1
|
Yang D, Su J, Chen Y, Chen G. The NF-κB pathway: Key players in neurocognitive functions and related disorders. Eur J Pharmacol 2024; 984:177038. [PMID: 39369877 DOI: 10.1016/j.ejphar.2024.177038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Perioperative neurocognitive disorder (PND) is a common complication of surgical anesthesia, yet its precise etiology remains unclear. Neuroinflammation is a key feature of PND, influenced by both patient -related and surgical variables. The nuclear factor-κB (NF-κB) transcription factor family plays a critical role in regulating the body's immunological proinflammatory response, which is pivotal in the development of PND. Surgery and anesthesia trigger the activation of the NF-κB signaling pathway, leading to the initiation of inflammatory cascades, disruption of the blood-brain barrier, and neuronal injury. Immune cells and glial cells are central to these pathological processes in PND. Furthermore, this study explores the interactions between NF-κB and various signaling molecules, including Tlr4, P2X, α7-nAChR, ROS, HIF-1α, PI3K/Ak, MicroRNA, Circular RNA, and histone deacetylases, within the context of PND. Targeting NF-κB as a therapeutic approach for PND shows promise as a potential treatment strategy.
Collapse
Affiliation(s)
- Danfeng Yang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Junwei Su
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
2
|
Tan L, Liu Q, Chen S, You R, Li X, Wen T, Peng Z. Neuroprotective effects of all-trans-retinoic acid are mediated via downregulation of TLR4/NF-κB signaling in a rat model of middle cerebral artery occlusion. NEUROSCIENCES (RIYADH, SAUDI ARABIA) 2024; 29:276-283. [PMID: 39379083 PMCID: PMC11460776 DOI: 10.17712/nsj.2024.4.20240010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/14/2024] [Indexed: 10/10/2024]
Abstract
OBJECTIVES To determine the effects of all-trans-retinoic acid (ATRA) on the post-stroke inflammatory response and elucidate the underlying molecular mechanisms. METHODS This animal experiment was conducted at Central Laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China during 2020-2022. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 1.5 h, and treated with ATRA at 2 and 24 h after reperfusion. Neurological deficit scores on behavioral tests, and cerebral infarct volume, microglial polarization, and the expression levels of inflammatory cytokines and proteins associated with TLR4/NF-κB signaling were assessed. RESULTS The ATRA administration reduced cerebral infarct volume and ameliorated neurological deficit scores in MCAO rats. Additionally, ATRA relieved cerebral edema and downregulated the secretion of proinflammatory cytokines after stroke. Finally, ATRA attenuated the polarization of the microglia toward the M1 phenotype and promoted the activation of the beneficial M2 phenotype; the underlying mechanism potentially involved the suppression of the TLR4/NF-κB signaling pathway. CONCLUSION The ATRA treatment promoted functional recovery in an experimental model of ischemic stroke by attenuating neural inflammation. ATRA potentially modulated microglia-mediated neuroinflammation via the downregulation of the TLR4/NF-κB signaling pathway, which makes it a candidate treatment for post-stroke neuroinflammation.
Collapse
Affiliation(s)
- Lixi Tan
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| | - Qian Liu
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| | - Songfa Chen
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| | - Rongjiao You
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| | - Xinyue Li
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| | - Tao Wen
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| | - Zhongxing Peng
- From the Department of Neurology (Tan, Chen, You, Li, Wen, Peng), and from First Affiliated Hospital of Guangdong Pharmaceutical College, and from Liwan Central Hospital of Guangzhou (Tan), Guangzhou, China
| |
Collapse
|
3
|
Alsharief M. How Do Retinoids Affect Alzheimer's Disease and Can They Be Novel Drug Candidates? Cureus 2024; 16:e57548. [PMID: 38572181 PMCID: PMC10990449 DOI: 10.7759/cureus.57548] [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] [Accepted: 04/03/2024] [Indexed: 04/05/2024] Open
Abstract
Alzheimer's disease is a chronic, neurological condition that faces many challenges in its management and therapy nowadays highlighting the importance and urgent need of researching new ways of approaching this disease. Retinoic acid and its derivatives, collectively known as the retinoids, are considered promising agents that have disease-modifying properties in affecting Alzheimer's disease. This thesis aims to address the research questions of what the role of retinoids is in Alzheimer's disease, and whether they can be used as a novel drug candidate for treating this condition. Retinoids' properties and agonistic actions on the nuclear receptors retinoic acid receptor (RAR) and retinoic X receptor (RXR) affect various pathways as well as their underlying genetic factors that compose important pathophysiological hallmarks causing the progression of Alzheimer's disease as amyloid β (Aβ) production and deposition, neurofibrillary tangle (NFT) formation and phosphorylation, and inflammatory and autoimmune responses. Retinoic acid inhibits the amplification of these pathways and modifies the disease progression in animal models, proposing a solid basis for human trials. Hence, investigating retinoids as pharmacological agents in human trials has been conducted, and several synthetic analogues have been developed to address issues concerning retinoic acid's instability and short half-life, as well as adverse drug reactions. The most prominent of these analogues is tamibarotene, a stable retinoic acid derivative with a higher half-life, higher specificity to target receptors, and fewer adverse reactions. A number of criteria that explain what a novel drug candidate should have when managing Alzheimer's disease have been formulated, and which also explain why most novel drug candidates other than retinoic acid have failed in achieving clinical results. Most of these candidates share one common trait which is a single-target approach in targeting disease pathways. This means that when administering these agents, their actions are to target a single disease-causing pathway at a time but do not affect other pathways. On the other hand, tamibarotene is a novel drug candidate that targets a range of pathways at once and provides a more comprehensive approach in its pharmacological actions.
Collapse
Affiliation(s)
- Mazen Alsharief
- International Postgraduate Medical Training Scheme (IPGMTS), University Hospitals Birmingham, Birmingham, GBR
| |
Collapse
|
4
|
Singh A, Kumar Singh N. Pre-clinical Evidence-based Neuroprotective Potential of Naringin against Alzheimer's Disease-like Pathology: A Comprehensive Review. Curr Pharm Biotechnol 2024; 25:1112-1123. [PMID: 37526460 DOI: 10.2174/1389201024666230801095526] [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: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023]
Abstract
Neurodegenerative disorders (NDs) are a group of progressive, chronic, and disabling disorders that are highly prevalent and the incidence is on a constant rise globally. Alzheimer's disease (AD), one of the most common neurodegenerative disorders is hallmarked by cognitive impairment, amyloid-β (Aβ) deposition, hyperphosphorylation of tau protein, cholinergic dysfunction, mitochondrial toxicity, and neurodegeneration. Available therapeutic agents only provide symptomatic relief and their use are limited due to serious side effects. Recent research has recognized flavonoids as potential multi-target biomolecules that can reduce the pathogenesis of AD. Naringin, a natural citrus flavonoid has been traditionally used to treat various NDs including AD, and has gained special attention because exhibits a neuroprotective effect by affecting numerous signaling pathways with minimum adverse effects. Naringin reduces deposition of Aβ, hyperphosphorylation of tau protein, cholinergic dysfunction, oxidative stress burden, mitochondrial toxicity, the activity of glutamate receptors, and apoptosis of the neuronal cells. Additionally, it reduces the expression of phosphorylated-P38/P38 and the NF-κB signaling pathway, showing that a wide range of molecular targets is involved in naringin's neuroprotective action. The present study describes the possible pharmacological targets, signaling pathways, and molecular mechanisms of naringin involved in neuroprotection against AD-like pathology. Based on the above pre-clinical reports it can be concluded that naringin could be an alternative therapeutic agent for the management of AD-like manifestation. Thus, there is a strong recommendation to perform more preclinical and clinical studies to develop naringin as a novel molecule that could be a multi-target drug to counteract AD.
Collapse
Affiliation(s)
- Ashini Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Niraj Kumar Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| |
Collapse
|
5
|
Takano-Kawabe K, Matoba K, Nakamura Y, Moriyama M. Low Density Lipoprotein Receptor-related Protein 2 Expression and Function in Cultured Astrocytes and Microglia. Neurochem Res 2024; 49:199-211. [PMID: 37702891 DOI: 10.1007/s11064-023-04022-7] [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: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
Activation of glial cells, astrocytes and microglia, has been observed in neurodegenerative diseases including Alzheimer's disease (AD). Amyloid β (Aβ), which is aggregated and the aggregation is detected as characteristic pathology in AD brain, is known to be produced by neurons and to activate glial cells. Clearance of Aβ from the brain via active transport system is important to prevent the accumulation and aggregation. Low density lipoprotein receptor-related protein 2 (LRP2/megalin) is an Aβ transporter. However, expression and contribution of LRP2 in astrocytes and microglia remain to be clarified. In the present study, we examined the expression of LRP2 and its roles in cultured astrocytes prepared from rat embryonic brain cortex and mouse microglial cell line BV-2. Both cultured rat astrocytes and BV-2 cells expressed LRP2 mRNA detected by RT-PCR. When lipopolysaccharide (LPS) or all-trans retinoic acid (ATRA) were added to BV-2 cells, LRP2 mRNA expression and uptake of microbeads, Aβ and insulin were increased. On the other hand, LPS decreased LRP2 expression and uptake of Aβ and insulin in cultured astrocytes. Knockdown of LRP2 using siRNA attenuated the LPS- or ATRA-increased uptake of microbeads, Aβ and insulin in BV-2 cells. These results suggest that LRP2 was expressed in both astrocytes and microglia and might be involved in endocytosis activities. Adequate control of LRP2 expression and function in astrocytes and microglia might regulate Aβ and insulin levels in brain and would be a potential target in AD pathology.
Collapse
Affiliation(s)
- Katsura Takano-Kawabe
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan.
| | - Kazuyuki Matoba
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| | - Yoichi Nakamura
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| | - Mitsuaki Moriyama
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| |
Collapse
|
6
|
Est CB, Murphy RM. An in vitro model for vitamin A transport across the human blood-brain barrier. eLife 2023; 12:RP87863. [PMID: 37934575 PMCID: PMC10629827 DOI: 10.7554/elife.87863] [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] [Indexed: 11/08/2023] Open
Abstract
Vitamin A, supplied by the diet, is critical for brain health, but little is known about its delivery across the blood-brain barrier (BBB). Brain microvascular endothelial-like cells (BMECs) differentiated from human-derived induced pluripotent stem cells (iPSCs) form a tight barrier that recapitulates many of the properties of the human BBB. We paired iPSC-derived BMECs with recombinant vitamin A serum transport proteins, retinol-binding protein (RBP), and transthyretin (TTR), to create an in vitro model for the study of vitamin A (retinol) delivery across the human BBB. iPSC-derived BMECs display a strong barrier phenotype, express key vitamin A metabolism markers, and can be used for quantitative modeling of retinol accumulation and permeation. Manipulation of retinol, RBP, and TTR concentrations, and the use of mutant RBP and TTR, yielded novel insights into the patterns of retinol accumulation in, and permeation across, the BBB. The results described herein provide a platform for deeper exploration of the regulatory mechanisms of retinol trafficking to the human brain.
Collapse
Affiliation(s)
- Chandler B Est
- Department of Chemical and Biological Engineering, University of WisconsinMadisonUnited States
| | - Regina M Murphy
- Department of Chemical and Biological Engineering, University of WisconsinMadisonUnited States
| |
Collapse
|
7
|
Almaguer J, Hindle A, Lawrence JJ. The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer's Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue. Antioxidants (Basel) 2023; 12:1921. [PMID: 38001775 PMCID: PMC10669734 DOI: 10.3390/antiox12111921] [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: 08/31/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 11/26/2023] Open
Abstract
There is accumulating evidence that vitamin A (VA) deficiency contributes to the pathogenesis and progression of Alzheimer's disease (AD). All-trans retinoic acid (ATRA), a metabolite of VA in the brain, serves distinct roles in the human hippocampus. Agonists of retinoic acid receptors (RAR), including ATRA, promote activation of the non-amyloidogenic pathway by enhancing expression of α-secretases, providing a mechanistic basis for delaying/preventing amyloid beta (Aβ) toxicity. However, whether ATRA is actually deficient in the hippocampi of patients with AD is not clear. Here, using a publicly available human transcriptomic dataset, we evaluated the extent to which ATRA-sensitive genes are dysregulated in hippocampal tissue from post-mortem AD brains, relative to age-matched controls. Consistent with ATRA deficiency, we found significant dysregulation of many ATRA-sensitive genes and significant upregulation of RAR co-repressors, supporting the idea of transcriptional repression of ATRA-mediated signaling. Consistent with oxidative stress and neuroinflammation, Nrf2 and NfkB transcripts were upregulated, respectively. Interestingly, transcriptional targets of Nrf2 were not upregulated, accompanied by upregulation of several histone deacetylases. Overall, our investigation of ATRA-sensitive genes in the human hippocampus bolsters the scientific premise of ATRA depletion in AD and that epigenetic factors should be considered and addressed as part of VA supplementation.
Collapse
Affiliation(s)
- Joey Almaguer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Ashly Hindle
- Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, Garrison Institute on Aging, Center of Excellence for Translational Neuroscience and Therapeutics, and Center of Excellence for Integrated Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
8
|
Nakanishi S, Kinoshita K, Kurauchi Y, Seki T, Kimura Y, Suzuki M, Suzuki K, Koyama H, Kagechika H, Katsuki H. Acyclic retinoid peretinoin reduces hemorrhage-associated brain injury in vitro and in vivo. Eur J Pharmacol 2023; 954:175899. [PMID: 37392831 DOI: 10.1016/j.ejphar.2023.175899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/03/2023]
Abstract
Peretinoin is an acyclic retinoid that stimulates retinoic acid receptors (NR1Bs) and produces therapeutic effects on hepatocellular cancer. We have previously shown that NR1B agonists such as Am80 and all trans-retinoic acid suppress pathogenic events in intracerebral hemorrhage. The present study addressed the actions of peretinoin and Am80 against cytotoxicity of a blood protease thrombin on cortico-striatal slice cultures obtained from neonatal rat brains. Application of 100 U/ml thrombin to the slice cultures for 72 h caused cell death in the cortical region and tissue shrinkage in the striatal region. Peretinoin (50 μM) and Am80 (1 μM) counteracted these cytotoxic effects of thrombin, and the effect of peretinoin and Am80 was blocked by LE540, an NR1B antagonist. A broad-spectrum kinase inhibitor K252a (3 μM) attenuated the cytoprotective effect of peretinoin in the cortical region, whereas a specific protein kinase A inhibitor KT5720 (1 μM) attenuated the protective effect of peretinoin in the cortical and the striatal regions. On the other hand, nuclear factor-κB (NF-κB) inhibitors such as pyrrolidine dithiocarbamate (50 μM) and Bay11-7082 (10 μM) prevented thrombin-induced shrinkage of the striatal region. Peretinoin and Am80 as well as Bay11-7082 blocked thrombin-induced nuclear translocation of NF-κB in striatal microglia and loss of striatal neurons. We also found that daily administration of peretinoin reduced histopathological injury and alleviated motor deficits in a mouse model of intracerebral hemorrhage. These results indicate that NR1B agonists including peretinoin may serve as a therapeutic option for hemorrhagic brain injury.
Collapse
Affiliation(s)
- Sakino Nakanishi
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keita Kinoshita
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahiro Seki
- Department of Pharmacology, School of Pharmacy, Himeji-Dokkyo University, Hyogo, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Masaaki Suzuki
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Keiichi Suzuki
- Field of Biological Molecular Sciences, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Hiroko Koyama
- Field of Biological Molecular Sciences, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan; Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Hiroyuki Kagechika
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
9
|
Abstract
Retinoic acid (RA) is a metabolite of vitamin A and is essential for development and growth as well as cellular metabolism. Through genomic and nongenomic actions, RA regulates a variety of physiological functions. Dysregulation of RA signaling is associated with many diseases. Targeting RA signaling has been proven valuable to human health. All-trans retinoic acid (AtRA) and anthracycline-based chemotherapy are the standard treatment of acute promyelocytic leukemia (APL). Both human and animal studies have shown a significant relationship between RA signaling and the development and progression of nonalcoholic fatty liver disease (NAFLD). In this review article, we will first summarize vitamin A metabolism and then focus on the role of RA signaling in NAFLD. AtRA inhibits the development and progression of NAFLD via regulating lipid metabolism, inflammation, thermogenesis, etc.
Collapse
Affiliation(s)
- Fathima N Cassim Bawa
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA 44272
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA 44272
| |
Collapse
|
10
|
Jakaria M, Belaidi AA, Bush AI, Ayton S. Vitamin A metabolites inhibit ferroptosis. Biomed Pharmacother 2023; 164:114930. [PMID: 37236031 DOI: 10.1016/j.biopha.2023.114930] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Vitamin A (retinol) is a lipid-soluble vitamin that acts as a precursor for several bioactive compounds, such as retinaldehyde (retinal) and isomers of retinoic acid. Retinol and all-trans-retinoic acid (atRA) penetrate the blood-brain barrier and are reported to be neuroprotective in several animal models. We characterised the impact of retinol and its metabolites, all-trans-retinal (atRAL) and atRA, on ferroptosis-a programmed cell death caused by iron-dependent phospholipid peroxidation. Ferroptosis was induced by erastin, buthionine sulfoximine or RSL3 in neuronal and non-neuronal cell lines. We found that retinol, atRAL and atRA inhibited ferroptosis with a potency superior to α-tocopherol, the canonical anti-ferroptotic vitamin. In contrast, we found that antagonism of endogenous retinol with anhydroretinol sensitises ferroptosis induced in neuronal and non-neuronal cell lines. Retinol and its metabolites atRAL and atRA directly interdict lipid radicals in ferroptosis since these compounds displayed radical trapping properties in a cell-free assay. Vitamin A, therefore, complements other anti-ferroptotic vitamins, E and K; metabolites of vitamin A, or agents that alter their levels, may be potential therapeutics for diseases where ferroptosis is implicated.
Collapse
Affiliation(s)
- Md Jakaria
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdel A Belaidi
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Scott Ayton
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
11
|
Est CB, Murphy RM. An in vitro model for vitamin A transport across the human blood-brain barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536348. [PMID: 37090623 PMCID: PMC10120720 DOI: 10.1101/2023.04.11.536348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Vitamin A, supplied by the diet, is critical for brain health, but little is known about its delivery across the blood-brain barrier (BBB). Brain microvascular endothelial-like cells (BMECs) differentiated from human-derived induced pluripotent stem cells (iPSC) form a tight barrier that recapitulates many of the properties of the human BBB. We paired iPSC-derived BMECs with recombinant vitamin A serum transport proteins, retinol binding protein (RBP) and transthyretin (TTR), to create an in vitro model for the study of vitamin A (retinol) delivery across the human BBB. iPSC-derived BMECs display a strong barrier phenotype, express key vitamin A metabolism markers and can be used for quantitative modeling of retinol accumulation and permeation. Manipulation of retinol, RBP and TTR concentrations, and the use of mutant RBP and TTR, yielded novel insights into the patterns of retinol accumulation in, and permeation across, the BBB. The results described herein provide a platform for deeper exploration of the regulatory mechanisms of retinol trafficking to the human brain.
Collapse
Affiliation(s)
| | - Regina M. Murphy
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison 1415 Engineering Dr., Madison, WI 53706
| |
Collapse
|
12
|
Huang X, Wang YJ, Xiang Y. Bidirectional communication between brain and visceral white adipose tissue: Its potential impact on Alzheimer's disease. EBioMedicine 2022; 84:104263. [PMID: 36122553 PMCID: PMC9490488 DOI: 10.1016/j.ebiom.2022.104263] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
A variety of axes between brain and abdominal organs have been reported, but the interaction between brain and visceral white adipose tissue (vWAT) remains unclear. In this review, we summarized human studies on the association between brain and vWAT, and generalized their interaction and the underlying mechanisms according to animal and cell experiments. On that basis, we come up with the concept of the brain-vWAT axis (BVA). Furthermore, we analyzed the potential mechanisms of involvement of BVA in the pathogenesis of Alzheimer's disease (AD), including vWAT-derived fatty acids, immunological properties of vWAT, vWAT-derived retinoic acid and vWAT-regulated insulin resistance. The proposal of BVA may expand our understanding to some extent of how the vWAT impacts on brain health and diseases, and provide a novel approach to study the pathogenesis and treatment strategies of neurodegenerative disorders.
Collapse
|
13
|
Liu J, Lin Y, Yang Y, Guo Y, Shang Y, Zhou B, Liu T, Fan J, Wei C. Z-Guggulsterone attenuates cognitive defects and decreases neuroinflammation in APPswe/PS1dE9 mice through inhibiting the TLR4 signaling pathway. Biochem Pharmacol 2022; 202:115149. [PMID: 35714682 DOI: 10.1016/j.bcp.2022.115149] [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: 04/01/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022]
Abstract
Growing evidence indicates that inflammatory damage is implicated in the pathogenesis of Alzheimer's disease (AD). Z-Guggulsterone (Z-GS) is a natural steroid, which is extracted from Commiphora mukul and has anti-inflammatory effects in vivo and in vitro. In the present study, we investigated the disease-modifying effects of chronic Z-GS administration on the cognitive and neuropathological impairments in the transgenic mouse models of AD. We found that chronic Z-GS administration prevented learning and memory deficits in the APPswe/PS1dE9 mice. In addition, Z-GS treatment significantly decreased cerebral amyloid-β (Aβ) levels and plaque burden via inhibiting amyloid precursor protein (APP) processing by reducing beta-site APP cleaving enzyme 1 (BACE1) expression in the APPswe/PS1dE9 mice. We also found that Z-GS treatment markedly alleviated neuroinflammation and reduced synaptic defects in the APPswe/PS1dE9 mice. Furthermore, the activated TLR4/NF-κB signaling pathways in APPswe/PS1dE9 mice were remarkably inhibited by Z-GS treatment, which was achieved via suppressing the phosphorylation of JNK. Collectively, our data demonstrate that chronic Z-GS treatment restores cognitive defects and reverses multiple neuropathological impairments in the APPswe/PS1dE9 mice. This study provides novel insights into the neuroprotective effects and neurobiological mechanisms of Z-GS on AD, indicating that Z-GS is a promising disease-modifying agent for the treatment of AD.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Geriatrics, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Ye Lin
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yang Yang
- Department of Neurology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yane Guo
- Department of Neurology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yanchang Shang
- Department of Neurology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Zhou
- Department of Neurology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Tianlong Liu
- Department of Clinical Pharmacy, the 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou 730050, China
| | - Jiao Fan
- Institute of Geriatrics, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| | - Chao Wei
- Department of Neurology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
14
|
Yi-Bin W, Xiang L, Bing Y, Qi Z, Fei-Tong J, Minghong W, Xiangxiang Z, Le K, Yan L, Ping S, Yufei G, Ye X, Chun-Yan W. Inhibition of the CEBPβ-NFκB interaction by nanocarrier-packaged Carnosic acid ameliorates glia-mediated neuroinflammation and improves cognitive function in an Alzheimer's disease model. Cell Death Dis 2022; 13:318. [PMID: 35393391 PMCID: PMC8989877 DOI: 10.1038/s41419-022-04765-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/10/2022] [Accepted: 03/25/2022] [Indexed: 01/09/2023]
Abstract
Neuroinflammation occurs early in Alzheimer’s disease (AD). The initial stage of AD is related to glial dysfunction, which contributes to impairment of Aβ clearance and disruption of synaptic connection. CEBPβ, a member of the CCAAT-enhancer-binding protein (CEBP) family, modulates the expression of inflammation-associated genes, and its expression is elevated in brains undergoing degeneration and injured brains. However, the mechanism underlying CEBPβ-mediated chronic inflammation in AD is unclear. In this study, we observed that increases in the levels of nuclear CEBPβ facilitated the interaction of CEBPβ with the NFκB p65 subunit, increasing the transcription of proinflammatory cytokines in the APP/PS1 mouse brain. Oral administration of nanocarrier-packaged carnosic acid (CA) reduced the aberrant activation of microglia and astrocytes and diminished mature IL-1β, TNFα and IL-6 production in the APP/PS1 mouse brain. CA administration reduced β-amyloid (Aβ) deposition and ameliorated cognitive impairment in APP/PS1 mice. We observed that CA blocked the interaction of CEBPβ with NFκB p65, and chromatin immunoprecipitation revealed that CA reduced the transcription of the NFκB target genes TNFα and IL-6. We confirmed that CA alleviated inflammatory mediator-induced neuronal degeneration and reduced Aβ secretion by inhibiting the CEBPβ-NFκB signalling pathway in vitro. Sulfobutyl ether-beta-cyclodextrin (SBEβCD) was used as the encapsulation agent for the CA-loaded nanocarrier to overcome the poor water solubility and enhance the brain bioavailability of CA. The CA nanoparticles (NPs) had no obvious toxicity. We demonstrated a feasible SBEβCD-based nanodelivery system targeting the brain. Our data provide experimental evidence that CA-loaded NPs are potential therapeutic agents for AD treatment.
Collapse
Affiliation(s)
- Wang Yi-Bin
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Li Xiang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Yang Bing
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Zhang Qi
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Jiao Fei-Tong
- Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China
| | - Wang Minghong
- Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China
| | - Zhang Xiangxiang
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Kang Le
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Li Yan
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Sui Ping
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Gao Yufei
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Xu Ye
- Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China.
| | - Wang Chun-Yan
- Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang, 110122, China. .,Translational Medicine Laboratory, School of Basic Medical Sciences, Jilin Medical University, Jilin, 132013, China.
| |
Collapse
|
15
|
Vitória JJM, Trigo D, da Cruz E Silva OAB. Revisiting APP secretases: an overview on the holistic effects of retinoic acid receptor stimulation in APP processing. Cell Mol Life Sci 2022; 79:101. [PMID: 35089425 PMCID: PMC11073327 DOI: 10.1007/s00018-021-04090-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide and is characterized by the accumulation of the β-amyloid peptide (Aβ) in the brain, along with profound alterations in phosphorylation-related events and regulatory pathways. The production of the neurotoxic Aβ peptide via amyloid precursor protein (APP) proteolysis is a crucial step in AD development. APP is highly expressed in the brain and is complexly metabolized by a series of sequential secretases, commonly denoted the α-, β-, and γ-cleavages. The toxicity of resulting fragments is a direct consequence of the first cleaving event. β-secretase (BACE1) induces amyloidogenic cleavages, while α-secretases (ADAM10 and ADAM17) result in less pathological peptides. Hence this first cleavage event is a prime therapeutic target for preventing or reverting initial biochemical events involved in AD. The subsequent cleavage by γ-secretase has a reduced impact on Aβ formation but affects the peptides' aggregating capacity. An array of therapeutic strategies are being explored, among them targeting Retinoic Acid (RA) signalling, which has long been associated with neuronal health. Additionally, several studies have described altered RA levels in AD patients, reinforcing RA Receptor (RAR) signalling as a promising therapeutic strategy. In this review we provide a holistic approach focussing on the effects of isoform-specific RAR modulation with respect to APP secretases and discuss its advantages and drawbacks in subcellular AD related events.
Collapse
Affiliation(s)
- José J M Vitória
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
16
|
Chen BW, Zhang KW, Chen SJ, Yang C, Li PG. Vitamin A Deficiency Exacerbates Gut Microbiota Dysbiosis and Cognitive Deficits in Amyloid Precursor Protein/Presenilin 1 Transgenic Mice. Front Aging Neurosci 2021; 13:753351. [PMID: 34790112 PMCID: PMC8591312 DOI: 10.3389/fnagi.2021.753351] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/04/2021] [Indexed: 01/17/2023] Open
Abstract
Vitamin A deficiency (VAD) plays an essential role in the pathogenesis of Alzheimer’s disease (AD). However, the specific mechanism by which VAD aggravates cognitive impairment is still unknown. At the intersection of microbiology and neuroscience, the gut-brain axis is undoubtedly contributing to the formation and function of neurological systems, but most of the previous studies have ignored the influence of gut microbiota on the cognitive function in VAD. Therefore, we assessed the effect of VAD on AD pathology and the decline of cognitive function in AD model mice and determined the role played by the intestinal microbiota in the process. Twenty 8-week-old male C57BL/6J amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice were randomly assigned to either a vitamin A normal (VAN) or VAD diet for 45 weeks. Our results show that VAD aggravated the behavioral learning and memory deficits, reduced the retinol concentration in the liver and the serum, decreased the transcription of vitamin A (VA)-related receptors and VA-related enzymes in the cortex, increased amyloid-β peptides (Aβ40 and Aβ42) in the brain and gut, upregulate the translation of beta-site APP-cleaving enzyme 1 (BACE1) and phosphorylated Tau in the cortex, and downregulate the expression of brain-derived neurotrophic factor (BDNF) and γ-aminobutyric acid (GABA) receptors in the cortex. In addition, VAD altered the composition and functionality of the fecal microbiota as exemplified by a decreased abundance of Lactobacillus and significantly different α- and β-diversity. Of note, the functional metagenomic prediction (PICRUSt analysis) indicated that GABAergic synapse and retinol metabolism decreased remarkably after VAD intervention, which was in line with the decreased expression of GABA receptors and the decreased liver and serum retinol. In summary, the present study provided valuable facts that VAD exacerbated the morphological, histopathological, molecular biological, microbiological, and behavioral impairment in the APP/PS1 transgenic mice, and the intestinal microbiota may play a key mediator role in this mechanism.
Collapse
Affiliation(s)
- Bo-Wen Chen
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Kai-Wen Zhang
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Si-Jia Chen
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Chun Yang
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Peng-Gao Li
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
17
|
Biyong EF, Tremblay C, Leclerc M, Caron V, Alfos S, Helbling JC, Rodriguez L, Pernet V, Bennett DA, Pallet V, Calon F. Role of Retinoid X Receptors (RXRs) and dietary vitamin A in Alzheimer's disease: Evidence from clinicopathological and preclinical studies. Neurobiol Dis 2021; 161:105542. [PMID: 34737043 DOI: 10.1016/j.nbd.2021.105542] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Vitamin A (VitA), via its active metabolite retinoic acid (RA), is critical for the maintenance of memory function with advancing age. Although its role in Alzheimer's disease (AD) is not well understood, data suggest that impaired brain VitA signaling is associated with the accumulation of β-amyloid peptides (Aβ), and could thus contribute to the onset of AD. METHODS We evaluated the protective action of a six-month-long dietary VitA-supplementation (20 IU/g), starting at 8 months of age, on the memory and the neuropathology of the 3xTg-AD mouse model of AD (n = 11-14/group; including 4-6 females and 7-8 males). We also measured protein levels of Retinoic Acid Receptor β (RARβ) and Retinoid X Receptor γ (RXRγ) in homogenates from the inferior parietal cortex of 60 participants of the Religious Orders study (ROS) divided in three groups: no cognitive impairment (NCI) (n = 20), mild cognitive impairment (MCI) (n = 20) and AD (n = 20). RESULTS The VitA-enriched diet preserved spatial memory of 3xTg-AD mice in the Y maze. VitA-supplementation affected hippocampal RXR expression in an opposite way according to sex by tending to increase in males and decrease in females their mRNA expression. VitA-enriched diet also reduced the amount of hippocampal Aβ40 and Aβ42, as well as the phosphorylation of tau protein at sites Ser396/Ser404 (PHF-1) in males. VitA-supplementation had no effect on tau phosphorylation in females but worsened their hippocampal Aβ load. However, the expression of Rxr-β in the hippocampus was negatively correlated with the amount of both soluble and insoluble Aβ in both males and females. Western immunoblotting in the human cortical samples of the ROS study did not reveal differences in RARβ levels. However, it evidenced a switch from a 60-kDa-RXRγ to a 55-kDa-RXRγ in AD, correlating with ante mortem cognitive decline and the accumulation of neuritic plaques in the brain cortex. CONCLUSION Our data suggest that (i) an altered expression of RXRs receptors is a contributor to β-amyloid pathology in both humans and 3xTg-AD mice, (ii) a chronic exposure of 3xTg-AD mice to a VitA-enriched diet may be protective in males, but not in females.
Collapse
Affiliation(s)
- Essi F Biyong
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, Québec, Canada; LIA OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada), Canada
| | - Cyntia Tremblay
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada
| | - Manon Leclerc
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada
| | - Vicky Caron
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada
| | - Serge Alfos
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | | | - Léa Rodriguez
- CUO-Recherche, Centre de Recherche du CHU de Québec, Québec, QC, Canada; Département d'ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Vincent Pernet
- CUO-Recherche, Centre de Recherche du CHU de Québec, Québec, QC, Canada; Département d'ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Véronique Pallet
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; LIA OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada), Canada
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, Québec, Canada; LIA OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada), Canada.
| |
Collapse
|
18
|
Wołoszynowska-Fraser MU, Kouchmeshky A, McCaffery P. Vitamin A and Retinoic Acid in Cognition and Cognitive Disease. Annu Rev Nutr 2021; 40:247-272. [PMID: 32966186 DOI: 10.1146/annurev-nutr-122319-034227] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The history of vitamin A goes back over one hundred years, but our realization of its importance for the brain and cognition is much more recent. The brain is more efficient than other target tissues at converting vitamin A to retinoic acid (RA), which activates retinoic acid receptors (RARs). RARs regulate transcription, but their function in the cytoplasm to control nongenomic actions is also crucial. Controlled synthesis of RA is essential for regulating synaptic plasticity in regions of the brain involved in learning and memory, such as the hippocampus. Vitamin A deficiency results in a deterioration of these functions, and failure of RA signaling is perhaps associated with normal cognitive decline with age as well as with Alzheimer's disease. Further, several psychiatric and developmental disorders that disrupt cognition are also linked with vitamin A and point to their possible treatment with vitamin A or RA.
Collapse
Affiliation(s)
| | - Azita Kouchmeshky
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, United Kingdom;
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, United Kingdom;
| |
Collapse
|
19
|
Berner D, Hoja U, Zenkel M, Ross JJ, Uebe S, Paoli D, Frezzotti P, Rautenbach RM, Ziskind A, Williams SE, Carmichael TR, Ramsay M, Topouzis F, Chatzikyriakidou A, Lambropoulos A, Sundaresan P, Ayub H, Akhtar F, Qamar R, Zenteno JC, Cruz-Aguilar M, Astakhov YS, Dubina M, Wiggs J, Ozaki M, Kruse FE, Aung T, Reis A, Khor CC, Pasutto F, Schlötzer-Schrehardt U. The protective variant rs7173049 at LOXL1 locus impacts on retinoic acid signaling pathway in pseudoexfoliation syndrome. Hum Mol Genet 2021; 28:2531-2548. [PMID: 30986821 PMCID: PMC6644155 DOI: 10.1093/hmg/ddz075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
Abstract
LOXL1 (lysyl oxidase-like 1) has been identified as the major effect locus in pseudoexfoliation (PEX) syndrome, a fibrotic disorder of the extracellular matrix and frequent cause of chronic open-angle glaucoma. However, all known PEX-associated common variants show allele effect reversal in populations of different ancestry, casting doubt on their biological significance. Based on extensive LOXL1 deep sequencing, we report here the identification of a common non-coding sequence variant, rs7173049A>G, located downstream of LOXL1, consistently associated with a decrease in PEX risk (odds ratio, OR = 0.63; P = 6.33 × 10−31) in nine different ethnic populations. We provide experimental evidence for a functional enhancer-like regulatory activity of the genomic region surrounding rs7173049 influencing expression levels of ISLR2 (immunoglobulin superfamily containing leucine-rich repeat protein 2) and STRA6 [stimulated by retinoic acid (RA) receptor 6], apparently mediated by allele-specific binding of the transcription factor thyroid hormone receptor beta. We further show that the protective rs7173049-G allele correlates with increased tissue expression levels of ISLR2 and STRA6 and that both genes are significantly downregulated in tissues of PEX patients together with other key components of the STRA6 receptor-driven RA signaling pathway. siRNA-mediated downregulation of RA signaling induces upregulation of LOXL1 and PEX-associated matrix genes in PEX-relevant cell types. These data indicate that dysregulation of STRA6 and impaired retinoid metabolism are involved in the pathophysiology of PEX syndrome and that the variant rs7173049-G, which represents the first common variant at the broad LOXL1 locus without allele effect reversal, mediates a protective effect through upregulation of STRA6 in ocular tissues.
Collapse
Affiliation(s)
- Daniel Berner
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Hoja
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - James Julian Ross
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Steffen Uebe
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Paoli
- Department of Ophthalmology, Monfalcone Hospital, Gorizia, Italy
| | - Paolo Frezzotti
- Ophthalmology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Robyn M Rautenbach
- Division of Ophthalmology, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Ari Ziskind
- Division of Ophthalmology, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Susan E Williams
- Division of Ophthalmology, University of the Witwatersrand, Johannesburg, South Africa
| | - Trevor R Carmichael
- Division of Ophthalmology, University of the Witwatersrand, Johannesburg, South Africa
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fotis Topouzis
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anthi Chatzikyriakidou
- Department of Biology and Genetics, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Lambropoulos
- Department of Biology and Genetics, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Periasamy Sundaresan
- Dr. G.Venkataswamy Eye Research Institute, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, India
| | - Humaira Ayub
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Abbottabad, Pakistan
| | - Farah Akhtar
- Pakistan Institute of Ophthalmology, Al-Shifa Trust Eye Hospital, Rawalpindi, Pakistan
| | - Raheel Qamar
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Juan C Zenteno
- Genetics Department, Institute of Ophthalmology 'Conde de Valenciana', Mexico City, Mexico.,Biochemistry Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Marisa Cruz-Aguilar
- Genetics Department, Institute of Ophthalmology 'Conde de Valenciana', Mexico City, Mexico
| | - Yury S Astakhov
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St Petersburg, Russia
| | - Michael Dubina
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St Petersburg, Russia.,St Petersburg Academic University, St Petersburg, Russia
| | - Janey Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Mineo Ozaki
- Ozaki Eye Hospital, Hyuga, Miyazaki, Japan.,Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Friedrich E Kruse
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tin Aung
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Chiea Chuen Khor
- Singapore Eye Research Institute, Singapore.,Genome Institute of Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Francesca Pasutto
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
20
|
Chuang Y, Van I, Zhao Y, Xu Y. Icariin ameliorate Alzheimer's disease by influencing SIRT1 and inhibiting Aβ cascade pathogenesis. J Chem Neuroanat 2021; 117:102014. [PMID: 34407393 DOI: 10.1016/j.jchemneu.2021.102014] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
Of all types of dementia, Alzheimer's disease is the type that has the highest proportion of cases and is the cause of substantial medical and economic burden. The mechanism of Alzheimer's disease is closely associated with the aggregation of amyloid-β protein and causes neurotoxicity and extracellular accumulation in the brain and to intracellular neurofibrillary tangles caused by tau protein hyperphosphorylation in the brain tissue. Previous studies have demonstrated that sirtuin1 downregulation is involved in the pathological mechanism of Alzheimer's disease. The decrease of sirtuin1 level would cause Alzheimer's disease by means of promoting the amyloidogenic pathway to generate amyloid-β species and thereby triggering amyloid-β cascade reaction, such as tau protein hyperphosphorylation, neuron autophagy, neuroinflammation, oxidative stress, and neuron apoptosis. Currently, there is no effective treatment for Alzheimer's disease, it is necessary to develop new treatment strategies. According to the theory of traditional Chinese medicine and based on the mechanism of the disease, tonifying the kidneys is one of the principles for the treatment of Alzheimer's disease and Epimedium is a well-known Chinese medicine for tonifying kidney. Therefore, investigating the influence of the components of Epimedium on the pathological characteristics of Alzheimer's disease may provide a reference for the treatment of Alzheimer's disease in the future. In this article, we summarise the effects and mechanism of icariin, the main ingredient extracted from Epimedium, in ameliorating Alzheimer's disease by regulating sirtuin1 to inhibit amyloid-β protein and improve other amyloid-β cascade pathogenesis.
Collapse
Affiliation(s)
- Yaochen Chuang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China; Kiang Wu Nursing College of Macau, Macao, 999078, China
| | - Iatkio Van
- Kiang Wu Nursing College of Macau, Macao, 999078, China.
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China.
| |
Collapse
|
21
|
Zhi Z, Tang X, Wang Y, Chen R, Ji H. Sinensetin Attenuates Amyloid Beta 25-35-Induced Oxidative Stress, Inflammation, and Apoptosis in SH-SY5Y Cells Through the TLR4/NF-κB Signaling Pathway. Neurochem Res 2021; 46:3012-3024. [PMID: 34309775 DOI: 10.1007/s11064-021-03406-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 12/31/2022]
Abstract
Sinensetin (SIN) is an important active compound that exists widely in citrus plants, and has been reported to exhibit various pharmacological properties, including anti-oxidative, anti-inflammatory, and anti-tumor. This study was designed to examine whether SIN can protect against amyloid beta (Aβ)-induced neurotoxicity and to elucidate the underlying mechanism. Our results showed that pretreatment with SIN for 1 h, followed by co-treatment with Aβ plus SIN for 24 h, attenuated Aβ25-35-induced cell viability reduction, oxidative stress, inflammation, and apoptosis in a dose-dependent manner. Aβ25-35-induced upregulation of Toll-like receptor 4 (TLR4) expression and nuclear translocation of nuclear factor-kappaB (NF-κB) p65 subunit were inhibited by pretreatment with SIN. Furthermore, the protective effect of SIN was abrogated by TLR4 overexpression. Hence, our data suggested that SIN attenuated Aβ25-35-induced neurotoxicity through the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Zhongwen Zhi
- Department of Neurology, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Xiaohong Tang
- Department of Neurology, Hongze Huai'an District People's Hospital, Huai'an, 223100, Jiangsu, People's Republic of China
| | - Yuqian Wang
- Department of Neurology, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Rui Chen
- Department of Neurology, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, Jiangsu, People's Republic of China
| | - Hu Ji
- Department of Neurology, Kangda College of Nanjing Medical University Affiliated Lianshui County People's Hospital, 6 Hongri Avenue East, Lianshui County, Huai'an, 223400, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
|
23
|
All-trans retinoic acid reduces mammalian target of rapamycin via a Sirtuin1-dependent mechanism in neurons. Neuroreport 2021; 32:975-982. [PMID: 34050114 DOI: 10.1097/wnr.0000000000001672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neuroinflammation has emerged as a key contributor in the pathogenesis of Alzheimer's disease (AD). Mammalian target of rapamycin (mTOR) is a key regulator of metabolism, cell growth and protein synthesis. And an elevated mTOR activity has been detected in AD-affected brain areas. Previous studies have suggested that all-trans retinoic acid (atRA) and rapamycin (RAPA), an mTOR inhibitor, protect lipopolysaccharide (LPS)-induced neuronal inflammation through inhibiting nuclear import of NFκB. The aim of this study was to test the effects of atRA on mTOR expression. Here we discovered that mTOR and p-mTOR expression are elevated in LPS-treated mice or primary rat neurons, while atRA blocks the mTOR gene upregulation via a SIRT1-dependent mechanism. The results of this study demonstrated that atRA may protect LPS-induced neuronal inflammation through suppressing mTOR signaling.
Collapse
|
24
|
Uddin MS, Hasana S, Ahmad J, Hossain MF, Rahman MM, Behl T, Rauf A, Ahmad A, Hafeez A, Perveen A, Ashraf GM. Anti-Neuroinflammatory Potential of Polyphenols by Inhibiting NF-κB to Halt Alzheimer's Disease. Curr Pharm Des 2021; 27:402-414. [PMID: 33213314 DOI: 10.2174/1381612826666201118092422] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is an irrevocable chronic brain disorder featured by neuronal loss, microglial accumulation, and progressive cognitive impairment. The proper pathophysiology of this life-threatening disorder is not completely understood and no exact remedies have been found yet. Over the last few decades, research on AD has mainly highlighted pathomechanisms linked to a couple of the major pathological hallmarks, including extracellular senile plaques made of amyloid-β (Aβ) peptides, and intracellular neurofibrillary tangles (NFTs) made of tau proteins. Aβ can induce apoptosis, trigger an inflammatory response, and inhibit the synaptic plasticity of the hippocampus, which ultimately contributes to reducing cognitive functions and memory impairment. Recently, a third disease hallmark, the neuroinflammatory reaction that is mediated by cerebral innate immune cells, has become a spotlight in the current research area, assured by pre-clinical, clinical, and genetic investigations. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a cytokine producer, is significantly associated with physiological inflammatory proceedings and thus shows a promising candidate for inflammation- based AD therapy. Recent data reveal that phytochemicals, mainly polyphenol compounds, exhibit potential neuroprotective functions and these may be considered as a vital resource for discovering several drug candidates against AD. Interestingly, phytochemicals can easily interfere with the signaling pathway of NF-κB. This review represents the anti-neuroinflammatory potential of polyphenols as inhibitors of NF-κB to combat AD pathogenesis.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Sharifa Hasana
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
25
|
Reduction BACE1 expression via suppressing NF-κB mediated signaling by Tamibarotene in a mouse model of Alzheimer's disease. IBRO Neurosci Rep 2021; 10:153-160. [PMID: 33842919 PMCID: PMC8019995 DOI: 10.1016/j.ibneur.2021.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 01/19/2021] [Accepted: 02/09/2021] [Indexed: 01/27/2023] Open
Abstract
This present study examined the effect of Tamibarotene (AM80) in APP/PS1 mice, a well-established AD mouse model. AM80 was intraperitoneal administered to 3-month-old APP/PS1 mice at a dose of 5 mg/kg/day for 16 weeks. The results clearly showed that AM80 could reduce amyloid-β peptides through impact on APP processing and reduce microglia and astrocyte activation in APP/PS1 mice. The most notable finding in the present study was that inhibitory effect on BACE1 mediated by NF-κB pathway underlies the anti-inflammatory action of AM80. Moreover, AM80 could significantly decrease synaptic loss and enhance the expressions of Synapsin and Drebrin. Therefore, AM80 treatment may have the preclinical prevention of AD with new therapeutic strategies.
Collapse
|
26
|
Park JH, Kim J, Choi SY, Lee B, Lee JE, Park H, Moon JW, Park SH, Lee JM, Lee HS, Oh J. Albumin inhibits the nuclear translocation of Smad3 via interleukin-1beta signaling in hepatic stellate cells. Sci Rep 2021; 11:3196. [PMID: 33542418 PMCID: PMC7862402 DOI: 10.1038/s41598-021-82758-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Activation of quiescent hepatic stellate cells (HSCs) to myofibroblasts plays a key role in liver fibrosis. We had previously shown that albumin and its derivative, R-III (a retinol-binding protein—albumin domain III fusion protein), inhibited HSC activation by sequestering retinoic acid (RA) and that R-III administration reduced carbon tetrachloride (CCl4)-induced liver fibrosis. In this study, we aimed to elucidate the mechanism of action of albumin downstream of RA sequestration. Nuclear factor-κB p65 was evenly distributed in the cytoplasm in activated mouse HSCs, whereas albumin expression or R-III treatment (albumin/R-III) caused the nuclear translocation of p65, probably via RA sequestration, resulting in a dramatic increase in interleukin-1beta (IL-1β) expression. Albumin/R-III in turn induced the phosphorylation of Smad3 at the linker region, inhibiting its nuclear import in an IL-1β-dependent manner. Consistent with the in vitro results, the level of IL-1β mRNA expression was higher in CCl4/R-III-treated livers than in CCl4-treated livers. These findings reveal that albumin/R-III inhibits the transforming growth factor-β-Smad3 signaling as well as the retinoic acid receptor-mediated pathway, which probably contributes to the inhibition of HSC activation, and suggest that R-III may be an anti-fibrotic drug candidate.
Collapse
Affiliation(s)
- Ji Hoon Park
- Protein Drug Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong, 28160, Korea
| | - Janghyun Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - So-Young Choi
- Protein Drug Team at New Drug Development Center, Osong Medical Innovation Foundation, Osong, 28160, Korea
| | - Boram Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Jung-Eun Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Heekyung Park
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Ji Wook Moon
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Sun-Hwa Park
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea
| | - Jae Min Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Korea
| | - Hong Sik Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Korea
| | - Junseo Oh
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Korea. .,Department of Biomedical Science, Korea University Graduate School, Seoul, 02841, Korea.
| |
Collapse
|
27
|
Xing Z, Zuo Z, Hu D, Zheng X, Wang X, Yuan L, Zhou L, Qi F, Yao Z. Influenza vaccine combined with moderate-dose PD1 blockade reduces amyloid-β accumulation and improves cognition in APP/PS1 mice. Brain Behav Immun 2021; 91:128-141. [PMID: 32956831 DOI: 10.1016/j.bbi.2020.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Immune dysfunction is implicated in Alzheimer's disease (AD), whereas systemic immune modulation may be neuroprotective. Our previous results have indicated immune challenge with Bacillus Calmette-Guerin attenuates AD pathology in animal models by boosting the systemic immune system. Similarly, independent studies have shown that boosting systemic immune system, by blocking PD-1 checkpoint pathway, modifies AD. Here we hypothesized that influenza vaccine would potentiate function of moderate dose anti-PD-1 and therefore combining them might allow reducing the dose of PD-1 antibody needed to modify the disease. We found that moderate-dose PD-1 in combination with influenza vaccine effectively attenuated cognitive deficit and prevented amyloid-β pathology build-up in APP/PS1 mice in a mechanism dependent on recruitment of peripheral monocyte-derived macrophages into the brain. Eliminating peripheral macrophages abrogated the beneficial effect. Moreover, by comparing CD11b+ compartments in the mouse parenchyma, we observed an elevated subset of Ly6C+ microglia-like cells, which are reportedly derived from peripheral monocytes. In addition, myeloid-derived suppressor cells are strongly elevated in the transgenic model used and normalized by combination treatment, indicating restoration of brain immune homeostasis. Overall, our results suggest that revitalizing brain immunity by combining IV with moderate-dose PD-1 inhibition may represent a therapeutic immunotherapy for AD.
Collapse
Affiliation(s)
- Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Dandan Hu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, PR China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lifang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Lihua Zhou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Department of Anatomy, Sun Yat-sen University, School of Medicine, Guangzhou 510089, PR China.
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| |
Collapse
|
28
|
Dimethyl fumarate exerts neuroprotection by modulating calcineurin/NFAT1 and NFκB dependent BACE1 activity in Aβ1-42 treated neuroblastoma SH-SY5Y cells. Brain Res Bull 2020; 165:97-107. [DOI: 10.1016/j.brainresbull.2020.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/04/2020] [Accepted: 08/22/2020] [Indexed: 12/31/2022]
|
29
|
Hummel R, Ulbrich S, Appel D, Li S, Hirnet T, Zander S, Bobkiewicz W, Gölz C, Schäfer MK. Administration of all-trans retinoic acid after experimental traumatic brain injury is brain protective. Br J Pharmacol 2020; 177:5208-5223. [PMID: 32964418 PMCID: PMC7588818 DOI: 10.1111/bph.15259] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE All-trans retinoic acid (ATRA) is a vitamin A metabolite, important in the developing and mature brain. Pre-injury ATRA administration ameliorates ischaemic brain insults in rodents. This study examined the effects of post-traumatic ATRA treatment in experimental traumatic brain injury (TBI). EXPERIMENTAL APPROACH Male adult mice were subjected to the controlled cortical impact model of TBI or sham procedure and killed at 7 or 30 days post-injury (dpi). ATRA (10 mg kg-1, i.p.) was given immediately after the injury and 1, 2 and 3 dpi. Neurological function and sensorimotor coordination were evaluated. Brains were processed for (immuno-) histological, mRNA and protein analyses (qPCR and western blot). KEY RESULTS ATRA treatment reduced brain lesion size, reactive astrogliosis and axonal injury at 7 dpi, and hippocampal granule cell layer (GCL) integrity was protected at 7 and 30 dpi, independent of cell proliferation in neurogenic niches and blood-brain barrier damage. Neurological and motor deficits over time and the brain tissue loss at 30 dpi were not affected by ATRA treatment. ATRA decreased gene expression of markers for damage-associated molecular pattern (HMGB1), apoptosis (caspase-3 and Bax), activated microglia (TSPO), and reactive astrogliosis (GFAP, SerpinA3N) at 7 dpi and a subset of markers at 30 dpi (TSPO, GFAP). CONCLUSION AND IMPLICATIONS In experimental TBI, post-traumatic ATRA administration exerted brain protective effects, including long-term protection of GCL integrity, but did not affect neurological and motor deficits. Further investigations are required to optimize treatment regimens to enhance ATRA's brain protective effects and improve outcomes.
Collapse
Affiliation(s)
- Regina Hummel
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Sebastian Ulbrich
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Dominik Appel
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Shuailong Li
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Tobias Hirnet
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Sonja Zander
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Wieslawa Bobkiewicz
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Christina Gölz
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Michael K.E. Schäfer
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
- Focus Program Translational Neurosciences (FTN)Johannes Gutenberg‐University MainzMainzGermany
- Research Center for ImmunotherapyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| |
Collapse
|
30
|
Abstract
Nuclear receptors have a broad spectrum of biological functions in normal physiology and in the pathology of various diseases, including glomerular disease. The primary therapies for many glomerular diseases are glucocorticoids, which exert their immunosuppressive and direct podocyte protective effects via the glucocorticoid receptor (GR). As glucocorticoids are associated with important adverse effects and a substantial proportion of patients show resistance to these therapies, the beneficial effects of selective GR modulators are now being explored. Peroxisome proliferator-activated receptor-γ (PPARγ) agonism using thiazolidinediones has potent podocyte cytoprotective and nephroprotective effects. Repurposing of thiazolidinediones or identification of novel PPARγ modulators are potential strategies to treat non-diabetic glomerular disease. Retinoic acid receptor-α is the key mediator of the renal protective effects of retinoic acid, and repair of the endogenous retinoic acid pathway offers another potential therapeutic strategy for glomerular disease. Vitamin D receptor, oestrogen receptor and mineralocorticoid receptor modulators regulate podocyte injury in experimental models. Further studies are needed to better understand the mechanisms of these nuclear receptors, evaluate their synergistic pathways and identify their novel modulators. Here, we focus on the role of nuclear receptors in podocyte biology and non-diabetic glomerular disease.
Collapse
|
31
|
Endres K. Retinoic Acid and the Gut Microbiota in Alzheimer's Disease: Fighting Back-to-Back? Curr Alzheimer Res 2020; 16:405-417. [PMID: 30907321 DOI: 10.2174/1567205016666190321163705] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND There is growing evidence that the gut microbiota may play an important role in neurodegenerative diseases such as Alzheimer's disease. However, how these commensals influence disease risk and progression still has to be deciphered. OBJECTIVE The objective of this review was to summarize current knowledge on the interplay between gut microbiota and retinoic acid. The latter one represents one of the important micronutrients, which have been correlated to Alzheimer's disease and are used in initial therapeutic intervention studies. METHODS A selective overview of the literature is given with the focus on the function of retinoic acid in the healthy and diseased brain, its metabolism in the gut, and the potential influence that the bioactive ligand may have on microbiota, gut physiology and, Alzheimer's disease. RESULTS Retinoic acid can influence neuronal functionality by means of plasticity but also by neurogenesis and modulating proteostasis. Impaired retinoid-signaling, therefore, might contribute to the development of diseases in the brain. Despite its rather direct impact, retinoic acid also influences other organ systems such as gut by regulating the residing immune cells but also factors such as permeability or commensal microbiota. These in turn can also interfere with retinoid-metabolism and via the gutbrain- axis furthermore with Alzheimer's disease pathology within the brain. CONCLUSION Potentially, it is yet too early to conclude from the few reports on changed microbiota in Alzheimer's disease to a dysfunctional role in retinoid-signaling. However, there are several routes how microbial commensals might affect and might be affected by vitamin A and its derivatives.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
32
|
Latexin deficiency in mice up-regulates inflammation and aggravates colitis through HECTD1/Rps3/NF-κB pathway. Sci Rep 2020; 10:9868. [PMID: 32555320 PMCID: PMC7299958 DOI: 10.1038/s41598-020-66789-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
The function of Latexin (LXN) in inflammation has attracted attention. However, no data are available regarding its role in colitis. We report that LXN is a suppressor of colitis. LXN deficiency leads to the severity of colitis in DSS-induced mice, and LXN is required for the therapeutic effect of retinoic acid on colitis. Using a proteomics approach, we demonstrate that LXN interacts and forms a functional complex with HECTD1 (an E3 ubiquitin ligase) and ribosomal protein subunit3 (Rps3). IκBα is one of the substrates of HECTD1. Ectopic expression of LXN leads to IκBα accumulation in intestinal epithelial cells, however, LXN knockdown enhances the interaction of HECTD1 and Rps3, contributing to the ubiquitination degradation of IκBα, and subsequently enhances inflammatory response. Thus, our findings provided a novel mechanism underlying LXN modulates colitis via HECTD1/Rps3/NF-κB pathway and significant implications for the development of novel strategies for the treatment of colitis by targeting LXN.
Collapse
|
33
|
Wei C, Fan J, Sun X, Yao J, Guo Y, Zhou B, Shang Y. Acetyl-11-keto-β-boswellic acid ameliorates cognitive deficits and reduces amyloid-β levels in APPswe/PS1dE9 mice through antioxidant and anti-inflammatory pathways. Free Radic Biol Med 2020; 150:96-108. [PMID: 32109514 DOI: 10.1016/j.freeradbiomed.2020.02.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/16/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a complex disease involved oxidative stress and inflammation in its pathogenesis. Acetyl-11-keto-β-boswellic acid (AKBA) is an active triterpenoid compound from extracts of Boswellia serrata, which has been widely used as an antioxidant and anti-inflammatory agent. The present study was to determine whether AKBA, a novel candidate, could protect against cognitive and neuropathological impairments in AD. We found that AKBA treatment resulted in a significant improvement of learning and memory deficits, a dramatic decrease in cerebral amyloid-β (Aβ) levels and plaque burden, a profound alleviation in oxidative stress and inflammation, and a marked reduction in activated glial cells and synaptic defects in the APPswe/PS1dE9 mice. Furthermore, amyloid precursor protein (APP) processing was remarkably suppressed with AKBA treatment by inhibiting beta-site APP cleaving enzyme 1 (BACE1) protein expression to produce Aβ in the APPswe/PS1dE9 mice brains. Mechanistically, AKBA modulated antioxidant and anti-inflammatory pathways via increasing nuclear erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression, and via declining phosphorylation of inhibitor of nuclear factor-kappa B alpha (IκBα) and p65. Collectively, our findings provide evidence that AKBA protects neurons against oxidative stress and inflammation in AD, and this neuroprotective effect involves the Nrf2/HO-1 and nuclear factor-kappa B (NF-κB) signaling pathways.
Collapse
Affiliation(s)
- Chao Wei
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jiao Fan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xuan Sun
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiarui Yao
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yane Guo
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Bo Zhou
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanchang Shang
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
34
|
Medina DX, Chung EP, Teague CD, Bowser R, Sirianni RW. Intravenously Administered, Retinoid Activating Nanoparticles Increase Lifespan and Reduce Neurodegeneration in the SOD1 G93A Mouse Model of ALS. Front Bioeng Biotechnol 2020; 8:224. [PMID: 32292776 PMCID: PMC7118553 DOI: 10.3389/fbioe.2020.00224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of the retinoic acid (RA) signaling pathway is observed in amyotrophic lateral sclerosis (ALS) and other neurodegenerative disorders. Here, we investigated the therapeutic potential of retinoid activation via the RA receptor β (RARβ) in the SOD1G93A mouse model of ALS. Our approach utilized the RARβ agonist adapalene, which we previously found to be neuroprotective in vitro. Adapalene, like most retinoids, is poorly water soluble, which has thus far prevented effective drug delivery in vivo. To address this challenge, we encapsulated adapalene within nanoparticles (Adap-NPs) composed of poly(lactic acid)-poly(ethylene glycol) (PLA-PEG). Our data demonstrate that intravenous administration of Adap-NPs robustly activates retinoid signaling in the CNS. Chronic administration of Adap-NPs resulted in improved motor performance, prolonged lifespan, and neuroprotection in SOD1G93A mice. This study highlights retinoid signaling as a valuable therapeutic approach and presents a novel nanoparticle platform for the treatment of ALS.
Collapse
Affiliation(s)
- David X Medina
- Gregory W. Fulton ALS Center, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States.,Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Eugene P Chung
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Collin D Teague
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Robert Bowser
- Gregory W. Fulton ALS Center, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Rachael W Sirianni
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, United States.,Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
35
|
Ma LY, Liu SF, Du JH, Niu Y, Hou PF, Shu Q, Ma RR, Wu SD, Qu QM, Lv YL. Chronic ghrelin administration suppresses IKK/NF-κB/BACE1 mediated Aβ production in primary neurons and improves cognitive function via upregulation of PP1 in STZ-diabetic rats. Neurobiol Learn Mem 2020; 169:107155. [PMID: 31904547 DOI: 10.1016/j.nlm.2019.107155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 11/19/2019] [Accepted: 12/31/2019] [Indexed: 01/19/2023]
Abstract
Diabetic rats display cognition impairments accompanied by activation of NF-κB signalling and increased Aβ expression. Ghrelin has been suggested to improve cognition in diabetic rats. In this study, we investigated the role of ghrelin on cognition and NF-κB mediated Aβ production in diabetic rats. A diabetic rat model was established with streptozotocin (STZ) injection, and diabetic rats were intracerebroventricularly administered with ghrelin or (D-lys3)-GHRP-6 (DG). Our results showed that diabetic rats had cognition impairment in the Morris water maze test, accompanied by the higher expression of Aβ in the hippocampus. Western blot analysis showed that diabetic rats exhibited significantly decreased levels of GHSR-1a and protein phosphatase 1 (PP1) in the hippocampus and increased activation of the IKK/NF-κB/BACE1 pathway. Chronic ghrelin administration upregulated hippocampal PP1 expression, suppressed IKK/NF-κB/BACE1 mediated Aβ production, and improved cognition in STZ-induced diabetic rats. These effects were reversed by DG. Then, primary rat hippocampal neurons were isolated and treated with high glucose, followed by Ghrelin and DG, PP1 or IKK. Similar to the in vivo results, high glucose suppressed the expression levels of GHSR-1a and PP1, activated the IKK/NF-κB/BACE1 pathway, increased Aβ production. Ghrelin suppressed IKK/NF-κB/BACE1 induced Aβ production. This improvement was reversed by DG and a PP1 antagonist and was enhanced by the IKK antagonist. Our findings indicated that chronic ghrelin administration can suppress IKK/NF-κB/BACE1 mediated Aβ production in primary neurons with high glucose treatment and improve the cognition via PP1 upregulation in diabetic rats.
Collapse
Affiliation(s)
- Lou-Yan Ma
- The Second Department of Geriatrics, Ninth Hospital of Xi'an, Xi'an, China
| | - Song-Fang Liu
- Department of Endocrinology, Ninth Hospital of Xi'an, Xi'an, China
| | - Jun-Hui Du
- Department of Ophthalmology, Ninth Hospital of Xi'an, Xi'an, China
| | - Yu Niu
- Department of Endocrinology, Ninth Hospital of Xi'an, Xi'an, China
| | - Peng-Fei Hou
- Department of Neurosurgery, Ninth Hospital of Xi'an, Xi'an, China
| | - Qing Shu
- Department of Pharmacy, Ninth Hospital of Xi'an, Xi'an, China
| | - Ran-Ran Ma
- Department of Neurology, Ninth Hospital of Xi'an, Xi'an, China
| | - Song-Di Wu
- Department of Neurology, First Hospital of Xi'an, Xi'an, China.
| | - Qiu-Min Qu
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Ya-Li Lv
- Department of Neurology, Fourth Hospital of Xi'an, Xi'an, China.
| |
Collapse
|
36
|
Khatib T, Chisholm DR, Whiting A, Platt B, McCaffery P. Decay in Retinoic Acid Signaling in Varied Models of Alzheimer's Disease and In-Vitro Test of Novel Retinoic Acid Receptor Ligands (RAR-Ms) to Regulate Protective Genes. J Alzheimers Dis 2020; 73:935-954. [PMID: 31884477 PMCID: PMC7081102 DOI: 10.3233/jad-190931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2019] [Indexed: 12/22/2022]
Abstract
Retinoic acid has been previously proposed in the treatment of Alzheimer's disease (AD). Here, five transgenic mouse models expressing AD and frontotemporal dementia risk genes (i.e., PLB2APP, PLB2TAU, PLB1Double, PLB1Triple, and PLB4) were used to investigate if consistent alterations exist in multiple elements of the retinoic acid signaling pathway in these models. Many steps of the retinoic acid signaling pathway including binding proteins and metabolic enzymes decline, while the previously reported increase in RBP4 was only consistent at late (6 months) but not early (3 month) ages. The retinoic acid receptors were exceptional in their consistent decline in mRNA and protein with transcript decline of retinoic acid receptors β and γ by 3 months, before significant pathology, suggesting involvement in early stages of disease. Decline in RBP1 transcript may also be an early but not late marker of disease. The decline in the retinoic acid signaling system may therefore be a therapeutic target for AD and frontotemporal dementia. Thus, novel stable retinoic acid receptor modulators (RAR-Ms) activating multiple genomic and non-genomic pathways were probed for therapeutic control of gene expression in rat primary hippocampal and cortical cultures. RAR-Ms promoted the non-amyloidogenic pathway, repressed lipopolysaccharide induced inflammatory genes and induced genes with neurotrophic action. RAR-Ms had diverse effects on gene expression allowing particular RAR-Ms to be selected for maximal therapeutic effect. Overall the results demonstrated the early decline of retinoic acid signaling in AD and frontotemporal dementia models and the activity of stable and potent alternatives to retinoic acid as potential therapeutics.
Collapse
Affiliation(s)
- Thabat Khatib
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - David R. Chisholm
- Department of Chemistry, Durham University, Science Laboratories, South Road, Durham, UK
| | - Andrew Whiting
- Department of Chemistry, Durham University, Science Laboratories, South Road, Durham, UK
| | - Bettina Platt
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| |
Collapse
|
37
|
Huang P, Zheng N, Zhou HB, Huang J. Curcumin inhibits BACE1 expression through the interaction between ERβ and NFκB signaling pathway in SH-SY5Y cells. Mol Cell Biochem 2019; 463:161-173. [DOI: 10.1007/s11010-019-03638-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022]
|
38
|
Saadipour K, Tiberi A, Lombardo S, Grajales E, Montroull L, Mañucat-Tan NB, LaFrancois J, Cammer M, Mathews PM, Scharfman HE, Liao FF, Friedman WJ, Zhou XF, Tesco G, Chao MV. Regulation of BACE1 expression after injury is linked to the p75 neurotrophin receptor. Mol Cell Neurosci 2019; 99:103395. [PMID: 31422108 DOI: 10.1016/j.mcn.2019.103395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/24/2019] [Accepted: 08/08/2019] [Indexed: 12/25/2022] Open
Abstract
BACE1 is a transmembrane aspartic protease that cleaves various substrates and it is required for normal brain function. BACE1 expression is high during early development, but it is reduced in adulthood. Under conditions of stress and injury, BACE1 levels are increased; however, the underlying mechanisms that drive BACE1 elevation are not well understood. One mechanism associated with brain injury is the activation of injurious p75 neurotrophin receptor (p75), which can trigger pathological signals. Here we report that within 72 h after controlled cortical impact (CCI) or laser injury, BACE1 and p75 are increased and tightly co-expressed in cortical neurons of mouse brain. Additionally, BACE1 is not up-regulated in p75 null mice in response to focal cortical injury, while p75 over-expression results in BACE1 augmentation in HEK-293 and SY5Y cell lines. A luciferase assay conducted in SY5Y cell line revealed that BACE1 expression is regulated at the transcriptional level in response to p75 transfection. Interestingly, this effect does not appear to be dependent upon p75 ligands including mature and pro-neurotrophins. In addition, BACE1 activity on amyloid precursor protein (APP) is enhanced in SY5Y-APP cells transfected with a p75 construct. Lastly, we found that the activation of c-jun n-terminal kinase (JNK) by p75 contributes to BACE1 up-regulation. This study explores how two injury-induced molecules are intimately connected and suggests a potential link between p75 signaling and the expression of BACE1 after brain injury.
Collapse
Affiliation(s)
- Khalil Saadipour
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| | - Alexia Tiberi
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA; Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, Pisa, 56126, Italy
| | - Sylvia Lombardo
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Elena Grajales
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA
| | - Laura Montroull
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Noralyn B Mañucat-Tan
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - John LaFrancois
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Michael Cammer
- DART Microscopy Laboratory, NYU Langone Medical Center, New York, NY 10016, USA
| | - Paul M Mathews
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Helen E Scharfman
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - Giueseppina Tesco
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| |
Collapse
|
39
|
Kuo YC, Rajesh R. Challenges in the treatment of Alzheimer’s disease: recent progress and treatment strategies of pharmaceuticals targeting notable pathological factors. Expert Rev Neurother 2019; 19:623-652. [DOI: 10.1080/14737175.2019.1621750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| |
Collapse
|
40
|
Jha NK, Jha SK, Kar R, Nand P, Swati K, Goswami VK. Nuclear factor-kappa β as a therapeutic target for Alzheimer's disease. J Neurochem 2019; 150:113-137. [PMID: 30802950 DOI: 10.1111/jnc.14687] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/16/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a typical progressive, chronic neurodegenerative disorder with worldwide prevalence. Its clinical manifestation involves the presence of extracellular plaques and intracellular neurofibrillary tangles (NFTs). NFTs occur in brain tissues as a result of both Aβ agglomeration and Tau phosphorylation. Although there is no known cure for AD, research into possible cures and treatment options continues using cell-cultures and model animals/organisms. The nuclear factor-kappa β (NF-κβ) plays an active role in the progression of AD. Impairment to this signaling module triggers undesirable phenotypic changes such as neuroinflammation, activation of microglia, oxidative stress related complications, and apoptotic cell death. These imbalances further lead to homeostatic abnormalities in the brain or in initial stages of AD essentially pushing normal neurons toward the degeneration process. Interestingly, the role of NF-κβ signaling associated receptor-interacting protein kinase is currently observed in apoptotic and necrotic cell death, and has been reported in brains. Conversely, the NF-κβ signaling pathway has also been reported to be involved in normal brain functioning. This pathway plays a crucial role in maintaining synaptic plasticity and balancing between learning and memory. Since any impairment in the pathways associated with NF-κβ signaling causes altered neuronal dynamics, neurotherapeutics using compounds including, antioxidants, bioflavonoids, and non-steroidal anti-inflammatory drugs against such abnormalities offer possibilities to rectify aberrant excitatory neuronal activity in AD. In this review, we have provided an extensive overview of the crucial role of NF-κβ signaling in normal brain homeostasis. We have also thoroughly outlined several established pathomechanisms associated with NF-κβ pathways in AD, along with their respective therapeutic approaches.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Rohan Kar
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Parma Nand
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Kumari Swati
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Vineet Kumar Goswami
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| |
Collapse
|
41
|
Clemens V, Regen F, Le Bret N, Heuser I, Hellmann-Regen J. Retinoic Acid Enhances Apolipoprotein E Synthesis in Human Macrophages. J Alzheimers Dis 2019; 61:1295-1300. [PMID: 29376871 DOI: 10.3233/jad-170823] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apolipoprotein E (ApoE) represents a pivotal target in Alzheimer's disease (AD) and is modulated through retinoic acid (RA), an endogenous neuroprotective and anti-inflammatory compound. A major source of ApoE are microglia, which are pathologically activated in AD. Activated microglia are known to block RA signaling. This suggests a vicious cycle between inflammation, RA signaling, and ApoE homeostasis in AD pathogenesis. To test this hypothesis, we investigated effects of RA and proinflammatory activation on ApoE synthesis in primary human macrophage-derived microglial-like cells. Our results indicate that proinflammatory activation attenuates ApoE synthesis, an effect blocked by RA.
Collapse
Affiliation(s)
- Vera Clemens
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Francesca Regen
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Nathalie Le Bret
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Isabella Heuser
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Julian Hellmann-Regen
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| |
Collapse
|
42
|
Thygesen C, Ilkjær L, Kempf SJ, Hemdrup AL, von Linstow CU, Babcock AA, Darvesh S, Larsen MR, Finsen B. Diverse Protein Profiles in CNS Myeloid Cells and CNS Tissue From Lipopolysaccharide- and Vehicle-Injected APP SWE/PS1 ΔE9 Transgenic Mice Implicate Cathepsin Z in Alzheimer's Disease. Front Cell Neurosci 2018; 12:397. [PMID: 30459560 PMCID: PMC6232379 DOI: 10.3389/fncel.2018.00397] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/15/2018] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation, characterized by chronic activation of the myeloid-derived microglia, is a hallmark of Alzheimer’s disease (AD). Systemic inflammation, typically resulting from infection, has been linked to the progression of AD due to exacerbation of the chronic microglial reaction. However, the mechanism and the consequences of this exacerbation are largely unknown. Here, we mimicked systemic inflammation in AD with weekly intraperitoneal (i.p.) injections of APPSWE/PS1ΔE9 transgenic mice with E. coli lipopolysaccharide (LPS) from 9 to 12 months of age, corresponding to the period with the steepest increase in amyloid pathology. We found that the repeated LPS injections ameliorated amyloid pathology in the neocortex while increasing the neuroinflammatory reaction. To elucidate mechanisms, we analyzed the proteome of the hippocampus from the same mice as well as in unique samples of CNS myeloid cells. The repeated LPS injections stimulated protein pathways of the complement system, retinoid receptor activation and oxidative stress. CNS myeloid cells from transgenic mice showed enrichment in pathways of amyloid-beta clearance and elevated levels of the lysosomal protease cathepsin Z, as well as amyloid precursor protein, apolipoprotein E and clusterin. These proteins were found elevated in the proteome of both LPS and vehicle injected transgenics, and co-localized to CD11b+ microglia in transgenic mice and in primary murine microglia. Additionally, cathepsin Z, amyloid precursor protein, and apolipoprotein E appeared associated with amyloid plaques in neocortex of AD cases. Interestingly, cathepsin Z was expressed in microglial-like cells and co-localized to CD68+ microglial lysosomes in AD cases, and it was expressed in perivascular cells in AD and control cases. Taken together, our results implicate systemic LPS administration in ameliorating amyloid pathology in early-to-mid stage disease in the APPSWE/PS1ΔE9 mouse and attract attention to the potential disease involvement of cathepsin Z expressed in CNS myeloid cells in AD.
Collapse
Affiliation(s)
- Camilla Thygesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.,Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Laura Ilkjær
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stefan J Kempf
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Anne Louise Hemdrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | - Alicia A Babcock
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sultan Darvesh
- Department of Medicine (Neurology and Geriatric Medicine) - Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.,Department of Chemistry and Physics, Mount Saint Vincent University, Halifax, NS, Canada
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
43
|
Uniyal S, Dhasmana A, Tyagi A, Muyal JP. ATRA reduces inflammation and improves alveolar epithelium regeneration in emphysematous rat lung. Biomed Pharmacother 2018; 108:1435-1450. [PMID: 30372846 DOI: 10.1016/j.biopha.2018.09.166] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Pulmonary emphysema characterized by alveolar wall destruction is resultant of persistent chronic inflammation. All-trans retinoic acid (ATRA) has been reported to reverse elastase-induced emphysema in rats. However, the underlying molecular mechanisms are so far unknown. OBJECTIVE To investigate the therapeutic potential effect of ATRA via the amelioration of the ERK/JAK-STAT pathways in the lungs of emphysematous rats. METHODS In silico analysis was done to find the binding efficiency of ATRA with receptor and ligands of ERK & JAK-STAT pathway. Emphysema was induced by porcine pancreatic elastase in Sprague-Dawley rats and ATRA was supplemented as therapy. Lungs were harvested for histopathological, genomics and proteomics analysis. RESULTS AND DISCUSSION In silico docking, analysis confirms that ATRA interferes with the normal binding of ligands (TNF-α, IL6ST) and receptors (TNFR1, IL6) of ERK/JAK-STAT pathways respectively. ATRA restored the histology, proteases/antiproteases balance, levels of inflammatory markers, antioxidants, expression of candidate genes of ERK and JAK-STAT pathways in the therapy group. CONCLUSION ATRA ameliorates ERK/JAK-STAT pathway in emphysema condition, resulting in alveolar epithelium regeneration. Hence, ATRA may prove to be a potential drug in the treatment of emphysema.
Collapse
Affiliation(s)
- Swati Uniyal
- Department of Biotechnology, School of Biotechnology, Gautam Buddha University, Greater Noida, 201308, Uttar Pradesh, India.
| | - Anupam Dhasmana
- Himalayan School of Biosciences and Cancer Research Institute, Swami Rama Himalayan University, Dehradun, India.
| | - Amit Tyagi
- Division of Nuclear Medicine, Institute of Nuclear Medicine and Allied Sciences, DRDO, New Delhi, India.
| | - Jai Prakash Muyal
- Department of Biotechnology, School of Biotechnology, Gautam Buddha University, Greater Noida, 201308, Uttar Pradesh, India.
| |
Collapse
|
44
|
Biliverdin reductase-A impairment links brain insulin resistance with increased Aβ production in an animal model of aging: Implications for Alzheimer disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3181-3194. [PMID: 29981845 DOI: 10.1016/j.bbadis.2018.07.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 06/21/2018] [Accepted: 07/03/2018] [Indexed: 12/26/2022]
|
45
|
Du Y, Zhao Y, Li C, Zheng Q, Tian J, Li Z, Huang TY, Zhang W, Xu H. Inhibition of PKCδ reduces amyloid-β levels and reverses Alzheimer disease phenotypes. J Exp Med 2018; 215:1665-1677. [PMID: 29739836 PMCID: PMC5987914 DOI: 10.1084/jem.20171193] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 01/11/2018] [Accepted: 03/29/2018] [Indexed: 12/28/2022] Open
Abstract
β-amyloid protein (Aβ) plays a central role in the pathogenesis of Alzheimer disease (AD). Aβ is generated from sequential cleavage of amyloid precursor protein (APP) by β-site APP-cleaving enzyme 1 (BACE1) and the γ-secretase complex. Although activation of some protein kinase C (PKC) isoforms such as PKCα and ε has been shown to regulate nonamyloidogenic pathways and Aβ degradation, it is unclear whether other PKC isoforms are involved in APP processing/AD pathogenesis. In this study, we report that increased PKCδ levels correlate with BACE1 expression in the AD brain. PKCδ knockdown reduces BACE1 expression, BACE1-mediated APP processing, and Aβ production. Conversely, overexpression of PKCδ increases BACE1 expression and Aβ generation. Importantly, inhibition of PKCδ by rottlerin markedly reduces BACE1 expression, Aβ levels, and neuritic plaque formation and rescues cognitive deficits in an APP Swedish mutations K594N/M595L/presenilin-1 with an exon 9 deletion-transgenic AD mouse model. Our study indicates that PKCδ plays an important role in aggravating AD pathogenesis, and PKCδ may be a potential target in AD therapeutics.
Collapse
Affiliation(s)
- Ying Du
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Yingjun Zhao
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Chuan Li
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Qiuyang Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, The Collaborative Innovation Center for Brain Science, Medical College, Xiamen University, Xiamen, China
| | - Jing Tian
- College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Marine Bioresources and Eco-Environmental Science, Shenzhen University, Shenzhen, China
| | - Zhuyi Li
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Timothy Y Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Wei Zhang
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xian, China
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA.,Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, The Collaborative Innovation Center for Brain Science, Medical College, Xiamen University, Xiamen, China
| |
Collapse
|
46
|
Marwarha G, Schommer J, Lund J, Schommer T, Ghribi O. Palmitate-induced C/EBP homologous protein activation leads to NF-κB-mediated increase in BACE1 activity and amyloid beta genesis. J Neurochem 2018; 144:761-779. [PMID: 29315574 PMCID: PMC6371812 DOI: 10.1111/jnc.14292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/29/2017] [Accepted: 12/23/2017] [Indexed: 12/12/2022]
Abstract
The etiology of Alzheimer's disease (AD) is egregiously comprehended, but epidemiological studies have posited that diets rich in the saturated fatty acid palmitic acid (palmitate) are a significant risk factor. The production and accumulation of amyloid beta peptide (Aβ) is considered the core pathological molecular event in the pathogenesis of AD. The rate-limiting step in Aβ genesis from amyloid-β precursor protein (AβPP) is catalyzed by the enzyme β-site amyloid precursor protein cleaving enzyme 1 (BACE1), the expression and enzymatic activity of which is significantly up-regulated in the AD brain. In this study, we determined the molecular mechanisms that potentially underlie the palmitate-induced up-regulation in BACE1 expression and augmented Aβ production. We demonstrate that a palmitate-enriched diet and exogenous palmitate treatment evoke an increase in BACE1 expression and activity leading to enhanced Aβ genesis in the mouse brain and SH-SY5Y-APPSwe cells, respectively, through the activation of the transcription factor NF-κB. Chromatin immunoprecipitation (ChIP) assays and luciferase reporter assays revealed that palmitate enhances BACE1 expression by increasing the binding of NF-κB in the BACE1 promoter followed by an enhancement in the transactivation of the BACE1 promoter. Elucidation and delineation of upstream molecular events unveiled a critical role of the endoplasmic reticulum stress-associated transcription factor, C/EBP homologous protein (CHOP) in the palmitate-induced NF-κB activation, as CHOP knock-down cells and Chop-/- mice do not exhibit the same degree of NF-κB activation in response to the palmitate challenge. Our study delineates a novel CHOP-NF-κB signaling pathway that mediates palmitate-induced up-regulation of BACE1 expression and Aβ genesis.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jared Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jonah Lund
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Trevor Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| |
Collapse
|
47
|
Seo EJ, Fischer N, Efferth T. Phytochemicals as inhibitors of NF-κB for treatment of Alzheimer’s disease. Pharmacol Res 2018; 129:262-273. [DOI: 10.1016/j.phrs.2017.11.030] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/19/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022]
|
48
|
Zhang M, Jiang X, Qu M, Gu H, Sha Q, Hua F. Salubrinal abrogates palmitate-induced leptin resistance and endoplasmic reticulum stress via nuclear factor kappa-light-chain-enhancer of activated B cell pathway in mHypoE-44 hypothalamic neurons. Diabetes Metab Syndr Obes 2018; 11:893-899. [PMID: 30584344 PMCID: PMC6287548 DOI: 10.2147/dmso.s179346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The prevalence of obesity is growing rapidly and has become a global problem that increases the risk for many diseases. It is influenced by many factors, including consumption of the Western-style diet, characterized as a high-fat diet. Within the central nervous system, the hypothalamus is a critical site in maintaining energy homeostasis and sensing nutrient status, including palmitate, the major component of high-fat-diet. METHODS In the present study, we conducted a variety of studies to investigate the specific role of salubrinal on palmitate-induced hypothalamic cell death, leptin signaling, and ER stress in an embryonic hypothalamic cell line. Experiments were also performed to identify the underlying mechanisms of the protective effect of salubrinal. RESULTS Our results indicate that salubrinal protects hypothalamic cells against PA-induced ER stress and improves hypothalamic leptin sensitivity. CONCLUSION Taken together, our findings conclusively reveal that salubrinal abrogates palmitate-induced hypothalamic leptin resistance and ER stress via NF-κB pathway.
Collapse
Affiliation(s)
- Min Zhang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Xiaohong Jiang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China,
| | - Meidi Qu
- Department of Clinical Nutrition, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Hongliu Gu
- Department of Clinical Nutrition, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Qi Sha
- Department of Clinical Nutrition, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China,
| |
Collapse
|
49
|
Jones SV, Kounatidis I. Nuclear Factor-Kappa B and Alzheimer Disease, Unifying Genetic and Environmental Risk Factors from Cell to Humans. Front Immunol 2017; 8:1805. [PMID: 29312321 PMCID: PMC5732234 DOI: 10.3389/fimmu.2017.01805] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/30/2017] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, an eversible, progressive disease that causes problems with memory, thinking, language, planning, and behavior. There are a number of risk factors associated with developing AD but the exact cause remains unknown. The predominant theory is that excessive build-up of amyloid protein leads to cell death, brain atrophy, and cognitive and functional decline. However, the amyloid hypothesis has not led to a single successful treatment. The recent failure of Solanezumab, a monoclonal antibody to amyloid, in a large phase III trial was emblematic of the repeated failure of anti-amyloid therapeutics. New disease targets are urgently needed. The innate immune system is increasingly being implicated in the pathology of number of chronic diseases. This focused review will summarize the role of transcription factor nuclear factor-kappa B (NF-κB), a key regulator of innate immunity, in the major genetic and environmental risk factors in cellular, invertebrate and vertebrate models of AD. The paper will also explore the relationship between NF-κB and emerging environmental risk factors in an attempt to assess the potential for this transcription factor to be targeted for disease prevention.
Collapse
Affiliation(s)
- Simon Vann Jones
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | - Ilias Kounatidis
- Laboratory of Cell Biology, Development and Genetics, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Celik E, Sanlier N. Effects of nutrient and bioactive food components on Alzheimer's disease and epigenetic. Crit Rev Food Sci Nutr 2017; 59:102-113. [PMID: 28799782 DOI: 10.1080/10408398.2017.1359488] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly and is a chronic neurodegenerative disease that is becoming widespread. For this reason, in recent years factors affecting the development, progression and cognitive function of the AD have been emphasized. Nutrients and other bioactive nutrients are among the factors that are effective in AD. In particular, vitamins A, C and E, vitamins B1, B6 and B12, folate, magnesium, choline, inositol, anthocyanins, isoflavones etc. nutrients and bioactive nutrients are known to be effective in the development of AD. Nutrients and nutrient components may also have an epigenetic effect on AD. At the same time, nutrients and bioactive food components slow down the progression of the disease. For this reason, the effect of nutrients and food components on AD was examined in this review.
Collapse
Affiliation(s)
- Elif Celik
- a Gazi University , Faculty of Health Sciences, Nutrition and Dietetics Department , Ankara , Turkey
| | - Nevin Sanlier
- a Gazi University , Faculty of Health Sciences, Nutrition and Dietetics Department , Ankara , Turkey
| |
Collapse
|