
Rivastigmine attenuates the Alzheimer's disease related protein degradation and apoptotic neuronal death signalling. Biochem J 2021; 478:1435-1451. [PMID: 33660768 DOI: 10.1042/bcj20200754] [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: 09/23/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
Rivastigmine is a clinical drug for patients of Alzheimer's disease (AD) exerting its inhibitory effect on acetylcholinesterase activity however, its effect on other disease-related pathological mechanisms are not yet known. This study was conducted to evaluate the effect of rivastigmine on protein aggregation and degradation related mechanisms employing streptozotocin (STZ) induced experimental rat model. The known inhibitory effect of rivastigmine on cognition and acetylcholinesterase activity was observed in both cortex and hippocampus and further its effect on tau level, amyloid aggregation, biochemical alterations, endoplasmic reticulum (ER) stress, calcium homeostasis, proteasome activity and apoptosis was estimated. STZ administration in rat brain caused significant cognitive impairment, augmented acetylcholinesterase activity, tau phosphorylation and amyloid aggregation which were significantly inhibited with rivastigmine treatment. STZ also caused significant biochemical alterations which were attenuated with rivastigmine treatment. Since AD pathology is related to protein aggregation and we have found disease-related amyloid aggregation, further the investigation was done to decipher the ER functionality and apoptotic signalling. STZ caused significantly altered level of ER stress related markers (GRP78, GADD153 and caspase-12) which were significantly inhibited with rivastigmine treatment. Furthermore, the effect of rivastigmine was estimated on proteasome activity in both regions. Rivastigmine treatment significantly enhances the proteasome activity and may contributes in removal of amyloid aggregation. In conclusion, findings suggested that along with inhibitory effect of rivastigmine on acetylcholinesterase activity and up to some extent on cognition, it has significant effect on disease-related biochemical alterations, ER functionality, protein degradation machinery and neuronal apoptosis.
Collapse
|
41
|
Duran-Aniotz C, Moreno-Gonzalez I, Gamez N, Perez-Urrutia N, Vegas-Gomez L, Soto C, Morales R. Amyloid pathology arrangements in Alzheimer's disease brains modulate in vivo seeding capability. Acta Neuropathol Commun 2021; 9:56. [PMID: 33785065 PMCID: PMC8008576 DOI: 10.1186/s40478-021-01155-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/14/2021] [Indexed: 12/23/2022] Open
Abstract
Amyloid-β (Aβ) misfolding is one of the hallmark pathological features of Alzheimer's disease (AD). AD can manifest with diverse symptomatology including variable rates of cognitive decline, duration of clinical disease, and other detrimental changes. Several reports suggest that conformational diversity in misfolded Aβ is a leading factor for clinical variability in AD, analogous to what it has been described for prion strains in prion diseases. Notably, prion strains generate diverse patterns of misfolded protein deposition in the brains of affected individuals. Here, we tested the in vivo prion-like transmission features of four AD brains displaying particular patterns of amyloidosis. AD brains induced different phenotypes in recipient mice, as evaluated by their specific seeding activity, as well as the total amount of Aβ deposited surrounding vascular structures and the reactivity of amyloid pathology to thioflavin S. Our results support the notion that AD-subtypes are encoded in disease-associated Aβ. Further research exploring whether AD include a spectrum of different clinical conditions or syndromes may pave the way to personalized diagnosis and treatments.
Collapse
Affiliation(s)
- Claudia Duran-Aniotz
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Universidad de los Andes, Facultad de Medicina, Av. San Carlos de Apoquindo, 2200, Las Condes, Santiago, Chile
| | - Ines Moreno-Gonzalez
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, 29010, Malaga, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Nazaret Gamez
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, 29010, Malaga, Spain
| | - Nelson Perez-Urrutia
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
| | - Laura Vegas-Gomez
- Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, 29010, Malaga, Spain
| | - Claudio Soto
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA
- Universidad de los Andes, Facultad de Medicina, Av. San Carlos de Apoquindo, 2200, Las Condes, Santiago, Chile
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, 6431 Fannin, St. Houston, TX, 77030, USA.
- Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile.
| |
Collapse
|
42
|
Shigematsu K, Takeda T, Komori N, Tahara K, Yamagishi H. Hypothesis: Intravenous administration of mesenchymal stem cells is effective in the treatment of Alzheimer's disease. Med Hypotheses 2021; 150:110572. [PMID: 33799163 DOI: 10.1016/j.mehy.2021.110572] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 01/24/2023]
Abstract
We propose the intravenous administration of autologous adipose-derived stem cells as a new treatment for Alzheimer's disease. We hypothesize that the stem cells will secrete neprilysin in the brain to break down and remove amyloid deposits in the Alzheimer's brain. We have shown a case of skin amyloid deposition that disappeared after stem cell administration and confirmed that the stem cells administered had neprilysin activity. In addition to neprilysin secretion, other mechanisms of action of stem cells include nerve regeneration, nerve repair, growth factor secretion, anti-inflammatory effects, and angiogenesis. The harvesting of adipose-derived stem cells is minimally invasive, and intravenous administration can be safely repeated. We hope that the efficacy of this new treatment will be verified and that it will bring a ray of hope to patients suffering from this incurable disease.
Collapse
Affiliation(s)
- Kazuo Shigematsu
- Department of Neurology, Minami Kyoto Hospital, National Hospital Organization, Kyoto, Japan; Nagituji Hospital. Kyoto, Japan.
| | | | | | | | | |
Collapse
|
43
|
Cunha R, Lafeta L, Fonseca EA, Barbosa A, Romano-Silva MA, Vieira R, Jorio A, Malard LM. Nonlinear and vibrational microscopy for label-free characterization of amyloid-β plaques in Alzheimer's disease model. Analyst 2021; 146:2945-2954. [DOI: 10.1039/d1an00074h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Multimodal optical imaging was used for characterization of amyloid-β plaques in mouse brain tissues. We obtained high-resolution images for different biomarkers and investigated vibrational fingerprints that could be used for diagnostic purposes.
Collapse
Affiliation(s)
- Renan Cunha
- Departamento de Física
- ICEx
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Lucas Lafeta
- Departamento de Física
- ICEx
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Emerson A. Fonseca
- Departamento de Física
- ICEx
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Alexandre Barbosa
- Departamento de Física
- ICEx
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Marco A. Romano-Silva
- Departamento de Saúde Mental
- Faculdade de Medicina
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Rafael Vieira
- Departamento de Bioquímica e Imunologia
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Ado Jorio
- Departamento de Física
- ICEx
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Leandro M. Malard
- Departamento de Física
- ICEx
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| |
Collapse
|
44
|
Sacchini S, Díaz-Delgado J, Espinosa de Los Monteros A, Paz Y, Bernaldo de Quirós Y, Sierra E, Arbelo M, Herráez P, Fernández A. Amyloid-beta peptide and phosphorylated tau in the frontopolar cerebral cortex and in the cerebellum of toothed whales: aging versus hypoxia. Biol Open 2020; 9:bio054734. [PMID: 33037014 PMCID: PMC7657478 DOI: 10.1242/bio.054734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/24/2020] [Indexed: 11/26/2022] Open
Abstract
Hypoxia could be a possible risk factor for neurodegenerative alterations in cetaceans' brain. Among toothed whales, the beaked whales are particularly cryptic and routinely dive deeper than 1000 m for about 1 h in order to hunt squids and fishes. Samples of frontal cerebral and cerebellar cortex were collected from nine animals, representing six different species of the suborder Odontoceti. Immunohistochemical analysis employed anti-β-amyloid (Aβ) and anti-neurofibrillary tangle (NFT) antibodies. Six of nine (67%) animals showed positive immunolabeling for Aβ and/or NFT. The most striking findings were intranuclear Aβ immunopositivity in cerebral cortical neurons and NFT immunopositivity in cerebellar Purkinje neurons with granulovacuolar degeneration. Aβ plaques were also observed in one elderly animal. Herein, we present immunohistopathological findings classic of Alzheimer's and other neurodegenerative diseases in humans. Our findings could be linked to hypoxic phenomena, as they were more extensive in beaked whales. Despite their adaptations, cetaceans could be vulnerable to sustained and repetitive brain hypoxia.
Collapse
Affiliation(s)
- Simona Sacchini
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Josué Díaz-Delgado
- Laboratory of Wildlife Comparative Pathology (LAPCOM), School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, 05508-270 SP, Brazil
- Texas A&M Veterinary Medical Diagnostic Laboratory, Pathology Division, College Station, TX 77843, USA
| | - Antonio Espinosa de Los Monteros
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Yania Paz
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Yara Bernaldo de Quirós
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Eva Sierra
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Manuel Arbelo
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Pedro Herráez
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| | - Antonio Fernández
- Veterinary Histology and Pathology, Institute of Animal Health, University of Las Palmas de Gran Canaria, Veterinary School, c/Transmontaña s/n, 35416 Arucas
| |
Collapse
|
45
|
Menet R, Bourassa P, Calon F, ElAli A. Dickkopf-related protein-1 inhibition attenuates amyloid-beta pathology associated to Alzheimer's disease. Neurochem Int 2020; 141:104881. [PMID: 33068684 DOI: 10.1016/j.neuint.2020.104881] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) constitutes the leading cause of dementia worldwide. It is associated to amyloid-β (Aβ) aggregation and tau hyper-phosphorylation, accompanied by a progressive cognitive decline. Evidence suggests that the canonical Wnt pathway is deregulated in AD. Pathway activity is mediated by β-catenin stabilization in the cytosol, and subsequent translocation to the nucleus to regulate the expression of several genes implicated in brain homeostasis and functioning. It was recently proposed that Dickkopf-related protein-1 (DKK1), an endogenous antagonist of the pathway, might be implicated in AD pathogenesis. Here, we hypothesized that canonical Wnt pathway deactivation associated to DKK1 induction contributes to late-onset AD pathogenesis, and thus DKK1 neutralization could attenuate AD pathology. For this purpose, human post-mortem AD brain samples were used to assess pathway activity, and aged APPswe/PS1 mice were used to investigate DKK1 in late-onset AD-like pathology and therapy. Our findings indicate that β-catenin levels progressively decrease in the brain of AD patients, correlating with the duration of symptoms. Next, we found that Aβ pathology in APPswe/PS1 mediates DKK1 induction in the brain. Pharmacological neutralization of DKK1's biological activity in APPswe/PS1 mice restores pathway activity by stabilizing β-catenin, attenuates Aβ pathology, and ameliorates the memory of mice. Attenuation of AD-like pathology upon DKK1 inhibition is accompanied by a reduced protein expression of beta-site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1). Moreover, DKK1 inhibition enhances vascular density, promotes blood-brain barrier (BBB) integrity by increasing claudin 5, glucose transporter-1 (GLUT1), and ATP-binding cassette sub-family B member-1 (ABCB1) protein expression, as well as ameliorates synaptic plasticity by increasing brain-derived neurotrophic factor (BDNF), and postsynaptic density protein-95 (PSD-95) protein expression. DKK1 conditional induction reduces claudin 5, abcb1, and psd-95 mRNA expression, validating its inhibition effects. Our results indicate that neutralization of DKK1's biological activity attenuates AD-like pathology by restoring canonical Wnt pathway activity.
Collapse
Affiliation(s)
- Romain Menet
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Bourassa
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Frédéric Calon
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
46
|
Wang L, Liu S, Xu J, Watanabe N, Mayo KH, Li J, Li X. Emodin inhibits aggregation of amyloid-β peptide 1-42 and improves cognitive deficits in Alzheimer's disease transgenic mice. J Neurochem 2020; 157:1992-2007. [PMID: 32799401 DOI: 10.1111/jnc.15156] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022]
Abstract
Aggregation of amyloid-β peptide 1-42 (Aβ42) initiates the onset of Alzheimer's disease (AD), and all the drugs designed to attenuate AD have failed in clinical trials. Emodin reduces levels of β-amyloid, tau aggregation, oxidative stress, and inflammatory response, demonstrating AD therapeutic potential, whereas its effect on the accumulation of the amyloid-β protein is not well understood. In this work, we investigated emodin activity on Aβ aggregation using a range of biochemical, biophysical, and cell-based approaches. We provide evidence to suggest that emodin blocks Aβ42 fibrillogenesis and Aβ-induced cytotoxicity, displaying a greater effect than that of curcumin. Through adopting three short peptides (Aβ1-16, Aβ17-33, and Aβ28-42), it was proven that emodin interacts with the Leu17-Gly33 sequence. Furthermore, our findings indicated that Val18 and Phe19 in Aβ42 are the target residues with which emodin interacts according amino acid mutation experiments. When fed to 8-month-old B6C3-Tg mice for 2 months, high-dose emodin ameliorates cognitive impairment by 60%-70%. Pathological results revealed that levels of Aβ deposition in the brains of AD mice treated with a high dose of emodin decreased by 50%-70%. Therefore, our study indicates that emodin may represent a promising drug for AD treatment.
Collapse
Affiliation(s)
- Lichun Wang
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Sitong Liu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China.,College of Life Sciences, Jilin University, Changchun, China
| | - Jiaqi Xu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Nobumoto Watanabe
- Bio-Active Compounds Discovery Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Kevin H Mayo
- Biochemistry, Molecular Biology, and Biophysics, college of Biological Science, University of Minnesota, Minneapolis, MN, USA
| | - Jiang Li
- Affiliated Stomatology Hospital of Guangzhong Medical University, Guangzhou, China
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| |
Collapse
|
47
|
Lim JKH, Li QX, He Z, Vingrys AJ, Chinnery HR, Mullen J, Bui BV, Nguyen CTO. Retinal Functional and Structural Changes in the 5xFAD Mouse Model of Alzheimer's Disease. Front Neurosci 2020; 14:862. [PMID: 32903645 PMCID: PMC7438734 DOI: 10.3389/fnins.2020.00862] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease is characterized by the aberrant deposition of protein in the brain and is the leading cause of dementia worldwide. Increasingly, there have been reports of the presence of these protein hallmarks in the retina. In this study, we assayed the retina of 5xFAD mice, a transgenic model of amyloid deposition known to exhibit dementia-like symptoms with age. Using OCT, we found that the retinal nerve fiber layer was thinner in 5xFAD at 6, 12, and 17 months of age compared with wild-type littermates, but the inner plexiform layer was thicker at 6 months old. Retinal function showed reduced ganglion cell responses to light in 5xFAD at 6, 12, and 17 months of age. This functional loss was observed in the outer retina at 17 months of age but not in younger mice. We showed using immunohistochemistry and ELISA that soluble and insoluble amyloid was present in the retina and brain at all ages. In conclusion, we report that amyloid is present in brain and retina of 5xFAD mice and that the pattern of neuronal dysfunction occurs in the inner retina at the early ages and progresses to encompass the outer retina with age. This implies that the inner retina is more sensitive to amyloid changes in early disease and that the outer retina is also affected with disease progression.
Collapse
Affiliation(s)
- Jeremiah K H Lim
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia.,Optometry and Vision Science, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Qiao-Xin Li
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Algis J Vingrys
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Jamie Mullen
- AstraZeneca Neuroscience, Cambridge, MA, United States
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Christine T O Nguyen
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
48
|
Tian J, Guo L, Sui S, Driskill C, Phensy A, Wang Q, Gauba E, Zigman JM, Swerdlow RH, Kroener S, Du H. Disrupted hippocampal growth hormone secretagogue receptor 1α interaction with dopamine receptor D1 plays a role in Alzheimer's disease. Sci Transl Med 2020; 11:11/505/eaav6278. [PMID: 31413143 PMCID: PMC6776822 DOI: 10.1126/scitranslmed.aav6278] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 06/24/2019] [Indexed: 12/12/2022]
Abstract
Hippocampal lesions are a defining pathology of Alzheimer's disease (AD). However, the molecular mechanisms that underlie hippocampal synaptic injury in AD have not been fully elucidated. Current therapeutic efforts for AD treatment are not effective in correcting hippocampal synaptic deficits. Growth hormone secretagogue receptor 1α (GHSR1α) is critical for hippocampal synaptic physiology. Here, we report that GHSR1α interaction with β-amyloid (Aβ) suppresses GHSR1α activation, leading to compromised GHSR1α regulation of dopamine receptor D1 (DRD1) in the hippocampus from patients with AD. The simultaneous application of the selective GHSR1α agonist MK0677 with the selective DRD1 agonist SKF81297 rescued Ghsr1α function from Aβ inhibition, mitigating hippocampal synaptic injury and improving spatial memory in an AD mouse model. Our data reveal a mechanism of hippocampal vulnerability in AD and suggest that a combined activation of GHSR1α and DRD1 may be a promising approach for treating AD.
Collapse
Affiliation(s)
- Jing Tian
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Lan Guo
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shaomei Sui
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.,Department of Neurology, Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong 250014, China
| | - Christopher Driskill
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Qi Wang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.,Department of Neurology, Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong 250014, China
| | - Esha Gauba
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jeffrey M Zigman
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Heng Du
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
49
|
Capsaicin consumption reduces brain amyloid-beta generation and attenuates Alzheimer's disease-type pathology and cognitive deficits in APP/PS1 mice. Transl Psychiatry 2020; 10:230. [PMID: 32661266 PMCID: PMC7359297 DOI: 10.1038/s41398-020-00918-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/17/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of age-related dementia and is currently incurable. The failures of current clinical trials and the establishment of modifiable risk factors have shifted the AD intervention from treatment to prevention in the at-risk population. Previous studies suggest that there is a geographic overlap between AD incidence and spicy food consumption. We previously reported that capsaicin-rich diet consumption was associated with better cognition and lower serum Amyloid-beta (Aβ) levels in people aged 40 years and over. In the present study, we found that intake of capsaicin, the pungent ingredient in chili peppers, reduced brain Aβ burden and rescued cognitive decline in APP/PS1 mice. Our in vivo and in vitro studies revealed that capsaicin shifted Amyloid precursor protein (APP) processing towards α-cleavage and precluded Aβ generation by promoting the maturation of a disintegrin and metalloproteinase 10 (ADAM10). We also found that capsaicin alleviated other AD-type pathologies, such as tau hyperphosphorylation, neuroinflammation and neurodegeneration. The present study suggests that capsaicin is a potential therapeutic candidate for AD and warrants clinical trials on chili peppers or capsaicin as dietary supplementation for the prevention and treatment of AD.
Collapse
|
50
|
Xu CJ, Wang JL, Jing-Pan, Min-Liao. Tph2 Genetic Ablation Contributes to Senile Plaque Load and Astrogliosis in APP/PS1 Mice. Curr Alzheimer Res 2020; 16:219-232. [PMID: 30827242 DOI: 10.2174/1567205016666190301110110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/20/2018] [Accepted: 01/11/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Amyloid-β (Aβ) accumulation plays a critical role in the pathogenesis of Alzheimer's disease (AD) lesions. Deficiency of Serotonin signaling recently has been linked to the increased Aβ level in transgenic mice and humans. In addition, tryptophan hydroxylase-2 (Tph2), a second tryptophan hydroxylase isoform, controls brain serotonin synthesis. However, it remains to be determined that whether Tph2 deficient APP/PS1mice affect the formation of Aβ plaques in vivo. METHODS Both quantitative and qualitative immunochemistry methods, as well as Congo red staining were used to evaluate the Aβ load and astrogliosis in these animals. RESULTS we studied alterations of cortex and hippocampus in astrocytes and senile plaques by Tph2 conditional knockout (Tph2 CKO) AD mice from 6-10 months of age. Using Congo red staining and immunostained with Aβ antibody, we showed that plaques load or plaques numbers significantly increased in Tph2 CKO experimental groups at 8 to 10 months old, compared to wild type (WT) group, respectively. Using GFAP+ astrocytes immunofluorescence method, we found that the density of GFAP+ astrocytes markedly enhanced in Tph2 CKO at 10 months. We showed Aβ plaques co-localized autophagic markers LC3 and p62. Nevertheless, we did not observe any co-localization between GFAP+ astrocytes and autophagic markers, but detected the co-localization between βIII-tubulin+ neurons and autophagic markers. CONCLUSION Overall, our work provides the preliminary evidence in vivo that Tph2 plays a role in amyloid plaques generation.
Collapse
Affiliation(s)
- Chao-Jin Xu
- Department of Histology & Embryology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jun-Ling Wang
- Centre for Reproductive Medicine, Affiliated Hospital 1 of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jing-Pan
- Department of Histology & Embryology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Min-Liao
- Department of Histology & Embryology, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| |
Collapse
|