1
|
Cheng Y, Zhao F, Wang J, Luo H, Mao R, Yu Y, Wang Y, Tan J, Hao X, Wang Y. Preparation, activity, and mechanistic insights of processed Polygala tenuifolia glycoprotein in ameliorating Alzheimer's disease. Int J Biol Macromol 2025; 309:143069. [PMID: 40220839 DOI: 10.1016/j.ijbiomac.2025.143069] [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: 02/09/2025] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
This study examined the efficacy and mechanism of Polygala tenuifolia glycoprotein (ZPG) in alleviating Alzheimer's disease (AD) for clinical application. ZPG's effects were tested in scopolamine hydrobromide-induced AD mice using behavioral, histological, and biomarker investigations. Additionally, 16S rDNA sequencing and lipidomics revealed ZPG's impact on gut microbiota and lipid metabolism, supported by pathway enrichment and correlation analyses. JNK pathway modulation was studied in vitro with purified and characterized ZPG-2. Results showed ZPG significantly improved cognitive deficits, reduced hippocampal pathology, and normalized APP, p-JNK, bax, and bcl-2 levels in AD mice. It also modulated gut microbiota and lipid metabolism, particularly glycerophospholipid pathways. ZPG-2 exhibited neuroprotective effects in Aβ25-35-induced PC12 cells by reducing apoptosis, inhibiting LDH release, and regulating oxidative stress and JNK activity. Structural analysis identified ZPG-2 as a 26 kDa glycoprotein with an O-linked glycopeptide bond and random coil conformation. Correlation analysis showed significant gut microbiota-AD biomarker relationships. These findings suggest ZPG may alleviate AD by reducing oxidative stress, inhibiting apoptosis, modulating gut microbiota, enhancing lipid metabolism, and suppressing the JNK signaling pathway. ZPG may be medicinal, however, more research is needed to validate its efficacy and mechanisms. This study lays the foundation for ZPG as an AD therapy for the future.
Collapse
Affiliation(s)
- Yangang Cheng
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Fuxia Zhao
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Jing Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Hong Luo
- The University of Adelaide, Adelaide, SA 5005, Australia
| | - Rui Mao
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Yuetong Yu
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Yan Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Jinyan Tan
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China.
| | - Xuliang Hao
- Shanxi Academy of Traditional Chinese Medicine, Shanxi, Taiyuan 030024, China.
| | - Yingli Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China.
| |
Collapse
|
2
|
Zhou L, Zhang M, Zheng Q, Song Y, Yan Z, Wang H, Xiong Y, Chen Y, Cai Z, Yuan J. Exploring the Mechanism of Kai-Xin-San to Improve Cognitive Deficits in AD Rats Induced by D-Gal and Aβ 25-35 Based on Multi-Omics and Network Analysis. Biomed Chromatogr 2025; 39:e70047. [PMID: 40033867 DOI: 10.1002/bmc.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease for which there are no effective drugs. Kai-Xin-San (KXS), with definite curative effects, is widely used for the prevention and treatment of AD in China. But its mechanism is not yet fully understood. Based on our established rat model and previous pharmacodynamics study, Multi-omics (metabolomics, proteomics) and network analysis were integrated to explore the holistic mechanism of anti-AD effects of KXS. The key pathways were validated with western blot and ELISA methods. Morris water maze and Nissl staining showed that KXS could ameliorate cognitive deficits and pathological morphology of the hippocampus in AD rats. A total of nine metabolites were identified, which were related to pyrimidine metabolism, riboflavin metabolism, tyrosine metabolism, tryptophan metabolism, and glycerophospholipid metabolism. Proteomics results indicated that the improvement of cognitive deficits by KXS was closely related to the regulation of oxidative phosphorylation in mitochondria. Western blotting results showed that KXS significantly inhibited the expression of Mt-nd2 and Ndufb6 in AD rats. Integrated analysis indicated that the anti-AD targets of KXS were interrelated and KXS could exert its anti-AD effect by reducing oxidative stress, neurotoxicity, and inflammation.
Collapse
Affiliation(s)
- Lifen Zhou
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Min Zhang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
- Nanchang Key Laboratory for Quality and Safety Risk Assessment of Health Food and Its Contact Materials, Nanchang Inspection and Testing Center, Nanchang, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yonggui Song
- Laboratory Animal Science and Technology Development Center, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhihong Yan
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Huijuan Wang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yongchang Xiong
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ying Chen
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhinan Cai
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jinbin Yuan
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
3
|
Lu S, Tao Z, Wang G, Na K, Wu L, Zhang L, Li X, Guo X. Mannuronate Oligosaccharides Ameliorate Experimental Colitis and Secondary Neurological Dysfunction by Manipulating the Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2935-2950. [PMID: 39846727 DOI: 10.1021/acs.jafc.4c10378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Microbiota dysfunction induces intestinal disorders and neurological diseases. Mannuronate oligosaccharides (MAOS), a kind of alginate oligosaccharide (AOS), specifically exert efficacy in shaping gut microbiota and relieving cognitive impairment. However, the key regulatory factors involved, such as the specific strains and metabolites as well as their regulatory mechanisms, remain unclear at present. This research investigates how MAOS specifically impact the gut-brain axis in vivo and in vitro. The results showed that pretreatment with MAOS significantly ameliorated dextran sodium sulfate (DSS)-induced colitis and secondary nerve injury. This preventive mechanism operates through the regulation of serum DOPC abundance and the gut-brain axis, achieved by inhibiting the TLR4/MyD88/NF-κB pathway. These findings underscore the crucial role of dietary MAOS in the prevention of colitis and neurological disorders, providing a rationale for the application of MAOS in disease prevention and functional food ingredients.
Collapse
Affiliation(s)
- Shuang Lu
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| | - Zhengxiong Tao
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| | - Gan Wang
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| | - Kai Na
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| | - Lisha Wu
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| | - Li Zhang
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| | - Xiangyu Li
- Hubei Province Nutrition Chemicals Biosynthetic Engineering Technology Research Center, Wuhan 430073, China
| | - Xiaohua Guo
- College of Life Science, South-Central Minzu University, No. 182, Minyuan Road, Hongshan District, Wuhan 430074, China
| |
Collapse
|
4
|
Conway T, Seidler K, Barrow M. Unlocking choline's potential in Alzheimer's disease: A narrative review exploring the neuroprotective and neurotrophic role of phosphatidylcholine and assessing its impact on memory and learning. Clin Nutr ESPEN 2024; 64:177-195. [PMID: 39357562 DOI: 10.1016/j.clnesp.2024.09.024] [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: 06/24/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND AIMS Growing evidence suggests nutritional intervention may influence the development and progression of Alzheimer's Disease (AD). Choline, an essential dietary nutrient plays a critical role in neurological development and brain function, however, its effects on AD in humans is unclear. The research aims to investigate mechanistic links between dietary choline intake and cognitive functioning, focusing on the role of phosphatidylcholine (PC) in neuroplasticity and its interaction with amyloid beta (Aβ) peptides in neuron membranes. Additionally, human evidence on the potential benefits of PC interventions on AD, cognition, and proposed mechanisms are evaluated. METHODS A reproducible systematic literature search was performed using a three-tranche strategy, consisting of a review, mechanism, and intervention search. Using PubMed as the main database, 1254 titles and abstracts were screened, 149 papers were read in full and 65 peer-reviewed papers were accepted, critically appraised, and analysed in a narrative review. RESULTS Predominantly preclinical evidence demonstrated that PC enhances neuroplasticity, a key biological substrate for cognition, by activating intracellular neuronal signalling pathways or through neuron membrane function. Molecular dynamic simulation methods provided a mechanistic understanding of the interconnection between neuronal PC content and the potential behaviour and trajectory of Aβ peptide aggregation. The results indicate that the neuronal membrane composition of PC is critical to inhibiting Aβ aggregation and neuronal damage, protecting the neuron from Aβ toxicity. This might provide a foundation for optimising cellular PC which may prove beneficial in the treatment or prevention of neurodegenerative disease. Altered PC metabolism in AD was evidenced in observational studies; however, whether this relationship represents a cause or consequence of AD remains to be determined. Human intervention studies did not produce conclusive evidence supporting its effectiveness in enhancing cognitive function. This lack of consistency primarily stems from methodological constraints within the conducted studies. Human observational research provided the most compelling evidence linking a higher dietary PC intake to a reduced risk of dementia and significant improvements in cognitive testing. CONCLUSION Despite the lack of randomised control trials (RCTs) assessing the efficacy of lecithin/PC to improve cognition in AD patients, there exists promising evidence supporting its neuroprotective and neurotrophic role. This review establishes an evidence-based framework through chains of mechanistic evidence, that may provide potential strategies for enhanced neuroprotection and reduced neurodegeneration caused by AD. Considering the escalating global burden of AD and the current shortcomings in effective treatments, this review together with the limitations and gaps identified in the existing research presents valuable insights that emphasise the urgency of more comprehensive research into the relationship between PC and AD.
Collapse
Affiliation(s)
- Tara Conway
- Centre for Nutrition Education and Lifestyle Management (CNELM), PO Box 3739, Wokingham, Berkshire, RG40 9UA, UK.
| | - Karin Seidler
- Centre for Nutrition Education and Lifestyle Management (CNELM), PO Box 3739, Wokingham, Berkshire, RG40 9UA, UK.
| | - Michelle Barrow
- Centre for Nutrition Education and Lifestyle Management (CNELM), PO Box 3739, Wokingham, Berkshire, RG40 9UA, UK.
| |
Collapse
|
5
|
Mak KM, Shekhar AC. Soybean polyenylphosphatidylcholine (PPC) is beneficial in liver and extrahepatic tissue injury: An update in experimental research. Anat Rec (Hoboken) 2024; 307:2162-2186. [PMID: 37814787 DOI: 10.1002/ar.25333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/11/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
Polyenylphosphatidylcholine (PPC) is a purified polyunsaturated phosphatidylcholine extract of soybeans. This article updates PPC's beneficial effects on various forms of liver cell injury and other tissues in experimental research. PPC downregulates hepatocyte CYP2E1 expression and associated hepatotoxicity, as well as attenuates oxidative stress, apoptosis, lipoprotein oxidation and steatosis in alcoholic and nonalcoholic liver injury. PPC inhibits pro-inflammatory cytokine production, while stimulating anti-inflammatory cytokine secretion in ethanol or lipopolysaccharide-stimulated Kupffer cells/macrophages. It promotes M2-type macrophage polarization and metabolic reprogramming of glucose and lipid metabolism. PPC mitigates steatosis in NAFLD through inhibiting polarization of pro-inflammatory M1-type Kupffer cells, alleviating metabolic inflammation, remodeling hepatic lipid metabolism, correcting imbalances between lipogenesis and lipolysis and enhancing lipoprotein secretion from hepatocytes. PPC is antifibrotic by preventing progression of alcoholic hepatic fibrosis in baboons and also prevents CCl4-induced fibrosis in rats. PPC supplementation replenishes the phosphatidylcholine content of damaged cell membranes, resulting in increased membrane fluidity and functioning. Phosphatidylcholine repletion prevents increased membrane curvature of the endoplasmic reticulum and Golgi and decreases sterol regulatory element binding protein-1-mediated lipogenesis, reducing steatosis. PPC remodels gut microbiota and affects hepatic lipid metabolism via the gut-hepatic-axis and also alleviates brain inflammatory responses and cognitive impairment via the gut-brain-axis. Additionally, PPC protects extrahepatic tissues from injury caused by various toxic compounds by reducing oxidative stress, inflammation, and membrane damage. It also stimulates liver regeneration, enhances sensitivity of cancer cells to radiotherapy/chemotherapy, and inhibits experimental hepatocarcinogenesis. PPC's beneficial effects justify it as a supportive treatment of liver disease.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya C Shekhar
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
6
|
Aisyah R, Ohshima N, Watanabe D, Nakagawa Y, Sakuma T, Nitschke F, Nakamura M, Sato K, Nakahata K, Yokoyama C, Marchioni CR, Kumrungsee T, Shimizu T, Sotomaru Y, Takeo T, Nakagata N, Izumi T, Miura S, Minassian BA, Yamamoto T, Wada M, Yanaka N. GDE5/Gpcpd1 activity determines phosphatidylcholine composition in skeletal muscle and regulates contractile force in mice. Commun Biol 2024; 7:604. [PMID: 38769369 PMCID: PMC11106330 DOI: 10.1038/s42003-024-06298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Glycerophosphocholine (GPC) is an important precursor for intracellular choline supply in phosphatidylcholine (PC) metabolism. GDE5/Gpcpd1 hydrolyzes GPC into choline and glycerol 3-phosphate; this study aimed to elucidate its physiological function in vivo. Heterozygous whole-body GDE5-deficient mice reveal a significant GPC accumulation across tissues, while homozygous whole-body knockout results in embryonic lethality. Skeletal muscle-specific GDE5 deletion (Gde5 skKO) exhibits reduced passive force and improved fatigue resistance in electrically stimulated gastrocnemius muscles in vivo. GDE5 deficiency also results in higher glycolytic metabolites and glycogen levels, and glycerophospholipids alteration, including reduced levels of phospholipids that bind polyunsaturated fatty acids (PUFAs), such as DHA. Interestingly, this PC fatty acid compositional change is similar to that observed in skeletal muscles of denervated and Duchenne muscular dystrophy mouse models. These are accompanied by decrease of GDE5 expression, suggesting a regulatory role of GDE5 activity for glycerophospholipid profiles. Furthermore, a DHA-rich diet enhances contractile force and lowers fatigue resistance, suggesting a functional relationship between PC fatty acid composition and muscle function. Finally, skinned fiber experiments show that GDE5 loss increases the probability of the ryanodine receptor opening and lowers the maximum Ca2+-activated force. Collectively, GDE5 activity plays roles in PC and glucose/glycogen metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Rahmawati Aisyah
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | | | - Daiki Watanabe
- Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, Japan
- Graduate School of Sport and Health Sciences, Osaka University of Health and Sport Sciences, Osaka, Japan
| | - Yoshiko Nakagawa
- Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Tetsushi Sakuma
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Felix Nitschke
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Minako Nakamura
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Koji Sato
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Kaori Nakahata
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Chihiro Yokoyama
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Charlotte R Marchioni
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Takahiko Shimizu
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Yusuke Sotomaru
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Toru Takeo
- Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Takashi Izumi
- Graduate School of Medicine, Gunma University, Gunma, Japan
- Faculty of Health Care, Teikyo Heisei University, Tokyo, Japan
| | - Shinji Miura
- Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Berge A Minassian
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Takashi Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Masanobu Wada
- Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, Japan
| | - Noriyuki Yanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
7
|
Liss KHH, Mousa M, Bucha S, Lutkewitte A, Allegood J, Cowart LA, Finck BN. Dynamic changes in the mouse hepatic lipidome following warm ischemia reperfusion injury. Sci Rep 2024; 14:3584. [PMID: 38351300 PMCID: PMC10864394 DOI: 10.1038/s41598-024-54122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
Liver failure secondary to metabolic dysfunction-associated steatotic liver disease (MASLD) has become the most common cause for liver transplantation in many parts of the world. Moreover, the prevalence of MASLD not only increases the demand for liver transplantation, but also limits the supply of suitable donor organs because steatosis predisposes grafts to ischemia-reperfusion injury (IRI). There are currently no pharmacological interventions to limit hepatic IRI because the mechanisms by which steatosis leads to increased injury are unclear. To identify potential novel mediators of IRI, we used liquid chromatography and mass spectrometry to assess temporal changes in the hepatic lipidome in steatotic and non-steatotic livers after warm IRI in mice. Our untargeted analyses revealed distinct differences between the steatotic and non-steatotic response to IRI and highlighted dynamic changes in lipid composition with marked changes in glycerophospholipids. These findings enhance our knowledge of the lipidomic changes that occur following IRI and provide a foundation for future mechanistic studies. A better understanding of the mechanisms underlying such changes will lead to novel therapeutic strategies to combat IRI.
Collapse
Affiliation(s)
- Kim H H Liss
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Muhammad Mousa
- Department of Medicine, Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Shria Bucha
- Washington University in St. Louis, St. Louis, MO, USA
| | - Andrew Lutkewitte
- Department of Medicine, Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Brian N Finck
- Department of Medicine, Division of Nutritional Science and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
8
|
Zhu H, Wu Y, Zhuang Z, Xu J, Chen F, Wang Q, Tang Q. Ampelopsis japonica aqueous extract improves ovulatory dysfunction in PCOS by modulating lipid metabolism. Biomed Pharmacother 2024; 170:116093. [PMID: 38159378 DOI: 10.1016/j.biopha.2023.116093] [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: 10/11/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a highly prevalent endocrine and metabolic disorder that is closely associated with the proliferation and apoptosis of ovarian granulosa cells (GCs). Ampelopsis japonica (AJ) is the dried tuberous root of Ampelopsis japonica (Thunb.) Makino (A. japonica), with anti-inflammatory, antioxidant, antibacterial, antiviral, wound-healing, and antitumor properties; however, it is unclear whether this herb has a therapeutic effect on PCOS. Therefore, this study aimed to investigate the pharmacological effect of AJ on PCOS and reveal its potential mechanism of action. A PCOS rat model was established using letrozole. After establishing the PCOS model, the rats received oral treatment of AJ and Diane-35 (Positive drug: ethinylestradiol + cyproterone tablets) for 2 weeks. Lipidomics was conducted using liquid-phase mass spectrometry and chromatography. AJ significantly regulated serum hormone levels and attenuated pathological variants in the ovaries of rats with PCOS. Furthermore, AJ significantly reduced the apoptotic rate of ovarian GCs. Lipidomic analysis revealed that AJ modulated glycerolipid and glycerophospholipid metabolic pathways mediated by lipoprotein lipase (Lpl), diacylglycerol choline phosphotransferase (Chpt1), and choline/ethanolamine phosphotransferase (Cept1). Therefore, we established that AJ may reduce ovarian GC apoptosis by modulating lipid metabolism, ultimately improving ovulatory dysfunction in PCOS. Therefore, AJ is a novel candidate for PCOS treatment.
Collapse
Affiliation(s)
- Huiqing Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Yuanyuan Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Ziming Zhuang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Jing Xu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Feilong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Qirui Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China.
| | - Qingfa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China.
| |
Collapse
|
9
|
Huang Z. Evidence that Alzheimer's Disease Is a Disease of Competitive Synaptic Plasticity Gone Awry. J Alzheimers Dis 2024; 99:447-470. [PMID: 38669548 PMCID: PMC11119021 DOI: 10.3233/jad-240042] [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: 04/28/2024]
Abstract
Mounting evidence indicates that a physiological function of amyloid-β (Aβ) is to mediate neural activity-dependent homeostatic and competitive synaptic plasticity in the brain. I have previously summarized the lines of evidence supporting this hypothesis and highlighted the similarities between Aβ and anti-microbial peptides in mediating cell/synapse competition. In cell competition, anti-microbial peptides deploy a multitude of mechanisms to ensure both self-protection and competitor elimination. Here I review recent studies showing that similar mechanisms are at play in Aβ-mediated synapse competition and perturbations in these mechanisms underpin Alzheimer's disease (AD). Specifically, I discuss evidence that Aβ and ApoE, two crucial players in AD, co-operate in the regulation of synapse competition. Glial ApoE promotes self-protection by increasing the production of trophic monomeric Aβ and inhibiting its assembly into toxic oligomers. Conversely, Aβ oligomers, once assembled, promote the elimination of competitor synapses via direct toxic activity and amplification of "eat-me" signals promoting the elimination of weak synapses. I further summarize evidence that neuronal ApoE may be part of a gene regulatory network that normally promotes competitive plasticity, explaining the selective vulnerability of ApoE expressing neurons in AD brains. Lastly, I discuss evidence that sleep may be key to Aβ-orchestrated plasticity, in which sleep is not only induced by Aβ but is also required for Aβ-mediated plasticity, underlining the link between sleep and AD. Together, these results strongly argue that AD is a disease of competitive synaptic plasticity gone awry, a novel perspective that may promote AD research.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
10
|
Wang X, Wang L, Luo M, Bu Q, Liu C, Jiang L, Xu R, Wang S, Zhang H, Zhang J, Wan X, Li H, Wang Y, Liu B, Zhao Y, Chen Y, Dai Y, Li M, Wang H, Tian J, Zhao Y, Cen X. Integrated lipidomic and transcriptomic analysis reveals clarithromycin-induced alteration of glycerophospholipid metabolism in the cerebral cortex of mice. Cell Biol Toxicol 2023; 39:771-793. [PMID: 34458952 DOI: 10.1007/s10565-021-09646-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023]
Abstract
Clarithromycin (CLA) has been widely used in the treatment of bacterial infection. Research reveals the adverse effects on the central nervous system among patients receiving CLA treatment; whereas, a relevant underlying mechanism remains considerably unclear. According to our research, an integrated lipidomic and transcriptomic analysis was applied to explore the effect of CLA on neurobehavior. CLA treatment caused anxiety-like behaviors dose-dependently during open field as well as elevated plus maze trials on mice. Transcriptomes and LC/MS-MS-based metabolomes were adopted for investigating how CLA affected lipidomic profiling as well as metabolic pathway of the cerebral cortex. CLA exposure greatly disturbed glycerophospholipid metabolism and the carbon chain length of fatty acids. By using whole transcriptome sequencing, we found that CLA significantly downregulated the mRNA expression of CEPT1 and CHPT1, two key enzymes involved in the synthesis of glycerophospholipids, supporting the findings from the lipidomic profiling. Also, CLA causes changes in neuronal morphology and function in vitro, which support the existing findings concerning neurobehavior in vivo. We speculate that altered glycerophospholipid metabolism may be involved in the neurobehavioral effect of CLA. Our findings contribute to understanding the mechanisms of CLA-induced adverse effects on the central nervous system. 1. Clarithromycin treatment caused anxiety-like behavior with dose-dependent response both in the open field and elevated plus maze test in mice; 2. Clarithromycin exposing predominately disturbed the metabolism of glycerophospholipids in the cerebral cortex of mice; 3. Clarithromycin application remarkably attenuated CEPT1 and CHPT1 gene expression, which participate in the last step in the synthesis of glycerophospholipids; 4. The altered glycerophospholipid metabolomics may be involved in the abnormal neurobehavior caused by clarithromycin.
Collapse
Affiliation(s)
- Xiaojie Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Liang Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Mingyi Luo
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Qian Bu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Chunqi Liu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Linhong Jiang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Rui Xu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Shaomin Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Haoluo Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Jiamei Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Xuemei Wan
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Hongchun Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Yonghai Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Bin Liu
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Ying Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Yuanyuan Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Yanping Dai
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Min Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Hongbo Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Jingwei Tian
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Yinglan Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, #1 Keyuan Road, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
11
|
Jin Y, Hu X, Meng F, Luo Q, Liu H, Yang Z. Sevoflurane Exposure of Clinical Doses in Pregnant Rats Induces Vcan Changes without Significant Neural Apoptosis in the Offspring. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020190. [PMID: 36837392 PMCID: PMC9965787 DOI: 10.3390/medicina59020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/17/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023]
Abstract
Background and Objectives: Sevoflurane is a commonly used inhalational anaesthetic in clinics. Prolonged exposure to sevoflurane can induce significant changes in lipid metabolism and neuronal damage in the developing brain. However, the effect of exposure of pregnant rats to clinical doses of sevoflurane remains unclear. Materials and Methods: Twenty-eight pregnant rats were randomly and equally divided into sevoflurane exposure (S) group, control (C) and a blank group at gestational day (G) 18; Rats in S group received 2% sevoflurane with 98% oxygen for 6 h in an anesthetizing chamber, while C group received 100% oxygen at an identical flow rate for 6 h in an identical chamber. Partial least squares discriminant analysis (PLS-DA), ultra performance liquid chromatography/time-of-flight mass spectrometry(UPLC/TOF-MS) and MetaboAnalyst were used to analysis acquire metabolomics profiles, and immunohistochemical changes of neuronalapoptosis in hippocampus and cortex of neonatal rats were also analyzed. Results: This study aimed to explore lipidomics and transcriptomics changes related to 2% sevoflurane exposure for 6 h in the developing brains of newborn offspring rats. Ultra-performance liquid chromatography/time-of-flight mass spectrometry (UPLC/TOF-MS) and RNA sequencing (RNA-seq) analyses were used to acquire metabolomics and transcriptomics profiles. We used RNA-seq to analyse the expression of the coding and non-coding transcripts in neural cells of the cerebral cortex. No significant differences in arterial oxygen tension (PaO2), arterial carbon dioxide tension (PaCO2), or arterial blood gas were found between the groups. The relative standard deviation (RSD) of retention times was <1.53%, and the RSDs of peak areas ranged from 2.13% to 8.51%. Base peak chromatogram (BPC) profiles showed no differences between the groups. We evaluated the partial least square-discriminant analysis (PLS-DA) model. In negative ion mode, R2X was over 70%, R2Y was over 93%, and Q2 (cum) was over 80%. Cell apoptosis was not remarkably enhanced by TUNEL and haematoxylin and eosin (HE) staining in the sevoflurane-exposed group compared to the control group (p > 0.05). Glycerophospholipid (GP) and sphingolipid metabolism disturbances might adversely influence neurodevelopment in offspring. The expression of mRNAs (Vcan gene, related to neuronal development, function and repair) of the sevoflurane group was significantly increased in the differential genes by qRT-PCR verification. Conclusions: GP and sphingolipid metabolism homeostasis may be potential therapeutic approaches against inhalational anaesthetic-induced neurodegenerative disorders. Meanwhile, sevoflurane-induced Vcan changes indicated some lipidomic and transcriptomic changes, even if neural cell apoptosis was not significantly changed in the usual clinical dose of sevoflurane exposure.
Collapse
Affiliation(s)
- Yi Jin
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200025, China
- Shanghai Municipal Key Clinical Specialty, Shanghai 200025, China
- Department of Anesthesiology, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Xiaoxue Hu
- Department of Anesthesiology, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Fanhua Meng
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qing Luo
- Department of Anesthesiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Henry Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, The University of Pennsylvania, 3401 Spruce Street, Philadelphia, PA 19104, USA
- Correspondence: (H.L.); (Z.Y.)
| | - Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200025, China
- Shanghai Municipal Key Clinical Specialty, Shanghai 200025, China
- Correspondence: (H.L.); (Z.Y.)
| |
Collapse
|
12
|
Transcriptomics and Lipid Metabolomics Analysis of Subcutaneous, Visceral, and Abdominal Adipose Tissues of Beef Cattle. Genes (Basel) 2022; 14:genes14010037. [PMID: 36672778 PMCID: PMC9858949 DOI: 10.3390/genes14010037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Fat deposition traits are influenced by genetics and environment, which affect meat quality, growth rate, and energy metabolism of domestic animals. However, at present, the molecular mechanism of fat deposition is not entirely understood in beef cattle. Therefore, the current study conducted transcriptomics and lipid metabolomics analysis of subcutaneous, visceral, and abdominal adipose tissue (SAT, VAT, and AAT) of Huaxi cattle to investigate the differences among these adipose tissues and systematically explore how candidate genes interact with metabolites to affect fat deposition. These results demonstrated that compared with SAT, the gene expression patterns and metabolite contents of VAT and AAT were more consistent. Particularly, SCD expression, monounsaturated fatty acid (MUFA) and triglyceride (TG) content were higher in SAT, whereas PCK1 expression and the contents of saturated fatty acid (SFA), diacylglycerol (DG), and lysoglycerophosphocholine (LPC) were higher in VAT. Notably, in contrast to PCK1, 10 candidates including SCD, ELOVL6, ACACA, and FABP7 were identified to affect fat deposition through positively regulating MUFA and TG, and negatively regulating SFA, DG, and LPC. These findings uncovered novel gene resources and offered a theoretical basis for future investigation of fat deposition in beef cattle.
Collapse
|
13
|
Lo Van A, Bernoud-Hubac N, Lagarde M. Esterification of Docosahexaenoic Acid Enhances Its Transport to the Brain and Its Potential Therapeutic Use in Brain Diseases. Nutrients 2022; 14:4550. [PMID: 36364810 PMCID: PMC9656701 DOI: 10.3390/nu14214550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 10/22/2023] Open
Abstract
Docosahexaenoic acid-containing lysophosphatidylcholine (DHA-LysoPC) is presented as the main transporter of DHA from blood plasma to the brain. This is related to the major facilitator superfamily domain-containing protein 2A (Mfsd2a) symporter expression in the blood-brain barrier that recognizes the various lyso-phospholipids that have choline in their polar head. In order to stabilize the DHA moiety at the sn-2 position of LysoPC, the sn-1 position was esterified by the shortest acetyl chain, creating the structural phospholipid 1-acetyl,2-docosahexaenoyl-glycerophosphocholine (AceDoPC). This small structure modification allows the maintaining of the preferential brain uptake of DHA over non-esterified DHA. Additional properties were found for AceDoPC, such as antioxidant properties, especially due to the aspirin-like acetyl moiety, as well as the capacity to generate acetylcholine in response to the phospholipase D cleavage of the polar head. Esterification of DHA within DHA-LysoPC or AceDoPC could elicit more potent neuroprotective effects against neurological diseases.
Collapse
Affiliation(s)
- Amanda Lo Van
- Univ Lyon, INSA Lyon, CNRS, LaMCoS, UMR5259, 69621 Villeurbanne, France
| | | | | |
Collapse
|
14
|
Choi J, Song I, Lee S, You M, Kwon J. Protective Effects of Phosphatidylcholine against Hepatic and Renal Cell Injury from Advanced Glycation End Products. Medicina (B Aires) 2022; 58:medicina58111519. [PMID: 36363476 PMCID: PMC9692888 DOI: 10.3390/medicina58111519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022] Open
Abstract
Background and Objectives: Receptors of the advanced glycation products (RAGE) are activated to promote cell death and contributes to chronic diseases such as diabetes and inflammation. Advanced glycation end products (AGEs), which interact with RAGE are complex compounds synthesized during diabetes development and are presumed to play a significant role in pathogenesis of diabetes. Phosphatidylcholine (PC), a polyunsaturated fatty acid found in egg yolk, mustard, and soybean, is thought to exert anti-inflammatory activity. We investigated the effects of PC on AGEs-induced hepatic and renal cell injury. Materials and Methods: In this study, we evaluated cytokine and NF-κB/MAPK signal pathway activity in AGEs induced human liver (HepG2) cells and human kidney (HK2) cells with and without PC treatment. Results: PC reduced RAGE expression and attenuated levels of inflammatory cytokines and NF-kB/MAPK signaling. Moreover, cells treated with PC exhibited a significant reduction in cytotoxicity, oxidative stress, and inflammatory factor levels. Conclusions: These findings suggest that PC could be an effective functional material for hepatic and renal injury involving with oxidative stress caused by AGEs during diabetic conditions.
Collapse
Affiliation(s)
- Jihye Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Korea
| | - Inbong Song
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Korea
| | - Sangmin Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Korea
| | - Myungjo You
- Laboratory of Veterinary Parasitology, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Korea
| | - Jungkee Kwon
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Korea
- Correspondence: ; Tel.: +82-63-850-0951
| |
Collapse
|
15
|
Roy P, Tomassoni D, Nittari G, Traini E, Amenta F. Effects of choline containing phospholipids on the neurovascular unit: A review. Front Cell Neurosci 2022; 16:988759. [PMID: 36212684 PMCID: PMC9541750 DOI: 10.3389/fncel.2022.988759] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The roles of choline and of choline-containing phospholipids (CCPLs) on the maintenance and progress of neurovascular unit (NVU) integrity are analyzed. NVU is composed of neurons, glial and vascular cells ensuring the correct homeostasis of the blood-brain barrier (BBB) and indirectly the function of the central nervous system. The CCPLs phosphatidylcholine (lecithin), cytidine 5′-diphosphocholine (CDP-choline), choline alphoscerate or α-glyceryl-phosphorylcholine (α-GPC) contribute to the modulation of the physiology of the NVU cells. A loss of CCPLs contributes to the development of neurodegenerative diseases such as Alzheimer’s disease, multiple sclerosis, Parkinson’s disease. Our study has characterized the cellular components of the NVU and has reviewed the effect of lecithin, of CDP-choline and α-GPC documented in preclinical studies and in limited clinical trials on these compounds. The interesting results obtained with some CCPLs, in particular with α-GPC, probably would justify reconsideration of the most promising molecules in larger attentively controlled studies. This can also contribute to better define the role of the NVU in the pathophysiology of brain disorders characterized by vascular impairment.
Collapse
Affiliation(s)
- Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Giulio Nittari
- School of Medicinal and Health Products Sciences, University of Camerino, Camerino, Italy
| | - Enea Traini
- School of Medicinal and Health Products Sciences, University of Camerino, Camerino, Italy
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, University of Camerino, Camerino, Italy
- *Correspondence: Francesco Amenta,
| |
Collapse
|
16
|
Chen G, Guo L, Zhao X, Ren Y, Chen H, Liu J, Jiang J, Liu P, Liu X, Hu B, Wang N, Peng H, Xu G, Tao H. Serum Metabonomics Reveals Risk Factors in Different Periods of Cerebral Infarction in Humans. Front Mol Biosci 2022; 8:784288. [PMID: 35242810 PMCID: PMC8887861 DOI: 10.3389/fmolb.2021.784288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/31/2021] [Indexed: 12/26/2022] Open
Abstract
Studies of key metabolite variations and their biological mechanisms in cerebral infarction (CI) have increased our understanding of the pathophysiology of the disease. However, how metabolite variations in different periods of CI influence these biological processes and whether key metabolites from different periods may better predict disease progression are still unknown. We performed a systematic investigation using the metabonomics method. Various metabolites in different pathways were investigated by serum metabolic profiling of 143 patients diagnosed with CI and 59 healthy controls. Phe-Phe, carnitine C18:1, palmitic acid, cis-8,11,14-eicosatrienoic acid, palmitoleic acid, 1-linoleoyl-rac-glycerol, MAG 18:1, MAG 20:3, phosphoric acid, 5α-dihydrotestosterone, Ca, K, and GGT were the major components in the early period of CI. GCDCA, glycocholate, PC 36:5, LPC 18:2, and PA showed obvious changes in the intermediate time. In contrast, trans-vaccenic acid, linolenic acid, linoleic acid, all-cis-4,7,10,13,16-docosapentaenoic acid, arachidonic acid, DHA, FFA 18:1, FFA 18:2, FFA 18:3, FFA 20:4, FFA 22:6, PC 34:1, PC 36:3, PC 38:4, ALP, and Crea displayed changes in the later time. More importantly, we found that phenylalanine metabolism, medium-chain acylcarnitines, long-chain acylcarnitines, choline, DHEA, LPC 18:0, LPC 18:1, FFA 18:0, FFA 22:4, TG, ALB, IDBIL, and DBIL played vital roles in the development of different periods of CI. Increased phenylacetyl-L-glutamine was detected and may be a biomarker for CI. It was of great significance that we identified key metabolic pathways and risk metabolites in different periods of CI different from those previously reported. Specific data are detailed in the Conclusion section. In addition, we also explored metabolite differences of CI patients complicated with high blood glucose compared with healthy controls. Further work in this area may inform personalized treatment approaches in clinical practice for CI by experimentally elucidating the pathophysiological mechanisms.
Collapse
Affiliation(s)
- Guoyou Chen
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Li Guo
- Department of Anesthesia, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Xinjie Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yachao Ren
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Hongyang Chen
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Jincheng Liu
- Academic Affairs Office, Harbin Medical University-Daqing, Daqing, China
| | - Jiaqi Jiang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Peijia Liu
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoying Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Bo Hu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Na Wang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Haisheng Peng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Guowang Xu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Haiquan Tao
- Department of Neurosurgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Cerebrovascular Diseases Department, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
17
|
Tang H, Li Y, Kakinen A, Andrikopoulos N, Sun Y, Kwak E, Davis TP, Ding F, Ke PC. Graphene quantum dots obstruct the membrane axis of Alzheimer's amyloid beta. Phys Chem Chem Phys 2021; 24:86-97. [PMID: 34878460 PMCID: PMC8771921 DOI: 10.1039/d1cp04246g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a primary form of dementia with debilitating consequences, but no effective cure is available. While the pathophysiology of AD remains multifactorial, the aggregation of amyloid beta (Aβ) mediated by the cell membrane is known to be the cause for the neurodegeneration associated with AD. Here we examined the effects of graphene quantum dots (GQDs) on the obstruction of the membrane axis of Aβ in its three representative forms of monomers (Aβ-m), oligomers (Aβ-o), and amyloid fibrils (Aβ-f). Specifically, we determined the membrane fluidity of neuroblastoma SH-SY5Y cells perturbed by the Aβ species, especially by the most toxic Aβ-o, and demonstrated their recovery by GQDs using confocal fluorescence microscopy. Our computational data through discrete molecular dynamics simulations further revealed energetically favorable association of the Aβ species with the GQDs in overcoming peptide-peptide aggregation. Overall, this study positively implicated GQDs as an effective agent in breaking down the membrane axis of Aβ, thereby circumventing adverse downstream events and offering a potential therapeutic solution for AD.
Collapse
Affiliation(s)
- Huayuan Tang
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Yuhuan Li
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Nicholas Andrikopoulos
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yunxiang Sun
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China
| | - Eunbi Kwak
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia,The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Thomas P. Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia,The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| |
Collapse
|
18
|
Study of the potential neuroprotective effect of Dunaliella salina extract in SH-SY5Y cell model. Anal Bioanal Chem 2021; 414:5357-5371. [PMID: 34923590 PMCID: PMC9242911 DOI: 10.1007/s00216-021-03819-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/03/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023]
Abstract
Alzheimer’s disease (AD) is the most common form of dementia caused by a progressive loss of neurons from different regions of the brain. This multifactorial pathophysiology has been widely characterized by neuroinflammation, extensive oxidative damage, synaptic loss, and neuronal cell death. In this sense, the design of multi-target strategies to prevent or delay its progression is a challenging goal. In the present work, different in vitro assays including antioxidant, anti-inflammatory, and anti-cholinergic activities of a carotenoid-enriched extract from Dunaliella salina microalgae obtained by supercritical fluid extraction are studied. Moreover, its potential neuroprotective effect in the human neuron-like SH-SY5Y cell model against remarkable hallmarks of AD was also evaluated. In parallel, a comprehensive metabolomics study based on the use of charged-surface hybrid chromatography (CSH) and hydrophilic interaction liquid chromatography (HILIC) coupled to high-resolution tandem mass spectrometry (Q-TOF MS/MS) was applied to evaluate the effects of the extract on the metabolism of the treated cells. The use of advanced bioinformatics and statistical tools allowed the identification of more than 314 metabolites in SH-SY5Y cells, of which a great number of phosphatidylcholines, triacylglycerols, and fatty acids were significantly increased, while several phosphatidylglycerols were decreased, compared to controls. These lipidomic changes in cells along with the possible role exerted by carotenoids and other minor compounds on the cell membrane might explain the observed neuroprotective effect of the D. salina extract. However, future experiments using in vivo models to corroborate this hypothesis must be carried out.
Collapse
|
19
|
Gallego R, Suárez-Montenegro ZJ, Ibáñez E, Herrero M, Valdés A, Cifuentes A. In vitro Neuroprotective Potential and Lipidomics Study of Olive Leaves Extracts Enriched in Triterpenoids. Front Nutr 2021; 8:769218. [PMID: 34708068 PMCID: PMC8542692 DOI: 10.3389/fnut.2021.769218] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's Disease (AD) is the most common form of dementia that is associated with extracellular amyloid beta (Aβ) plaque formation. Genetic, environmental, and nutrition factors have been suggested as contributors to oxidative stress and neuroinflammation events that are connected to AD etiology, and secondary metabolites, such as triterpenes, have shown promising results in AD prevention. In this work, the neuroprotective and anti-inflammatory potential of an olive leaves fraction enriched in triterpenoid compounds obtained using supercritical fluid extraction (SFE) and dynamic adsorption/desorption using sea sand as adsorbent has been performed. In addition, a comprehensive lipidomics study of the response of SH-SY5Y neuroblastoma cell line to this fraction was carried out using advanced analytical methodologies, namely, charged-surface hybrid chromatography-quadrupole-time of flight mass spectrometry (CSH-Q-TOF MS/MS). The use of freely available lipidomic annotation tools and databases, and stringent cut-off filters allowed the annotation of more than 250 intracellular lipids. Advanced bioinformatics and statistical tools showed a number of phosphatidylcholines and phosphatidylethanolamines significantly increased, which could explain the protection against the cell death caused by Aβ1-42. Moreover, several triacylglycerols were found decreased. These results suggest triterpenoids from olive leaves as good neuroprotective candidates, and open a new gate for future experiments using in vivo models to corroborate this hypothesis.
Collapse
Affiliation(s)
- Rocío Gallego
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Zully J. Suárez-Montenegro
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
- Departamento de Procesos Industriales, Facultad de Ingeniería Agroindustrial, Universidad de Nariño, Pasto, Colombia
| | - Elena Ibáñez
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Miguel Herrero
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Alberto Valdés
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| | - Alejandro Cifuentes
- Foodomics Laboratory, Institute of Food Science Research (CIAL, CSIC), Madrid, Spain
| |
Collapse
|
20
|
Kuo YC, Ng IW, Rajesh R. Glutathione- and apolipoprotein E-grafted liposomes to regulate mitogen-activated protein kinases and rescue neurons in Alzheimer's disease. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112233. [PMID: 34225874 DOI: 10.1016/j.msec.2021.112233] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 01/29/2023]
Abstract
Neurodegenerative disorders, such as Alzheimer's disease (AD), present biomedical challenges due to inability of pharmaceuticals to permeate the blood-brain barrier (BBB) and lack of therapeutic specificity in definite sites against multiple pathologies. Phosphatidylcholine (PC)-liposomes carrying curcumin (CURC), quercetin (QU), epigallocatechin gallate (EGCG) and rosmarinic acid (RA) with crosslinked glutathione (GSH) and apolipoprotein E (ApoE) were fabricated to recognize brain microvascular endothelial cells and amyloid beta (Aβ), and reduce tau protein hyperphosphorylation for AD management. Addition of stearic acid to liposomal bilayers ameliorated particle stability, promoted drug entrapment efficiency, and prolonged drug release duration. The triple targeting liposomes boosted the capability of CURC, QU, EGCG and RA for crossing the BBB with the assistance of grafted GSH and ApoE and docking Aβ around SK-N-MC cells using ApoE and PC. Moreover, GSH-ApoE-PC-liposomes benefited the 4 medicines in simultaneously transporting to Aβ1-42-insulted neurons, in functioning against hyperphosphorylated mitogen-activated protein kinases, including p-c-Jun N-terminal protein kinase, p-extracellular signal-regulated protein kinase 1/2 and p-p38, in downregulating tau protein at S202, caspase-3 and interleukin-6, and in upregulating p-cyclic adenosine monophosphate response element-binding protein. GSH-ApoE-PC-liposomes can be promising colloidal carriers in delivering CURC, QU, EGCG and RA to degenerated neural tissue in a controlled manner, targeting pathological factors for neuroprotection, and raising preclinical effectualness for AD treatment.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC; Advanced Institute of Manufacturing with High-tech Innovations, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC.
| | - I-Wen Ng
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| |
Collapse
|
21
|
Li Y, Tang H, Andrikopoulos N, Javed I, Cecchetto L, Nandakumar A, Kakinen A, Davis TP, Ding F, Ke PC. The membrane axis of Alzheimer's nanomedicine. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000040. [PMID: 33748816 PMCID: PMC7971452 DOI: 10.1002/anbr.202000040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Alzheimer's disease (AD) is a major neurological disorder impairing its carrier's cognitive function, memory and lifespan. While the development of AD nanomedicine is still nascent, the field is evolving into a new scientific frontier driven by the diverse physicochemical properties and theranostic potential of nanomaterials and nanocomposites. Characteristic to the AD pathology is the deposition of amyloid plaques and tangles of amyloid beta (Aβ) and tau, whose aggregation kinetics may be curbed by nanoparticle inhibitors via sequence-specific targeting or nonspecific interactions with the amyloidogenic proteins. As literature implicates cell membrane as a culprit in AD pathogenesis, here we summarize the membrane axis of AD nanomedicine and present a new rationale that the field development may greatly benefit from harnessing our existing knowledge of Aβ-membrane interaction, nanoparticle-membrane interaction and Aβ-nanoparticle interaction.
Collapse
Affiliation(s)
- Yuhuan Li
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai, 200032, China
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Huayuan Tang
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Nicholas Andrikopoulos
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Luca Cecchetto
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Department of Chemical and Pharmaceutical Science, University of Trieste, Via Licio Giorgieri 1, 34127 Trieste, Italy
| | - Aparna Nandakumar
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Thomas P. Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Pu Chun Ke
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai, 200032, China
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| |
Collapse
|
22
|
Okuda M, Fujita Y, Sugimoto H. The Additive Effects of Low Dose Intake of Ferulic Acid, Phosphatidylserine and Curcumin, Not Alone, Improve Cognitive Function in APPswe/PS1dE9 Transgenic Mice. Biol Pharm Bull 2020; 42:1694-1706. [PMID: 31582657 DOI: 10.1248/bpb.b19-00332] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and its prevention and treatment is a worldwide issue. Many natural components considered to be effective against AD have been identified. However, almost all clinical trials of these components for AD reported inconclusive results. We thought that multiple factors such as amyloid β (Aβ) and tau progressed the pathology of AD and that a therapeutic effect would be obtained by using multiple active ingredients with different effects. Thus, in this study, we treated ferulic acid (FA), phosphatidylserine (PS) and curcumin (Cur) in combination or alone to APPswe/PS1dE9 transgenic mice and evaluated cognitive function by Y-maze test. Consequently, only the three-ingredient group exhibited a significant improvement in cognitive function compared to the control group. In addition, we determined the amounts of Aβ, brain-derived neurotrophic factor (BDNF), interleukin (IL)-1β, acetylcholine and phosphorylated tau in the mouse brains after the treatment. In the two-ingredient (FA and PS) group, a significant decrease in IL-1β and an increasing trend in acetylcholine were observed. In the Cur group, significant decreases in Aβ and phosphorylated tau and an increasing trend in BDNF were observed. In the three-ingredient group, all of them were observed. These results indicate that the intake of multiple active ingredients with different mechanisms of action for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Michiaki Okuda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University.,Green Tech Co., Ltd
| | - Yuki Fujita
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University.,Green Tech Co., Ltd
| | | |
Collapse
|
23
|
Yoon SJ, Long NP, Jung KH, Kim HM, Hong YJ, Fang Z, Kim SJ, Kim TJ, Anh NH, Hong SS, Kwon SW. Systemic and Local Metabolic Alterations in Sleep-Deprivation-Induced Stress: A Multiplatform Mass-Spectrometry-Based Lipidomics and Metabolomics Approach. J Proteome Res 2019; 18:3295-3304. [PMID: 31313932 DOI: 10.1021/acs.jproteome.9b00234] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sleep deprivation (SD) is known to be associated with metabolic disorders and chronic diseases. Complex metabolic alterations induced by SD at omics scale and the associated biomarker candidates have been proposed. However, in vivo systemic and local metabolic shift patterns of the metabolome and lipidome in acute and chronic partial SD models remain to be elucidated. In the present study, the serum, hypothalamus, and hippocampus CA1 of sleep-deprived rats (SD rats) from acute and chronic sleep restriction models were analyzed using three different omics platforms for the discovery and mechanistic assessment of systemic and local SD-induced dysregulated metabolites. We found a similar pattern of systemic metabolome alterations between two models, for which the area under the curve (AUC) of receiver operating characteristic curves was AUC = 0.847 and 0.930 with the pseudotargeted and untargeted metabolomics approach, respectively. However, SD-induced systemic lipidome alterations were significantly different and appeared to be model-dependent (AUC = 0.374). Comprehensive pathway analysis of the altered lipidome and metabolome in the hypothalamus indicated the abnormal behavior of eight metabolic and lipid metabolic pathways. The metabolic alterations of the hippocampus CA1 was subtle in two SD models. Collectively, these results extend our understanding of the quality of sleep and suggest metabolic targets in developing diagnostic biomarkers for better SD control.
Collapse
Affiliation(s)
- Sang Jun Yoon
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Nguyen Phuoc Long
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Kyung-Hee Jung
- Department of Biomedical Sciences, College of Medicine , Inha University , Incheon 22212 , Republic of Korea
| | - Hyung Min Kim
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Yu Jin Hong
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Zhenghuan Fang
- Department of Biomedical Sciences, College of Medicine , Inha University , Incheon 22212 , Republic of Korea
| | - Sun Jo Kim
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Tae Joon Kim
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Nguyen Hoang Anh
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine , Inha University , Incheon 22212 , Republic of Korea
| | - Sung Won Kwon
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| |
Collapse
|
24
|
Tirumanyam M, Nadella R, Kondammagari S, Borelli DPR, Nannepaga JS. Bacopa phospholipid complex retrieves aluminum maltolate complex-induced oxidative stress and apoptotic alterations in the brain regions of albino rat. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:12071-12079. [PMID: 30827024 DOI: 10.1007/s11356-019-04624-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/19/2019] [Indexed: 06/09/2023]
Abstract
Highly bioavailable plant phospholipid complex that can reverse aluminum maltolate (AlM)-induced toxicity is not yet reported. Hence, the present study was planned to investigate the impact of oxidative stress and apoptotic changes provoked by Al and ameliorative role of Bacopa phospholipid complex (BPC) in albino rats. The levels of antioxidant enzymes such as superoxide dismutase (SOD), catalase activity (CAT), glutathione peroxidase (GPx), and thiobarbituric acid-reactive substance (TBA-RS) were measured and immunohistochemistry analysis of apoptotic markers, Bax and Bcl-2, was done from the four brain regions such as the hippocampus, cerebral cortex, cerebellum, and medulla oblongata. The levels of antioxidant enzymes and apoptotic markers that were decreased on AlM induction showed a significant increase in their levels, almost as observed in the control, when treated with BPC and Bm. Our results indicate that both BPC and Bm showed a therapeutic effect against AlM toxicity; however, it was found that the therapeutic potential of BPC was more pronounced than Bm against AlM-induced neurotoxicity.
Collapse
Affiliation(s)
- Madhavi Tirumanyam
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women's University), Tirupati, Andhra Pradesh, 517 502, India
| | - Rasajna Nadella
- Department of Biological Sciences, IIIT-Srikakulam, Etcherla, 532 410, India
| | - Saritha Kondammagari
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women's University), Tirupati, Andhra Pradesh, 517 502, India
| | | | - John Sushma Nannepaga
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women's University), Tirupati, Andhra Pradesh, 517 502, India.
| |
Collapse
|
25
|
Structure of amyloid β 25-35 in lipid environment and cholesterol-dependent membrane pore formation. Sci Rep 2019; 9:2689. [PMID: 30804528 PMCID: PMC6389947 DOI: 10.1038/s41598-019-38749-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/08/2019] [Indexed: 01/14/2023] Open
Abstract
The amyloid β (Aβ) peptide and its shorter variants, including a highly cytotoxic Aβ25–35 peptide, exert their neurotoxic effect during Alzheimer’s disease by various mechanisms, including cellular membrane permeabilization. The intrinsic polymorphism of Aβ has prevented the identification of the molecular basis of Aβ pore formation by direct structural methods, and computational studies have led to highly divergent pore models. Here, we have employed a set of biophysical techniques to directly monitor Ca2+-transporting Aβ25–35 pores in lipid membranes, to quantitatively characterize pore formation, and to identify the key structural features of the pore. Moreover, the effect of membrane cholesterol on pore formation and the structure of Aβ25–35 has been elucidated. The data suggest that the membrane-embedded peptide forms 6- or 8-stranded β-barrel like structures. The 8-stranded barrels may conduct Ca2+ ions through an inner cavity, whereas the tightly packed 6-stranded barrels need to assemble into supramolecular structures to form a central pore. Cholesterol affects Aβ25–35 pore formation by a dual mechanism, i.e., by direct interaction with the peptide and by affecting membrane structure. Collectively, our data illuminate the molecular basis of Aβ membrane pore formation, which should advance both basic and clinical research on Alzheimer’s disease and membrane-associated pathologies in general.
Collapse
|
26
|
Alzheimer's brains show inter-related changes in RNA and lipid metabolism. Neurobiol Dis 2017; 106:1-13. [PMID: 28630030 PMCID: PMC5560656 DOI: 10.1016/j.nbd.2017.06.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) involves changes in both lipid and RNA metabolism, but it remained unknown if these differences associate with AD's cognition and/or post-mortem neuropathology indices. Here, we report RNA-sequencing evidence of inter-related associations between lipid processing, cognition level, and AD neuropathology. In two unrelated cohorts, we identified pathway-enriched facilitation of lipid processing and alternative splicing genes, including the neuronal-enriched NOVA1 and hnRNPA1. Specifically, this association emerged in temporal lobe tissue samples from donors where postmortem evidence demonstrated AD neuropathology, but who presented normal cognition proximate to death. The observed changes further associated with modified ATP synthesis and mitochondrial transcripts, indicating metabolic relevance; accordingly, mass-spectrometry-derived lipidomic profiles distinguished between individuals with and without cognitive impairment prior to death. In spite of the limited group sizes, tissues from persons with both cognitive impairment and AD pathology showed elevation in several drug-targeted genes of other brain, vascular and autoimmune disorders, accompanied by pathology-related increases in distinct lipid processing transcripts, and in the RNA metabolism genes hnRNPH2, TARDBP, CLP1 and EWSR1. To further detect 3'-polyadenylation variants, we employed multiple cDNA primer pairs. This identified variants that showed limited differences in scope and length between the tested cohorts, yet enabled superior clustering of demented and non-demented AD brains versus controls compared to total mRNA expression values. Our findings indicate inter-related cognition-associated differences in AD's lipid processing, alternative splicing and 3'-polyadenylation, calling for pursuing the underlying psychological and therapeutics implications.
Collapse
|
27
|
Xie B, Liu F, Dong X, Wang Y, Liu XM, Sun Y. Modulation effect of acidulated human serum albumin on Cu 2+ -mediated amyloid β-protein aggregation and cytotoxicity under a mildly acidic condition. J Inorg Biochem 2017; 171:67-75. [DOI: 10.1016/j.jinorgbio.2017.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 02/10/2017] [Accepted: 03/19/2017] [Indexed: 12/31/2022]
|
28
|
Khafaga AF. Exogenous phosphatidylcholine supplementation retrieve aluminum-induced toxicity in male albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:15589-15598. [PMID: 28523611 DOI: 10.1007/s11356-017-9151-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/01/2017] [Indexed: 05/25/2023]
Abstract
This study investigated the ameliorative potential of exogenous phosphatidylcholine (PC) against aluminum-induced toxicity in male albino rats. Four groups of rats were used for this study (N = 8): group I served as the control, group II (PC treated) received L-α-phosphatidylcholine (egg yolk-derived) 100 mg/kg bwt/day orally, group III (aluminum treated) received aluminum chloride 100 mg/kg bwt/day orally, and group VI (aluminum + PC treated) received similar oral dose of aluminum and PC (100 mg/kg bwt/day). Treatment was continued for 8 weeks. Results revealed that aluminum chloride treatment leading to a significant elevation in serum aspartate aminotransferase, serum alanine aminotransferase, urea, creatinine, malondialdehyde, serum cytokines (tumor necrosis factor-α, interleukin-6), and brain content of acetylcholine, as well as a significant reduction in serum-reduced glutathione, serum testosterone, and brain content of acetylcholinesterase. Moreover, aluminum administration caused significant histopathological alteration in liver, kidney, brain, testes, and epididymis. Co-treatment with exogenous PC resulted in significant improvement in intensity of histopathologic lesions, serum parameters, testosterone level, proinflammatory cytokines, and oxidative/antioxidative status. However, it does not affect the brain content of acetylcholine and acetylcholinesterase. Conclusively, treatment with exogenous PC can retrieve the adverse effect of aluminum toxicities through its antioxidative and anti-inflammatory properties.
Collapse
Affiliation(s)
- Asmaa Fahmy Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina-Rasheed, Behira, Egypt.
| |
Collapse
|
29
|
Li M, Dong X, Liu Y, Sun Y. Brazilin Inhibits Prostatic Acidic Phosphatase Fibrillogenesis and Decreases its Cytotoxicity. Chem Asian J 2017; 12:1062-1068. [PMID: 28303660 DOI: 10.1002/asia.201700058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/24/2017] [Indexed: 11/06/2022]
Abstract
A 39-amino acid peptide fragment that is derived from prostatic acidic phosphatase (PAP), PAP248-286 , is secreted in large amounts in human semen and forms amyloid fibrils. These fibrils can capture HIV virions and increase the attachment of virions to target cells; as such, they are called a "semen-derived enhancer of virus infection" (SEVI). Therefore, the inhibition of the formation of PAP248-286 amyloid fibrils is of great significance. Herein, we demonstrate that brazilin effectively inhibits PAP248-286 aggregation. The inhibitory effect increases with increasing brazilin concentration. Thioflavin T fluorescence assays and TEM observations confirmed that a few fibrils formed when brazilin was present with PAP248-286 in an equimolar concentration. Circular dichroism spectroscopy indicated that brazilin inhibited the secondary structural transitions from α-helices and random coils into β-sheets. Cytotoxicity assays showed that brazilin significantly decreased the cytotoxicity of the fibrils at 0.01 mmol L-1 . Isothermal titration calorimetry revealed that hydrophobic interactions were the main driving force for the binding of brazilin to the PAP248-286 monomer (dissociation constant, 4.03 μmol L-1 ), and that the binding affinity of brazilin for the fibrils was at least three orders of magnitude lower than that for the monomer. These results indicate that brazilin holds great potential as a small-molecule agent against SEVIs.
Collapse
Affiliation(s)
- Ming Li
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300354, P. R. China
| | - Xiaoyan Dong
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300354, P. R. China
| | - Yang Liu
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, College of Science, Shantou University, Shantou, Guangdong, 515063, P. R. China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300354, P. R. China
| |
Collapse
|
30
|
Effects of the duration of hyperlipidemia on cerebral lipids, vessels and neurons in rats. Lipids Health Dis 2017; 16:26. [PMID: 28143622 PMCID: PMC5282812 DOI: 10.1186/s12944-016-0401-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/27/2016] [Indexed: 01/09/2023] Open
Abstract
Background The present study was designed to investigate the effects of hyperlipidemia on the cerebral lipids, vessels and neurons of rats, and to provide experimental evidence for subsequent intervention. Method One hundred adult SD rats, half of which were male and half of which were female, were randomly divided into five groups on the basis of serum total cholesterol (TC) levels. Four groups were fed a hypercholesterolemic diet (rat chow supplemented with 4% cholesterol, 1% cholic acid and 0.5% thiouracil – this is also called a CCT diet) for periods of 1 week, 2 weeks, 3 weeks and 4 weeks, respectively. A control group was included. The levels of serum lipids, cerebral lipids, free fatty acids (FFA), interleukin-6 (IL-6), interleukin-1 (IL-1), tumor necrosis factor alpha (TNF-α), vascular endothelial growth factor (VEGF), oxidized low density lipoprotein (ox-LDL), A-beta precursor proteins (APP), amyloid beta (Aβ), glial fibrillary acidic protein (GFAP) and tight junction protein Claudin-5 were measured after the experiment. The pathologic changes and apoptosis of the rat brains were evaluated. Results Compared with the control group, after 1 week of a CCT diet, the levels of serum total cholesterol (TC), triglycerides (TG), low density lipoprotein cholesterol (LDL-C) and brain triglycerides had increased by 2.40, 1.29 and 1.75 and 0.3 times, respectively. The serum high density lipoprotein cholesterol (HDL-C) had decreased by 0.74 times (P < 0.05) and the expression of IL-1, TNF-α and GFAP in the brains had increased (P < 0.05). In the second week, the expression of FFA and APP in the brains, and the amount of apoptotic neurons, had increased (P < 0.05). In the third week, the levels of VEGF, Ox-LDL and Aβ had increased, and the expression of Claudin-5 had decreased in the brains (P < 0.05). In the fourth week, the levels of TC, LDL-C and the amount of apoptotic neurons had increased (P < 0.05). The correlation analysis showed a positive correlation among FFA, TNF-α, VEGF, ox-LDL, Aβ, GFAP and neuronal apoptosis in the rat brains, and they all were negatively correlated with Claudin-5 (P < 0.05). Conclusion Hyperlipidemia may activate astrocytes by means of high levels of TG that will have direct toxic effects on the cerebral vessels and neurons by causing the secretion of TNF-α and IL-1 in the brains of rats. In the metabolic procession, brain tissue was shown to generate FFA that aggravated the biosynthesis of ox-LDL. With the extension of the duration of hyperlipidemia, high levels of cerebral TC and LDL-C were shown to aggravate the deposition of Aβ, induce the secretion of VEGF, reduce the expression of tight junction protein Claudin-5 and change the permeability of blood–brain barriers to factors that could damage cerebral vessels and neurons.
Collapse
|
31
|
Ritchie SA, Jayasinge D, Wang L, Goodenowe DB. Improved specificity of serum phosphatidylcholine detection based on side-chain losses during negative electrospray ionization tandem mass spectrometry. Anal Bioanal Chem 2016; 408:7811-7823. [DOI: 10.1007/s00216-016-9884-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 12/13/2022]
|