1
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
2
|
Bassareo V, Maccioni R, Talani G, Zuffa S, El Abiead Y, Lorrai I, Kawamura T, Pantis S, Puliga R, Vargiu R, Lecca D, Enrico P, Peana A, Dazzi L, Dorrestein PC, Sanna PP, Sanna E, Acquas E. Receptor and metabolic insights on the ability of caffeine to prevent alcohol-induced stimulation of mesolimbic dopamine transmission. Transl Psychiatry 2024; 14:391. [PMID: 39341817 PMCID: PMC11438888 DOI: 10.1038/s41398-024-03112-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
The consumption of alcohol and caffeine affects the lives of billions of individuals worldwide. Although recent evidence indicates that caffeine impairs the reinforcing properties of alcohol, a characterization of its effects on alcohol-stimulated mesolimbic dopamine (DA) function was lacking. Acting as the pro-drug of salsolinol, alcohol excites DA neurons in the posterior ventral tegmental area (pVTA) and increases DA release in the nucleus accumbens shell (AcbSh). Here we show that caffeine, via antagonistic activity on A2A adenosine receptors (A2AR), prevents alcohol-dependent activation of mesolimbic DA function as assessed, in-vivo, by brain microdialysis of AcbSh DA and, in-vitro, by electrophysiological recordings of pVTA DA neuronal firing. Accordingly, while the A1R antagonist DPCPX fails to prevent the effects of alcohol on DA function, both caffeine and the A2AR antagonist SCH 58261 prevent alcohol-dependent pVTA generation of salsolinol and increase in AcbSh DA in-vivo, as well as alcohol-dependent excitation of pVTA DA neurons in-vitro. However, caffeine also prevents direct salsolinol- and morphine-stimulated DA function, suggesting that it can exert these inhibitory effects also independently from affecting alcohol-induced salsolinol formation or bioavailability. Finally, untargeted metabolomics of the pVTA showcases that caffeine antagonizes alcohol-mediated effects on molecules (e.g. phosphatidylcholines, fatty amides, carnitines) involved in lipid signaling and energy metabolism, which could represent an additional salsolinol-independent mechanism of caffeine in impairing alcohol-mediated stimulation of mesolimbic DA transmission. In conclusion, the outcomes of this study strengthen the potential of caffeine, as well as of A2AR antagonists, for future development of preventive/therapeutic strategies for alcohol use disorder.
Collapse
Affiliation(s)
- Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Riccardo Maccioni
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| | - Giuseppe Talani
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Irene Lorrai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomoya Kawamura
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sofia Pantis
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberta Puliga
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Romina Vargiu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Daniele Lecca
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Paolo Enrico
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandra Peana
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Laura Dazzi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Enrico Sanna
- Institute of Neuroscience - National Research Council (C.N.R.) of Italy, Cagliari, Italy
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria Monserrato, Monserrato, CA, Italy
| |
Collapse
|
3
|
Navab F, Foshati S, Vajdi M, Askari G, Moeinzadeh F, Heshamtipour H, Mirzaeian S, Rouhani MH. Is there any association between type of dietary fat and quality of life in hemodialysis patients? A cross-sectional study. Front Nutr 2024; 11:1430595. [PMID: 39439523 PMCID: PMC11495156 DOI: 10.3389/fnut.2024.1430595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/11/2024] [Indexed: 10/25/2024] Open
Abstract
Background Hemodialysis (HD) patients have a low quality of life (QOL), and dietary intakes may impact both somatic and psychosocial aspects of QOL. Nevertheless, the relationship between QOL and different dietary fats has not yet been evaluated. Objective The purpose of this study was to assess the association between QOL and the types/quantities of dietary fats intake in HD patients. Methods In this multi-center cross-sectional study, 251 adult patients under dialysis for at least 3 months were included. Participants' dietary intakes were collected using a validated 168-item semi-quantitative FFQ during the past year. Moreover, to assess QOL, Kidney Disease Quality of Life Short Form (KDQOL-SF 1/3) was used. The linear regression between QOL and different types of dietary fats was conducted. p < 0.05 was statistically significant. Results Overall, 66 women and 185 men participated in our study. Regression analysis adjusted for total calorie intake showed that there was a negative association between QOL and total fat (95% CI: -0.187, -0.043), SFA (95% CI: -0.688, -0.143), MUFA (95% CI: -0.389, -0.065) and PUFA (95% CI: -0.401, -0.056) when types of dietary fats were individually included to the regression analysis. When all types of dietary fats were simultaneously entered into the analysis, the association between QOL and MUFA (95% CI: -0.243, 1.031) and PUFA (95% CI: -1.159, 0.084) were attenuated. The regression coefficient for SFA remained significant (95% CI: -0.968, -0.138). Also, there was a marginally significant association between SFA and the risk of low QOL was observed when all types of dietary fats were simultaneously entered into the analysis (OR = 1.051, 95% CI: 0.998-1.104). Conclusion Our investigation found a negative association between SFA consumption and QOL among different types of dietary fats. Furthermore, SFA mediated the relationship between QOL, MUFA, PUFA, and total fat. So, modification of dietary fat intake could enhance QOL in HD patients.
Collapse
Affiliation(s)
- Fatemeh Navab
- Student Research Committee, Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sahar Foshati
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdi Vajdi
- Student Research Committee, Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Firouzeh Moeinzadeh
- Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Houri Heshamtipour
- Student Research Committee, Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Mohammad Hossein Rouhani
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Soltani S, Sangsefidi ZS, Asoudeh F, Torabynasab K, Zeraattalab-Motlagh S, Hejazi M, Khalighi Sikaroudi M, Meshkini F, Razmpoosh E, Abdollahi S. Effect of Low-Fat Diet on Depression Score in Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Clinical Trials. Nutr Rev 2024:nuae069. [PMID: 38899499 DOI: 10.1093/nutrit/nuae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
CONTEXT Current evidence on the effect of a low-fat (LF) diet on depression scores has been inconsistent. OBJECTIVE To explore the effect of an LF diet on depression scores of adults by systematic review and meta-analysis of randomized controlled trials (RCTs). DATA SOURCES The PubMed, ISI Web of Science, Scopus, and CENTRAL databases were searched from inception to June 7, 2023, to identify trials investigating the effect of an LF diet (fat intake ≤30% of energy intake) on the depression score. DATA EXTRACTION Random-effects meta-analyses were used to estimate pooled summary effects of an LF diet on the depression score (as Hedges g). DATA ANALYSIS Finding from 10 trials with 50 846 participants indicated no significant change in depression score following LF diets in comparison with usual diet (Hedges g = -0.11; 95% CI, -0.25 to 0.03; P = 0.12; I2 = 70.7% [for I2, 95% CI, 44%, 85%]). However, a significant improvement was observed in both usual diet and LF diets when the content of protein was 15-20% of calorie intake (LF, normal protein diet: n = 5, Hedges g = -0.21, 95% CI, -0.24 to -0.01, P = 0.04, I2 = 0%; usual, normal protein diet: n = 3, Hedges g = -0.28, 95% CI, -0.51 to -0.05, P = 0.01, I2 = 0%). Sensitivity analysis also found the depression score improved following LF diet intervention in participants without baseline depression. CONCLUSION This study revealed that LF diet may have small beneficial effect on depression score in the studies enrolled mentally healthy participants. Moreover, achieving to adequate dietary protein is likely to be a better intervention than manipulating dietary fat to improve depression scores. However, it is not clear whether this effect will last in the long term. Conducting more studies may change the results due to the low-certainty of evidence. SYSTEMATIC REVIEW REGISTRATION CRD42023420978 (https://www.crd.york.ac.uk/PROSPERO).
Collapse
Affiliation(s)
- Sepideh Soltani
- Research Center of Addiction and Behavioral Sciences, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zohreh Sadat Sangsefidi
- Department of Nutrition, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Farzaneh Asoudeh
- Research Center for Biochemistry and Nutrition in Metabolic Disease, Kashan University of Medical Sciences, Kashan, Iran
| | - Kimia Torabynasab
- Student Research Committee, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mahdi Hejazi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Khalighi Sikaroudi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Meshkini
- Department of Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elham Razmpoosh
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Shima Abdollahi
- Department of Nutrition, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
5
|
Osetrova M, Tkachev A, Mair W, Guijarro Larraz P, Efimova O, Kurochkin I, Stekolshchikova E, Anikanov N, Foo JC, Cazenave-Gassiot A, Mitina A, Ogurtsova P, Guo S, Potashnikova DM, Gulin AA, Vasin AA, Sarycheva A, Vladimirov G, Fedorova M, Kostyukevich Y, Nikolaev E, Wenk MR, Khrameeva EE, Khaitovich P. Lipidome atlas of the adult human brain. Nat Commun 2024; 15:4455. [PMID: 38796479 PMCID: PMC11127996 DOI: 10.1038/s41467-024-48734-y] [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: 12/15/2022] [Accepted: 05/07/2024] [Indexed: 05/28/2024] Open
Abstract
Lipids are the most abundant but poorly explored components of the human brain. Here, we present a lipidome map of the human brain comprising 75 regions, including 52 neocortical ones. The lipidome composition varies greatly among the brain regions, affecting 93% of the 419 analyzed lipids. These differences reflect the brain's structural characteristics, such as myelin content (345 lipids) and cell type composition (353 lipids), but also functional traits: functional connectivity (76 lipids) and information processing hierarchy (60 lipids). Combining lipid composition and mRNA expression data further enhances functional connectivity association. Biochemically, lipids linked with structural and functional brain features display distinct lipid class distribution, unsaturation extent, and prevalence of omega-3 and omega-6 fatty acid residues. We verified our conclusions by parallel analysis of three adult macaque brains, targeted analysis of 216 lipids, mass spectrometry imaging, and lipidome assessment of sorted murine neurons.
Collapse
Affiliation(s)
- Maria Osetrova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Anna Tkachev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Waltraud Mair
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | - Olga Efimova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Ilia Kurochkin
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | - Juat Chin Foo
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore
| | | | | | - Song Guo
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Daria M Potashnikova
- Department of Cell Biology and Histology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander A Gulin
- N. N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, Moscow, Russia
| | - Alexander A Vasin
- N. N. Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | | | - Gleb Vladimirov
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | | | - Evgeny Nikolaev
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute and Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine; National University of Singapore, Singapore, Singapore.
| | | | | |
Collapse
|
6
|
Nilén G, Larsson M, Hyötyläinen T, Keiter SH. A complex mixture of polycyclic aromatic compounds causes embryotoxic, behavioral, and molecular effects in zebrafish larvae (Danio rerio), and in vitro bioassays. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167307. [PMID: 37804991 DOI: 10.1016/j.scitotenv.2023.167307] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/09/2023]
Abstract
Polycyclic aromatic compounds (PACs) are prevalent in the environment, typically found in complex mixtures and high concentrations. Our understanding of the effects of PACs, excluding the 16 priority polycyclic aromatic hydrocarbons (16 PAHs), remains limited. Zebrafish embryos and in vitro bioassays were utilized to investigate the embryotoxic, behavioral, and molecular effects of a soil sample from a former gasworks site in Sweden. Additionally, targeted chemical analysis was conducted to analyze 87 PACs in the soil, fish, water, and plate material. CALUX® assays were used to assess the activation of aryl hydrocarbon and estrogen receptors, as well as the inhibition of the androgen receptor. Larval behavior was measured by analyzing activity during light and darkness and in response to mechanical stimulation. Furthermore, qPCR analyses were performed on a subset of 36 genes associated with specific adverse outcomes, and the total lipid content in the larvae was measured. Exposure to the sample resulted in embryotoxic effects (LC50 = 0.480 mg dry matter soil/mL water). The mixture also induced hyperactivity in darkness and hypoactivity in light and in response to the mechanical stimulus. qPCR analysis revealed differential regulation of 15 genes, including downregulation of opn1sw1 (eye pigmentation) and upregulation of fpgs (heart failure). The sample caused significant responses in three bioassays (ERα-, DR-, and PAH-CALUX), and the exposed larvae exhibited elevated lipid levels. Chemical analysis identified benzo[a]pyrene as the predominant compound in the soil and approximately half of the total PAC concentration was attributed to the 16 PAHs. This study highlights the value of combining in vitro and in vivo methods with chemical analysis to assess toxic mechanisms at specific targets and to elucidate the possible interactions between various pathways in an organism. It also enhances our understanding of the risks associated with environmental mixtures of PACs and their distribution during toxicity testing.
Collapse
Affiliation(s)
- Greta Nilén
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden.
| | - Maria Larsson
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| | - Tuulia Hyötyläinen
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| | - Steffen H Keiter
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| |
Collapse
|
7
|
Carpinter BA, Renhe DC, Bellei JCB, Vieira CD, Rodolphi CM, Ferreira MVR, de Freitas CS, Neto AFDS, Coelho EAF, Mietto BDS, Gomes FLR, Rocha VN, Scopel KKG. DHA-rich fish oil plays a protective role against experimental cerebral malaria by controlling inflammatory and mechanical events from infection. J Nutr Biochem 2024; 123:109492. [PMID: 37866427 DOI: 10.1016/j.jnutbio.2023.109492] [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: 02/04/2023] [Revised: 05/17/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Every year, thousands of children, particularly those under 5 years old, die because of cerebral malaria (CM). Following conventional treatment, approximately 25% of surviving individuals have lifelong severe neurocognitive sequelae. Therefore, improved conventional therapies or effective alternative therapies that prevent the severe infection are crucial. Omega-3 (Ω-3) polyunsaturated fatty acids (PUFAs) are known to have antioxidative and anti-inflammatory effects and protect against diverse neurological disorders, including Alzheimer's and Parkinson's diseases. However, little is known regarding the effects of Ω-3 PUFAs against parasitic infections. In this study, C57BL/6 mice received supplemental treatment of a fish oil rich in the Ω-3 PUFA, docosahexaenoic acid (DHA), which was started 15 days prior to infection with Plasmodium berghei ANKA and was maintained until the end of the study. Animals treated with the highest doses of DHA, 3.0 and 6.0 g/kg body weight, had 60 and 80% chance of survival, respectively, while all nontreated mice died by the 7th day postinfection due to CM. Furthermore, the parasite load during the critical period for CM development (5th to 11th day postinfection) was controlled in treated mice. However, after this period all animals developed high levels of parasitemia until the 20th day of infection. DHA treatment also effectively reduced blood-brain barrier (BBB) damage and brain edema and completely prevented brain hemorrhage and vascular occlusion. A strong anti-inflammatory profile was observed in the brains of DHA-treated mice, as well as, an increased number of neutrophil and reduced number of CD8+ T leukocytes in the spleen. Thus, this is the first study to demonstrate that the prophylactic use of DHA-rich fish oil exerts protective effects against experimental CM, reducing the mechanical and immunological events caused by the P. berghei ANKA infection.
Collapse
Affiliation(s)
- Bárbara Albuquerque Carpinter
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela Chaves Renhe
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Jéssica Correa Bezerra Bellei
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina David Vieira
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Cinthia Magalhães Rodolphi
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Camila Simões de Freitas
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno de Siqueira Mietto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Vinicius Novaes Rocha
- Department of Veterinary Medicine, Research Centre of Pathology and Veterinary Histology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| |
Collapse
|
8
|
Han X, Li Y, Chen X, Pan D, Mo J, Qiu J, Li Y, Chen Y, Huang Y, Shen Q, Tang Y. Platelet-activating factor antagonist-based intensive antiplatelet strategy in acute ischemic stroke: A propensity score matched with network pharmacology analysis. CNS Neurosci Ther 2023; 29:4082-4092. [PMID: 37435773 PMCID: PMC10651968 DOI: 10.1111/cns.14331] [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: 03/25/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Diterpene ginkgolides meglumine injection (DGMI) is a platelet-activating factor receptor (PAFR) antagonist that can be used to treat acute ischemic stroke (AIS). This study evaluated the efficacy and safety of an intensive antiplatelet strategy based on PAFR antagonists and explored the underlying mechanisms of PAFR antagonists in AIS treatment. METHODS This is a retrospective study applying propensity score methods to match AIS patients treated with DGMI to nontreated patients. The primary outcome was functional independence (modified Rankin Scale [mRS] 0-2) at 90 days. The safety outcome was bleeding risk. We used McNemar test to compare the efficacy outcome. Subsequently, the network pharmacology analysis was performed. RESULTS 161 AIS patients treated with DGMI in the study were matched with 161 untreated patients. Compared with untreated patients, DGMI-treated patients had a significantly higher rate of mRS ranking 0-2 at 90 days (82.0% vs. 75.8%, p < 0.001), without increased risk of bleeding. The gene enrichment analysis showed that the overlap genes of DGMI targeted and AIS-related enriched in thrombosis and inflammatory-related signaling pathways. CONCLUSIONS An intensive antiplatelet strategy of DGMI plus traditional antiplatelet agents is effective in treating AIS and may work by mediating post-stroke inflammation and thrombosis.
Collapse
Affiliation(s)
- Xiaoyan Han
- Department of NeurologyFirst People's Hospital of ZhaoqingZhaoqingPeople's Republic of China
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Youjia Li
- Department of NeurologyFirst People's Hospital of ZhaoqingZhaoqingPeople's Republic of China
| | - Xuemin Chen
- Guangdong Medical UniversityZhanjiangPeople's Republic of China
| | - Dong Pan
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Junning Mo
- Department of NeurologyFirst People's Hospital of ZhaoqingZhaoqingPeople's Republic of China
| | - Jiaming Qiu
- Department of NeurologyFirst People's Hospital of ZhaoqingZhaoqingPeople's Republic of China
| | - Yi Li
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Yan Chen
- Department of NeurologyFirst People's Hospital of ZhaoqingZhaoqingPeople's Republic of China
| | - Yan Huang
- Department of NeurologyFirst People's Hospital of ZhaoqingZhaoqingPeople's Republic of China
| | - Qingyu Shen
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Yamei Tang
- Department of Neurology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouPeople's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouPeople's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
9
|
Li Y, Zhang L, Mao M, He L, Wang T, Pan Y, Zhao X, Li Z, Mu X, Qian Y, Qiu J. Multi-omics analysis of a drug-induced model of bipolar disorder in zebrafish. iScience 2023; 26:106744. [PMID: 37207274 PMCID: PMC10189518 DOI: 10.1016/j.isci.2023.106744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/16/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Emerging studies demonstrate that inflammation plays a crucial role in the pathogenesis of bipolar disorder (BD), but the underlying mechanism remains largely unclear. Given the complexity of BD pathogenesis, we performed high-throughput multi-omic profiling (metabolomics, lipidomics, and transcriptomics) of the BD zebrafish brain to comprehensively unravel the molecular mechanism. Our research proved that in BD zebrafish, JNK-mediated neuroinflammation altered metabolic pathways involved in neurotransmission. On one hand, disturbed metabolism of tryptophan and tyrosine limited the participation of the monoamine neurotransmitters serotonin and dopamine in synaptic vesicle recycling. On the other hand, dysregulated metabolism of the membrane lipids sphingomyelin and glycerophospholipids altered the synaptic membrane structure and neurotransmitter receptors (chrnα7, htr1b, drd5b, and gabra1) activity. Our findings revealed that disturbance of serotonergic and dopaminergic synaptic transmission mediated by the JNK inflammatory cascade was the key pathogenic mechanism in a zebrafish model of BD, provides critical biological insights into the pathogenesis of BD.
Collapse
Affiliation(s)
- Yameng Li
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Lin Zhang
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Mingcai Mao
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Linjuan He
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Tiancai Wang
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yecan Pan
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiaoyu Zhao
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zishu Li
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiyan Mu
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yongzhong Qian
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Corresponding author
| | - Jing Qiu
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Corresponding author
| |
Collapse
|
10
|
Lacombe RJS, Smith ME, Perlman K, Turecki G, Mechawar N, Bazinet RP. Quantitative and carbon isotope ratio analysis of fatty acids isolated from human brain hemispheres. J Neurochem 2023; 164:44-56. [PMID: 36196762 DOI: 10.1111/jnc.15702] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 02/04/2023]
Abstract
Our knowledge surrounding the overall fatty acid profile of the adult human brain has been largely limited to extrapolations from brain regions in which the distribution of fatty acids varies. This is especially problematic when modeling brain fatty acid metabolism, therefore, an updated estimate of whole-brain fatty acid concentration is necessitated. Here, we sought to conduct a comprehensive quantitative analysis of fatty acids from entire well-characterized human brain hemispheres (n = 6) provided by the Douglas-Bell Canada Brain Bank. Additionally, exploratory natural abundance carbon isotope ratio (CIR; δ13 C, 13 C/12 C) analysis was performed to assess the origin of brain fatty acids. Brain fatty acid methyl esters (FAMEs) were quantified by gas chromatography (GC)-flame ionization detection and minor n-6 and n-3 polyunsaturated fatty acid pentafluorobenzyl esters by GC-mass spectrometry. Carbon isotope ratio values of identifiable FAMEs were measured by GC-combustion-isotope ratio mass spectrometry. Overall, the most abundant fatty acid in the human brain was oleic acid, followed by stearic acid (STA), palmitic acid (PAM), docosahexaenoic acid (DHA), and arachidonic acid (ARA). Interestingly, cholesterol as well as saturates including PAM and STA were most enriched in 13 C, while PUFAs including DHA and ARA were most depleted in 13 C. These findings suggest a contribution of endogenous synthesis utilizing dietary sugar substrates rich in 13 C, and a combination of marine, animal, and terrestrial PUFA sources more depleted in 13 C, respectively. These results provide novel insights on cerebral fatty acid origin and concentration, the latter serving as a valuable resource for future modeling of fatty acid metabolism in the human brain.
Collapse
Affiliation(s)
- R J Scott Lacombe
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Mackenzie E Smith
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Kelly Perlman
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Zivko C, Sagar R, Xydia A, Mahairaki V. Lipid Profiling in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1423:281-287. [PMID: 37525056 DOI: 10.1007/978-3-031-31978-5_29] [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: 08/02/2023]
Abstract
The human brain is the organ with the most lipids after adipose tissues. The rich heterogeneity of the neural lipidome is being actively investigated with the aim of shedding new light into the physiological and pathological roles these compounds play in the brain. This is particularly important for the study of increasingly common neurodegenerative pathologies, such as Alzheimer's disease (AD), whose underlying mechanisms are still insufficiently understood and for which there is no cure. The present text dives into the current knowledge of the lipid composition of the brain, with a particular focus on the application of lipid profiling to AD research.
Collapse
Affiliation(s)
- Cristina Zivko
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Ram Sagar
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Ariadni Xydia
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Agrawal I, Lim YS, Ng SY, Ling SC. Deciphering lipid dysregulation in ALS: from mechanisms to translational medicine. Transl Neurodegener 2022; 11:48. [DOI: 10.1186/s40035-022-00322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractLipids, defined by low solubility in water and high solubility in nonpolar solvents, can be classified into fatty acids, glycerolipids, glycerophospholipids, sphingolipids, and sterols. Lipids not only regulate integrity and fluidity of biological membranes, but also serve as energy storage and bioactive molecules for signaling. Causal mutations in SPTLC1 (serine palmitoyltransferase long chain subunit 1) gene within the lipogenic pathway have been identified in amyotrophic lateral sclerosis (ALS), a paralytic and fatal motor neuron disease. Furthermore, lipid dysmetabolism within the central nervous system and circulation is associated with ALS. Here, we aim to delineate the diverse roles of different lipid classes and understand how lipid dysmetabolism may contribute to ALS pathogenesis. Among the different lipids, accumulation of ceramides, arachidonic acid, and lysophosphatidylcholine is commonly emerging as detrimental to motor neurons. We end with exploring the potential ALS therapeutics by reducing these toxic lipids.
Collapse
|
13
|
Reid MM, Obenaus A, Mukherjee PK, Khoutorova L, Roque CR, Petasis NA, Oria RB, Belayev L, Bazan NG. Synergistic Neuroprotection by a PAF Antagonist Plus a Docosanoid in Experimental Ischemic Stroke: Dose-Response and Therapeutic Window. J Stroke Cerebrovasc Dis 2022; 31:106585. [PMID: 35717719 PMCID: PMC9976619 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/17/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVE We tested the hypothesis that blocking pro-inflammatory platelet-activating factor receptor (PAFR) with LAU-0901 (LAU) plus administering a selected docosanoid, aspirin-triggered neuroprotectin D1 (AT-NPD1), which activates cell-survival pathways after middle cerebral artery occlusion (MCAo), would lead to neurological recovery. Dose-response and therapeutic window were investigated. MATERIALS AND METHODS Male SD rats were subjected to 2 hours of MCAo. Behavior testing (days 1-7) and ex vivo MRI on day 7 were conducted. In dose-response, rats were treated with LAU (45 and 60 mg/kg; IP), AT-NPD1 (111, 222, 333 µg/kg; IV), LAU+AT-NPD1 (LAU at 3 hours and AT-NPD1 at 3.15 hours) or vehicle. In the therapeutic window, vehicle, LAU (60 mg/kg), AT-NPD1 (222 µg/kg), and LAU+AT-NPD1 were administered at 3, 4, 5, and 6 hours after onset of MCAo. RESULTS LAU and AT-NPD1 treatments alone improved behavior by 40-42% and 20-30%, respectively, and LAU+AT-NPD1 by 40% compared to the vehicle group. T2-weighted imaging (T2WI) volumes were reduced with all doses of LAU and AT-NPD1 by 73-90% and 67-83% and LAU+AT-NPD1 by 94% compared to vehicle. In the therapeutic window, LAU+AT-NPD1, when administered at 3, 4, 5, and 6 hours, improved behavior by 50, 56, 33, and 26% and reduced T2WI volumes by 93, 90, 82, and 84% compared to vehicle. CONCLUSIONS We have shown here for the first time that LAU plus AT-NPD1 treatment affords high-grade neuroprotection in MCAo, equaling or exceeding that afforded by LAU or AT-NPD1 alone at considerably moderate doses. It has a broad therapeutic window extending to 6 hours after stroke onset.
Collapse
Affiliation(s)
- Madigan M. Reid
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, CA, USA.
| | - Pranab K. Mukherjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA.
| | - Cassia R. Roque
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - Nicos A. Petasis
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Reinaldo B. Oria
- Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - Ludmila Belayev
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA.
| | - Nicolas G. Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| |
Collapse
|
14
|
Urso C, Zhou H. Palmitic acid-induced defects in cell cycle progression and cytokinesis in Neuro-2a cells. Cell Cycle 2022; 21:1048-1057. [PMID: 35171079 PMCID: PMC9037450 DOI: 10.1080/15384101.2022.2040769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Obesity is associated with elevated levels of free fatty acids (FFAs). Excessive saturated fatty acids (SFAs) exhibit significant deleterious cytotoxic effects in many types of cells. However, the effects of palmitic acid (PA), the most common circulating SFA, on cell cycle progression in neuronal cells have not been well-examined. The aim of this study was to examine whether PA affects the proliferation and cell cycle progression in mouse neuroblastoma Neuro-2a (N2a) cells. Our studies found that 200 µM PA significantly decreased DNA synthesis and mitotic index in N2a cells as early as 4 h following treatment. 24 h treatment with 200 µM PA significantly decreased the percentage of diploid (2 N) cells while dramatically increasing the percentage of tetraploid (4 N) cells as compared to the BSA control. Moreover, our studies found that 24 h treatment with 200 µM PA increased the percentage of binucleate cells as compared to the BSA control. Our studies also found that unsaturated fatty acids (UFAs), including linoleic acid, oleic acid, α-linolenic acid, and docosahexaenoic acid, were able to abolish PA-induced decrease of 2 N cells, increase of 4 N cells, and accumulation of binucleate cells. Taken together, these results suggest that PA may affect multiple aspects of the cell cycle progression in N2a cells, including decreased DNA synthesis, G2/M arrest, and cytokinetic failure, which could be abolished by UFAs.Abbreviations: 4-PBA, 4-Phenylbutyric Acid; ALA, α-linolenic acid; BrdU, 5-bromo-2'-deoxyuridine; DAPI, 4',6-diamidino-2-phenylindole; ER, endoplasmic reticulum; FFA, free fatty acids; FITC, fluorescein isothiocyanate; LA, linoleic acid; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; N2a, Neuro-2a; NAC, N-acetyl cysteine; OA, oleic acid; PA, palmitic acid; pHH3, Phosphorylation of histone H3; PI, propidium iodide; SFA, saturated fatty acids; PUFA, polyunsaturated fatty acids; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; UFA, unsaturated fatty acids.
Collapse
Affiliation(s)
- C.J. Urso
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA
| | - Heping Zhou
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA,CONTACT Heping Zhou Department of Biological Sciences, Seton Hall University, South Orange, NJ07079, USA
| |
Collapse
|
15
|
Cai D, Luo Z, Su J, Gan H, Wang Z, Liu X, Li S, Wu J, Chen J, Ma R, Huang M, Zhong G. Exposure-Response Analysis and Mechanism of Ginkgolide B’s Neuroprotective Effect in Acute Cerebral Ischemia/Reperfusion Stage in Rat. Biol Pharm Bull 2022; 45:409-420. [DOI: 10.1248/bpb.b21-00781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Dake Cai
- School of Pharmaceutical Science, Sun Yat-sen University
| | - Zhongxing Luo
- The Seventh Affiliated Hospital, Sun Yat-sen University
| | - Jiyan Su
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University
| | - Haining Gan
- Department of Pharmacology of Traditional Chinese Medicine, The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine
| | | | - Xiaolin Liu
- School of Pharmaceutical Science, Sun Yat-sen University
| | - Siyi Li
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine
| | - Jingjing Wu
- Department of Pharmacy, the First Affiliated Hospital, Sun Yat-sen University
| | - Jiangying Chen
- School of Pharmaceutical Science, Sun Yat-sen University
| | - Renqiang Ma
- Guangzhou Boji Medical Biotechnological Co., Ltd
| | - Min Huang
- School of Pharmaceutical Science, Sun Yat-sen University
| | - Guoping Zhong
- School of Pharmaceutical Science, Sun Yat-sen University
| |
Collapse
|
16
|
Zamarbide M, Martinez-Pinilla E, Gil-Bea F, Yanagisawa M, Franco R, Perez-Mediavilla A. Genetic Inactivation of Free Fatty Acid Receptor 3 Impedes Behavioral Deficits and Pathological Hallmarks in the APP swe Alzheimer's Disease Mouse Model. Int J Mol Sci 2022; 23:ijms23073533. [PMID: 35408893 PMCID: PMC8999053 DOI: 10.3390/ijms23073533] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/01/2023] Open
Abstract
The free fatty acid FFA3 receptor (FFA3R) belongs to the superfamily of G-protein-coupled receptors (GPCRs). In the intestine and adipose tissue, it is involved in the regulation of energy metabolism, but its function in the brain is unknown. We aimed, first, to investigate the expression of the receptor in the hippocampus of Alzheimer disease (AD) patients at different stages of the disease and, second, to assess whether genetic inactivation of the Ffar3 gene could affect the phenotypic features of the APPswe mouse model. The expression of transcripts for FFA receptors in postmortem human hippocampal samples and in the hippocampus of wild-type and transgenic mice was analyzed by RT-qPCR. We generated a double transgenic mouse, FFA3R−/−/APPswe, to perform cognition studies and to assess, by immunoblotting Aβ and tau pathologies and the differential expression of synaptic plasticity-related proteins. For the first time, the occurrence of the FFA3R in the human hippocampus and its overexpression, even in the first stages of AD, was demonstrated. Remarkably, FFA3R−/−/APPswe mice do not have the characteristic memory impairment of 12-month-old APPswe mice. Additionally, this newly generated transgenic line does not develop the most important Alzheimer’s disease (AD)-related features, such as amyloid beta (Aβ) brain accumulations and tau hyperphosphorylation. These findings are accompanied by increased levels of the insulin-degrading enzyme (IDE) and lower activity of the tau kinases GSK3β and Cdk5. We conclude that the brain FFA3R is involved in cognitive processes and that its inactivation prevents AD-like cognitive decline and pathological hallmarks.
Collapse
Affiliation(s)
- Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
| | - Eva Martinez-Pinilla
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Francisco Gil-Bea
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan;
| | - Rafael Franco
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Network Center, Neurodegenerative Diseases, CiberNed, Spanish National Health Institute “Carlos III”, 28031 Madrid, Spain
- Correspondence: (R.F.); (A.P.-M.); Tel.: +34-934021208 (R.F.); +34-948194700 (ext. 2033) (A.P.-M.)
| | - Alberto Perez-Mediavilla
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain
- Correspondence: (R.F.); (A.P.-M.); Tel.: +34-934021208 (R.F.); +34-948194700 (ext. 2033) (A.P.-M.)
| |
Collapse
|
17
|
Drosophila Lysophospholipase Gene swiss cheese Is Required for Survival and Reproduction. INSECTS 2021; 13:insects13010014. [PMID: 35055857 PMCID: PMC8781823 DOI: 10.3390/insects13010014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/28/2022]
Abstract
Simple Summary Biological evolution implies fitness of newly evolved organisms that have inherent adaptive traits because of mutations in genes. However, most mutations are detrimental, and they spoil the organism’s life, its survival and its ability to leave progeny. Some genes are extremely vital for an organism, and therefore, they tend to save their structure and do not mutate or do it very composedly. That is the case of the gene encoding PNPLA6 lysophospholipase domain that evolved in bacteria, and evolution obliged it to save its function in higher animals. In mammals, complete dysfunction of such a gene is lethal because of its high importance in placenta for early embryo development. Why is it conserved in other species, for instance insects, that have no placenta? Here we studied the role of the PNPLA6-encoding gene named swiss cheese in Drosophila melanogaster fitness. We have found that its dysfunction results in premature death of specimens and their inability to leave enough progeny. Thus, we provide the first evidence for significance of the gene that encodes the lysophospholipase enzyme in fitness of insects. Abstract Drosophila melanogaster is one of the most famous insects in biological research. It is widely used to analyse functions of different genes. The phosphatidylcholine lysophospholipase gene swiss cheese was initially shown to be important in the fruit fly nervous system. However, the role of this gene in non-nervous cell types has not been elucidated yet, and the evolutional explanation for the conservation of its function remains elusive. In this study, we analyse expression pattern and some aspects of the role of the swiss cheese gene in the fitness of Drosophila melanogaster. We describe the spatiotemporal expression of swiss cheese throughout the fly development and analyse the survival and productivity of swiss cheese mutants. We found swiss cheese to be expressed in salivary glands, midgut, Malpighian tubes, adipocytes, and male reproductive system. Dysfunction of swiss cheese results in severe pupae and imago lethality and decline of fertility, which is impressive in males. The latter is accompanied with abnormalities of male locomotor activity and courtship behaviour, accumulation of lipid droplets in testis cyst cells and decrease in spermatozoa motility. These results suggest that normal swiss cheese is important for Drosophila melanogaster fitness due to its necessity for both specimen survival and their reproductive success.
Collapse
|
18
|
Dominguez LJ, Veronese N, Vernuccio L, Catanese G, Inzerillo F, Salemi G, Barbagallo M. Nutrition, Physical Activity, and Other Lifestyle Factors in the Prevention of Cognitive Decline and Dementia. Nutrients 2021; 13:nu13114080. [PMID: 34836334 PMCID: PMC8624903 DOI: 10.3390/nu13114080] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Multiple factors combined are currently recognized as contributors to cognitive decline. The main independent risk factor for cognitive impairment and dementia is advanced age followed by other determinants such as genetic, socioeconomic, and environmental factors, including nutrition and physical activity. In the next decades, a rise in dementia cases is expected due largely to the aging of the world population. There are no hitherto effective pharmaceutical therapies to treat age-associated cognitive impairment and dementia, which underscores the crucial role of prevention. A relationship among diet, physical activity, and other lifestyle factors with cognitive function has been intensively studied with mounting evidence supporting the role of these determinants in the development of cognitive decline and dementia, which is a chief cause of disability globally. Several dietary patterns, foods, and nutrients have been investigated in this regard, with some encouraging and other disappointing results. This review presents the current evidence for the effects of dietary patterns, dietary components, some supplements, physical activity, sleep patterns, and social engagement on the prevention or delay of the onset of age-related cognitive decline and dementia.
Collapse
Affiliation(s)
- Ligia J. Dominguez
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy
- Correspondence: ; +39-0916554828
| | - Nicola Veronese
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Laura Vernuccio
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Giuseppina Catanese
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Flora Inzerillo
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| | - Giuseppe Salemi
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90100 Palermo, Italy;
- UOC of Neurology, University Hospital “Paolo Giaccone”, 90100 Palermo, Italy
| | - Mario Barbagallo
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy; (N.V.); (L.V.); (G.C.); (F.I.); (M.B.)
| |
Collapse
|
19
|
Implications of Phosphoinositide 3-Kinase-Akt (PI3K-Akt) Pathway in the Pathogenesis of Alzheimer's Disease. Mol Neurobiol 2021; 59:354-385. [PMID: 34699027 DOI: 10.1007/s12035-021-02611-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the foremost type of dementia that afflicts considerable morbidity and mortality in aged population. Several transcription molecules, pathways, and molecular mechanisms such as oxidative stress, inflammation, autophagy, and immune system interact in a multifaceted way that disrupt physiological processes (cell growth, differentiation, survival, lipid and energy metabolism, endocytosis) leading to apoptosis, tauopathy, β-amyloidopathy, neuron, and synapse loss, which play an important role in AD pathophysiology. Despite of stupendous advancements in pathogenic mechanisms, treatment of AD is still a nightmare in the field of medicine. There is compelling urgency to find not only symptomatic but effective disease-modifying therapies. Recently, phosphoinositide 3-kinase (PI3K) and Akt are identified as a pathway triggered by diverse stimuli, including insulin, growth factors, cytokines, and cellular stress, that link amyloid-β, neurofibrillary tangles, and brain atrophy. The present review aims to explore and analyze the role of PI3K-Akt pathway in AD and agents which may modulate Akt and have therapeutic prospects in AD. The literature was researched using keywords "PI3K-Akt" and "Alzheimer's disease" from PubMed, Web of Science, Bentham, Science Direct, Springer Nature, Scopus, and Google Scholar databases including books. Articles published from 1992 to 2021 were prioritized and analyzed for their strengths and limitations, and most appropriate ones were selected for the purpose of review. PI3K-Akt pathway regulates various biological processes such as cell proliferation, motility, growth, survival, and metabolic functions, and inhibits many neurotoxic mechanisms. Furthermore, experimental data indicate that PI3K-Akt signaling might be an important therapeutic target in treatment of AD.
Collapse
|
20
|
Cruz Flores VA, Menghani H, Mukherjee PK, Marrero L, Obenaus A, Dang Q, Khoutorova L, Reid MM, Belayev L, Bazan NG. Combined Therapy With Avastin, a PAF Receptor Antagonist and a Lipid Mediator Inhibited Glioblastoma Tumor Growth. Front Pharmacol 2021; 12:746470. [PMID: 34630114 PMCID: PMC8498947 DOI: 10.3389/fphar.2021.746470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/06/2021] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive, highly proliferative, invasive brain tumor with a poor prognosis and low survival rate. The current standard of care for GBM is chemotherapy combined with radiation following surgical intervention, altogether with limited efficacy, since survival averages 18 months. Improvement in treatment outcomes for patients with GBM requires a multifaceted approach due to the dysregulation of numerous signaling pathways. Recently emerging therapies to precisely modulate tumor angiogenesis, inflammation, and oxidative stress are gaining attention as potential options to combat GBM. Using a mouse model of GBM, this study aims to investigate Avastin (suppressor of vascular endothelial growth factor and anti-angiogenetic treatment), LAU-0901 (a platelet-activating factor receptor antagonist that blocks pro-inflammatory signaling), Elovanoid; ELV, a novel pro-homeostatic lipid mediator that protects neural cell integrity and their combination as an alternative treatment for GBM. Female athymic nude mice were anesthetized with ketamine/xylazine, and luciferase-modified U87MG tumor cells were stereotactically injected into the right striatum. On post-implantation day 13, mice received one of the following: LAU-0901, ELV, Avastin, and all three compounds in combination. Bioluminescent imaging (BLI) was performed on days 13, 20, and 30 post-implantation. Mice were perfused for ex vivo MRI on day 30. Bioluminescent intracranial tumor growth percentage was reduced by treatments with LAU-0901 (43%), Avastin (77%), or ELV (86%), individually, by day 30 compared to saline treatment. In combination, LAU-0901/Avastin, ELV/LAU-0901, or ELV/Avastin had a synergistic effect in decreasing tumor growth by 72, 92, and 96%, respectively. Additionally, tumor reduction was confirmed by MRI on day 30, which shows a decrease in tumor volume by treatments with LAU-0901 (37%), Avastin (67%), or ELV (81.5%), individually, by day 30 compared to saline treatment. In combination, LAU-0901/Avastin, ELV/LAU-0901, or ELV/Avastin had a synergistic effect in decreasing tumor growth by 69, 78.7, and 88.6%, respectively. We concluded that LAU-0901 and ELV combined with Avastin exert a better inhibitive effect in GBM progression than monotherapy. To our knowledge, this is the first study that demonstrates the efficacy of these novel therapeutic regimens in a model of GBM and may provide the basis for future therapeutics in GBM patients.
Collapse
Affiliation(s)
- Valerie A Cruz Flores
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Pediatrics, Hematology-Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Hemant Menghani
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Pediatrics, Hematology-Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Pranab K Mukherjee
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Luis Marrero
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California Irvin, Irvine, CA, United States
| | - Quan Dang
- Department of Pediatrics, School of Medicine, University of California Irvin, Irvine, CA, United States
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Madigan M Reid
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Ludmila Belayev
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
21
|
Garcia-Lopez R, Pombero A, Estirado A, Geijo-Barrientos E, Martinez S. Interneuron Heterotopia in the Lis1 Mutant Mouse Cortex Underlies a Structural and Functional Schizophrenia-Like Phenotype. Front Cell Dev Biol 2021; 9:693919. [PMID: 34327202 PMCID: PMC8313859 DOI: 10.3389/fcell.2021.693919] [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: 04/12/2021] [Accepted: 06/16/2021] [Indexed: 11/24/2022] Open
Abstract
LIS1 is one of the principal genes related to Type I lissencephaly, a severe human brain malformation characterized by an abnormal neuronal migration in the cortex during embryonic development. This is clinically associated with epilepsy and cerebral palsy in severe cases, as well as a predisposition to developing mental disorders, in cases with a mild phenotype. Although genetic variations in the LIS1 gene have been associated with the development of schizophrenia, little is known about the underlying neurobiological mechanisms. We have studied how the Lis1 gene might cause deficits associated with the pathophysiology of schizophrenia using the Lis1/sLis1 murine model, which involves the deletion of the first coding exon of the Lis1 gene. Homozygous mice are not viable, but heterozygous animals present abnormal neuronal morphology, cortical dysplasia, and enhanced cortical excitability. We have observed reduced number of cells expressing GABA-synthesizing enzyme glutamic acid decarboxylase 67 (GAD67) in the hippocampus and the anterior cingulate area, as well as fewer parvalbumin-expressing cells in the anterior cingulate cortex in Lis1/sLis1 mutants compared to control mice. The cFOS protein expression (indicative of neuronal activity) in Lis1/sLis1 mice was higher in the medial prefrontal (mPFC), perirhinal (PERI), entorhinal (ENT), ectorhinal (ECT) cortices, and hippocampus compared to control mice. Our results suggest that deleting the first coding exon of the Lis1 gene might cause cortical anomalies associated with the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
| | - Ana Pombero
- Instituto de Neurociencias, UMH-CSIC, Alicante, Spain
| | | | | | - Salvador Martinez
- Instituto de Neurociencias, UMH-CSIC, Alicante, Spain.,Centro de Investigación Biomédica En Red en Salud Mental-CIBERSAM-ISCIII, Valencia, Spain
| |
Collapse
|
22
|
Tian R, Abarientos A, Hong J, Hashemi SH, Yan R, Dräger N, Leng K, Nalls MA, Singleton AB, Xu K, Faghri F, Kampmann M. Genome-wide CRISPRi/a screens in human neurons link lysosomal failure to ferroptosis. Nat Neurosci 2021; 24:1020-1034. [PMID: 34031600 PMCID: PMC8254803 DOI: 10.1038/s41593-021-00862-0] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 04/23/2021] [Indexed: 02/08/2023]
Abstract
Single-cell transcriptomics provide a systematic map of gene expression in different human cell types. The next challenge is to systematically understand cell-type-specific gene function. The integration of CRISPR-based functional genomics and stem cell technology enables the scalable interrogation of gene function in differentiated human cells. Here we present the first genome-wide CRISPR interference and CRISPR activation screens in human neurons. We uncover pathways controlling neuronal response to chronic oxidative stress, which is implicated in neurodegenerative diseases. Unexpectedly, knockdown of the lysosomal protein prosaposin strongly sensitizes neurons, but not other cell types, to oxidative stress by triggering the formation of lipofuscin, a hallmark of aging, which traps iron, generating reactive oxygen species and triggering ferroptosis. We also determine transcriptomic changes in neurons after perturbation of genes linked to neurodegenerative diseases. To enable the systematic comparison of gene function across different human cell types, we establish a data commons named CRISPRbrain.
Collapse
Affiliation(s)
- Ruilin Tian
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Biophysics Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Anthony Abarientos
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Jason Hong
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Sayed Hadi Hashemi
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rui Yan
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - Nina Dräger
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Kun Leng
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, LLC, Glen Echo, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - Faraz Faghri
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, LLC, Glen Echo, MD, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
23
|
Lindhout IA, Murray TE, Richards CM, Klegeris A. Potential neurotoxic activity of diverse molecules released by microglia. Neurochem Int 2021; 148:105117. [PMID: 34186114 DOI: 10.1016/j.neuint.2021.105117] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/18/2021] [Accepted: 06/24/2021] [Indexed: 01/02/2023]
Abstract
Microglia are the professional immune cells of the brain, which support numerous physiological processes. One of the defensive functions provided by microglia involves secretion of cytotoxins aimed at destroying invading pathogens. It is also recognized that the adverse activation of microglia in diseased brains may lead to secretion of cytotoxic molecules, which could be damaging to the surrounding cells, including neurons. Several of these toxins, such as reactive oxygen and nitrogen species, L-glutamate, and quinolinic acid, are widely recognized and well-studied. This review is focused on a structurally diverse group of less-established microglia neurotoxins, which were selected by applying the two criteria that these molecules 1) can be released by microglia, and 2) have the potential to be directly harmful to neurons. The following 11 molecules are discussed in detail: amyloid beta peptides (Aβ); cathepsin (Cat)B and CatD; C-X-C motif chemokine ligand (CXCL)10 and CXCL12 (5-67); high mobility group box (HMGB)1; lymphotoxin (LT)-α; matrix metalloproteinase (MMP)-2 and MMP-9; platelet-activating factor (PAF); and prolyl endopeptidase (PEP). Molecular mechanisms of their release by microglia and neurotoxicity, as well as available evidence implicating their involvement in human neuropathologies are summarized. Further studies on several of the above molecules are warranted to confirm either their microglial origin in the brain or direct neurotoxic effects. In addition, investigations into the differential secretion patterns of neurotoxins by microglia in response to diverse stimuli are required. This research could identify novel therapeutic targets for neurological disorders involving adverse microglial activation.
Collapse
Affiliation(s)
- Ivan A Lindhout
- Department of Biology, University of British Columbia Okanagan Campus, 3187 University Way, Kelowna, British Columbia, V1V 1V7, Canada
| | - Taryn E Murray
- Department of Biology, University of British Columbia Okanagan Campus, 3187 University Way, Kelowna, British Columbia, V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, 3187 University Way, Kelowna, British Columbia, V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, 3187 University Way, Kelowna, British Columbia, V1V 1V7, Canada.
| |
Collapse
|
24
|
SREBP-1c Deficiency Affects Hippocampal Micromorphometry and Hippocampus-Dependent Memory Ability in Mice. Int J Mol Sci 2021; 22:ijms22116103. [PMID: 34198910 PMCID: PMC8201143 DOI: 10.3390/ijms22116103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 11/18/2022] Open
Abstract
Changes in structural and functional neuroplasticity have been implicated in various neurological disorders. Sterol regulatory element-binding protein (SREBP)-1c is a critical regulatory molecule of lipid homeostasis in the brain. Recently, our findings have shown the potential involvement of SREBP-1c deficiency in the alteration of novel modulatory molecules in the hippocampus and occurrence of schizophrenia-like behaviors in mice. However, the possible underlying mechanisms, related to neuronal plasticity in the hippocampus, are yet to be elucidated. In this study, we investigated the hippocampus-dependent memory function and neuronal architecture of hippocampal neurons in SREBP-1c knockout (KO) mice. During the passive avoidance test, SREBP-1c KO mice showed memory impairment. Based on Golgi staining, the dendritic complexity, length, and branch points were significantly decreased in the apical cornu ammonis (CA) 1, CA3, and dentate gyrus (DG) subregions of the hippocampi of SREBP-1c KO mice, compared with those of wild-type (WT) mice. Additionally, significant decreases in the dendritic diameters were detected in the CA3 and DG subregions, and spine density was also significantly decreased in the apical CA3 subregion of the hippocampi of KO mice, compared with that of WT mice. Alterations in the proportions of stubby and thin-shaped dendritic spines were observed in the apical subcompartments of CA1 and CA3 in the hippocampi of KO mice. Furthermore, the corresponding differential decreases in the levels of SREBP-1 expression in the hippocampal subregions (particularly, a significant decrease in the level in the CA3) were detected by immunofluorescence. This study suggests that the contributions of SREBP-1c to the structural plasticity of the mouse hippocampus may have underlain the behavioral alterations. These findings offer insights into the critical role of SREBP-1c in hippocampal functioning in mice.
Collapse
|
25
|
Huanglian Jiedu decoction remodels the periphery microenvironment to inhibit Alzheimer's disease progression based on the "brain-gut" axis through multiple integrated omics. ALZHEIMERS RESEARCH & THERAPY 2021; 13:44. [PMID: 33579351 PMCID: PMC7881564 DOI: 10.1186/s13195-021-00779-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND In recent years, excellent results have suggested an association between the "brain-gut" axis and Alzheimer's disease (AD) progression, yet the role of the "brain-gut" axis in AD pathogenesis still remains obscure. Herein, we provided a potential link between the central and peripheral neuroinflammatory disorders in AD progression. METHODS The Morris water maze (MWM) test, immunohistochemistry, ELISA, ProcartaPlex Multiplex immunoassay, multiple LC-MS/MS methods, and the V3-V4 regions of 16S rRNA genes were applied to explore potential biomarkers. RESULTS In Tg-APP/PS1 mice, gut dysbiosis and lipid metabolism were highly associated with AD-like neuroinflammation. The combination of inflammatory factors (IL-6 and INF-γ), phosphatidylcholines (PCs) and SCFA-producing bacteria were expected to be early diagnostic biomarkers for AD. Huanglian Jiedu decoction (HLJDD) suppressed gut dysbiosis and the associated Aβ accumulation, harnessed neuroinflammation and reversed cognitive impairment. CONCLUSION Together, our findings highlighted the roles of neuroinflammation induced by gut dysbiosis and lipid metabolism disorder in AD progression. This integrated metabolomics approach showed its potential to understand the complex mechanisms of HLJDD in the treatment of AD.
Collapse
|
26
|
Blanc M, Alfonso S, Bégout ML, Barrachina C, Hyötyläinen T, Keiter SH, Cousin X. An environmentally relevant mixture of polychlorinated biphenyls (PCBs) and polybrominated diphenylethers (PBDEs) disrupts mitochondrial function, lipid metabolism and neurotransmission in the brain of exposed zebrafish and their unexposed F2 offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 754:142097. [PMID: 32911150 DOI: 10.1016/j.scitotenv.2020.142097] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/25/2020] [Accepted: 08/29/2020] [Indexed: 06/11/2023]
Abstract
Polychlorinated biphenyls (PCBs) and polybrominated diphenyl ethers (PBDEs) are persistent organic pollutants still present in aquatic environments despite their total or partial ban. Previously, we observed that an environmentally realistic mixture of these compounds affects energy balance, growth, and reproduction in exposed zebrafish (F0), and behavior in their unexposed offspring (F1-F4). In the present work, we performed lipidomic and transcriptomic analyses on brains of zebrafish (F0-F2) from exposed and control lineages to identify molecular changes that could explain the observed phenotypes. The use of both technologies highlighted that F0 zebrafish displayed impaired mitochondrial function and lipid metabolism regulation (depletion in triacylglycerols and phospholipids) which can explain disruption of energy homeostasis. A subset of the regulated biological pathways related to energetic metabolism and neurotransmission were inherited in F2. In addition, there were increasing effects on epigenetic pathways from the F0 to the F2 generation. Altogether, we show that the effects of an environmental exposure to PCBs and PBDEs on energetic metabolism as well as neurotransmission extend over 2 generations of zebrafish, possibly due to transgenerational epigenetic inheritance.
Collapse
Affiliation(s)
- Mélanie Blanc
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden.
| | - Sébastien Alfonso
- MARBEC, Univ. Montpellier, CNRS, Ifremer, IRD, Route de Maguelone, F-34250 Palavas-les-Flots, France; COISPA Tecnologia & Ricerca, Stazione Sperimentale per lo Studio delle Risorse del Mare, Via dei Trulli, n 18, 70126 Bari, Italy
| | - Marie-Laure Bégout
- MARBEC, Univ. Montpellier, CNRS, Ifremer, IRD, Route de Maguelone, F-34250 Palavas-les-Flots, France
| | - Célia Barrachina
- MGX, Univ. Montpellier, CNRS, INSERM, Université Montpellier 2, Place Eugène Bataillon, F-34095 Montpellier, France
| | - Tuulia Hyötyläinen
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| | - Steffen H Keiter
- Man-Technology-Environment Research Centre (MTM), School of Science and Technology, Örebro University, Fakultetsgatan 1, S-701 82 Örebro, Sweden
| | - Xavier Cousin
- MARBEC, Univ. Montpellier, CNRS, Ifremer, IRD, Route de Maguelone, F-34250 Palavas-les-Flots, France; Université Paris-Saclay, AgroParisTech, INRAE, GABI, Domaine de Vilvert, F-78350 Jouy-en-Josas, France
| |
Collapse
|
27
|
Belayev L, Obenaus A, Mukherjee PK, Knott EJ, Khoutorova L, Reid MM, Roque CR, Nguyen L, Lee JB, Petasis NA, Oria RB, Bazan NG. Blocking pro-inflammatory platelet-activating factor receptors and activating cell survival pathways: A novel therapeutic strategy in experimental ischemic stroke. Brain Circ 2021; 6:260-268. [PMID: 33506149 PMCID: PMC7821809 DOI: 10.4103/bc.bc_36_20] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/10/2020] [Accepted: 10/18/2020] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Acute ischemic stroke triggers complex neurovascular, neuroinflammatory, and synaptic alterations. This study explores whether blocking pro-inflammatory platelet-activating factor receptor (PAF-R) plus selected docosanoids after middle cerebral artery occlusion (MCAo) would lead to neurological recovery. The following small molecules were investigated: (a) LAU-0901, a PAF-R antagonist that blocks pro-inflammatory signaling; and (b) derivatives of docosahexaenoic acid (DHA), neuroprotectin D1 (NPD1), and aspirin-triggered NPD1 (AT-NPD1), which activates cell survival pathways and are exert potent anti-inflammatory activity in the brain. MATERIALS AND METHODS Sprague-Dawley rats received 2 h MCAo and LAU-0901 (30 or 60 mg/kg, 2 h after stroke), NPD1, and AT-NPD1 (333 μg/kg), DHA (5 mg/kg), and their combination were administered intravenous at 3 h after stroke. Behavior testing and ex vivo magnetic resonance imaging were conducted on day 3 or 14 to assess lesion characteristics and lipidomic analysis on day 1. Series 1 (LAU-0901 + NPD1, 14d), Series 2 (LAU-0901 + AT-NPD1, 3d), and Series 3 (LAU-0901 + DHA, 1d). RESULTS All combinatory groups improved behavior compared to NPD1, AT-NPD1, or DHA treatments alone. Total lesion volumes were reduced with LAU-0901 + NPD1 by 62% and LAU-0901 + AT-NPD1 by 90% treatments versus vehicle groups. LAU-0901 and LAU-0901 + DHA increased the production of vasoactive lipid mediators (prostaglandins: PGE2, PGF2- α, 6-keto-PGF1- α, and PGD2) as well an inflammatory regulating mediator hydroxyoctadecadienoic acid. In contrast, LAU-0901 and LAU-0901 + DHA decreased the production of 12-hydroxyeicosatetraenoic acid, a pro-inflammatory mediator. CONCLUSION Combination therapy with LAU-0901 and selected docosanoids is more effective than the single therapy, affording synergistic neuroprotection, with restored pro-homeostatic lipid mediators and improved neurological recovery. Altogether, our findings support the combinatory therapy as the basis for future therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Ludmila Belayev
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California, Irvine, USA
| | - Pranab K Mukherjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Eric J Knott
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Madigan M Reid
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Cassia R Roque
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA.,Department of Morphology and Institute of Biomedicine, School of Medicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Federal University of Ceara, Fortaleza, Brazil
| | - Lawrence Nguyen
- Department of Pediatrics, School of Medicine, University of California, Irvine, USA
| | - Jeong Bin Lee
- Department of Pediatrics, School of Medicine, University of California, Irvine, USA
| | - Nicos A Petasis
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Reinaldo B Oria
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA.,Department of Morphology and Institute of Biomedicine, School of Medicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, Federal University of Ceara, Fortaleza, Brazil
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
28
|
The interaction between brain and liver regulates lipid metabolism in the TBI pathology. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166078. [PMID: 33444711 DOI: 10.1016/j.bbadis.2021.166078] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/28/2020] [Accepted: 01/03/2021] [Indexed: 12/31/2022]
Abstract
To shed light on the impact of systemic physiology on the pathology of traumatic brain injury (TBI), we examine the effects of TBI (concussive injury) and dietary fructose on critical aspects of lipid homeostasis in the brain and liver of young-adult rats. Lipids are integral components of brain structure and function, and the liver has a role on the synthesis and metabolism of lipids. Fructose is mainly metabolized in the liver with potential implications for brain function. Lipidomic analysis accompanied by unbiased sparse partial least squares discriminant analysis (sPLS-DA) identified lysophosphatidylcholine (LPC) and cholesterol ester (CE) as the top lipid families impacted by TBI and fructose in the hippocampus, and only LPC (16:0) was associated with hippocampal-dependent memory performance. Fructose and TBI elevated liver pro-inflammatory markers, interleukin-1α (IL-1α), Interferon-γ (IFN-γ) that correlated with hippocampal-dependent memory dysfunction, and monocyte chemoattractant protein-1 (MCP-1) positively correlated with LPC levels in the hippocampus. The effects of fructose were more pronounced in the liver, in agreement with the role of liver on fructose metabolism and suggest that fructose could exacerbate liver inflammation caused by TBI. The overall results indicate that TBI and fructose interact to influence systemic and central inflammation by engaging liver lipids. The impact of TBI and fructose diet on the periphery provides a therapeutic target to counteract the TBI pathogenesis.
Collapse
|
29
|
da Silva Chagas L, Sandre PC, de Velasco PC, Marcondes H, Ribeiro E Ribeiro NCA, Barreto AL, Alves Mauro LB, Ferreira JH, Serfaty CA. Neuroinflammation and Brain Development: Possible Risk Factors in COVID-19-Infected Children. Neuroimmunomodulation 2021; 28:22-28. [PMID: 33530091 PMCID: PMC7900470 DOI: 10.1159/000512815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/01/2020] [Indexed: 11/19/2022] Open
Abstract
COVID-19, a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) betacoronavirus, affects children in a different way than it does in adults, with milder symptoms. However, several cases of neurological symptoms with neuroinflammatory syndromes, such as the multisystem inflammatory syndrome (MIS-C), following mild cases, have been reported. As with other viral infections, such as rubella, influenza, and cytomegalovirus, SARS-CoV-2 induces a surge of proinflammatory cytokines that affect microglial function, which can be harmful to brain development. Along with the viral induction of neuroinflammation, other noninfectious conditions may interact to produce additional inflammation, such as the nutritional imbalance of fatty acids and polyunsaturated fatty acids and alcohol consumption during pregnancy. Additionally, transient thyrotoxicosis induced by SARS-CoV-2 with secondary autoimmune hypothyroidism has been reported, which could go undetected during pregnancy. Together, those factors may pose additional risk factors for SARS-CoV-2 infection impacting mechanisms of neural development such as synaptic pruning and neural circuitry formation. The present review discusses those conditions in the perspective of the understanding of risk factors that should be considered and the possible emergence of neurodevelopmental disorders in COVID-19-infected children.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Poliana Capucho Sandre
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Patricia Coelho de Velasco
- Department of Applied Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Henrique Marcondes
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | | | - Aline Loureiro Barreto
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Luiza Beatriz Alves Mauro
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Julia Huber Ferreira
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Claudio A Serfaty
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil,
- National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil,
| |
Collapse
|
30
|
Chi NF, Chang TH, Lee CY, Wu YW, Shen TA, Chan L, Chen YR, Chiou HY, Hsu CY, Hu CJ. Untargeted metabolomics predicts the functional outcome of ischemic stroke. J Formos Med Assoc 2020; 120:234-241. [PMID: 32414667 DOI: 10.1016/j.jfma.2020.04.026] [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] [Received: 11/15/2019] [Revised: 03/08/2020] [Accepted: 04/20/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND/PURPOSE Metabolites in blood have been found associated with the occurrence of vascular diseases, but its role in the functional recovery of stroke is unclear. The aim of this study is to investigate whether the untargeted metabolomics at the acute stage of ischemic stroke is able to predict functional recovery. METHODS One hundred and fifty patients with acute ischemic stroke were recruited and followed up for 3 months. Fasting blood samples within 7 days of stroke were obtained, liquid chromatography and mass spectrometry were applied to identify outcome-associated metabolites. The patients' clinical characteristics and identified metabolites were included for constructing the outcome prediction model using machine learning approaches. RESULTS By using multivariate analysis, 220 differentially expressed metabolites (DEMs) were discovered between patients with favorable outcomes (modified Rankin Scale, mRS ≤ 2 at 3 months, n = 77) and unfavorable outcomes (mRS ≥ 3 at 3 months, n = 73). After feature selection, 63 DEMs were chosen for constructing the outcome prediction model. The predictive accuracy was below 0.65 when including patients' clinical characteristics, and could reach 0.80 when including patients' clinical characteristics and 63 selected DEMs. The functional enrichment analysis identified platelet activating factor (PAF) as the strongest outcome-associated metabolite, which involved in proinflammatory mediators release, arachidonic acid metabolism, eosinophil degranulation, and production of reactive oxygen species. CONCLUSION Metabolomics is a potential method to explore the blood biomarkers of acute ischemic stroke. The patients with unfavorable outcomes had a lower PAF level compared to those with favorable outcomes.
Collapse
Affiliation(s)
- Nai-Fang Chi
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Neurology, Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan; Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chen-Yang Lee
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wei Wu
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| | - Ting-An Shen
- Graduate Institute of Biomedical Informatics, Taipei Medical University, Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yih-Ru Chen
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Chung Y Hsu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
31
|
Barón-Mendoza I, González-Arenas A. Relationship between the effect of polyunsaturated fatty acids (PUFAs) on brain plasticity and the improvement on cognition and behavior in individuals with autism spectrum disorder. Nutr Neurosci 2020; 25:387-410. [PMID: 32338174 DOI: 10.1080/1028415x.2020.1755793] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: This work aimed to compile information about the neuronal processes in which polyunsaturated fatty acids (PUFAs) could modulate brain plasticity, in order to analyze the role of nutritional intervention with the ω-3 and ω-6 fatty acids as a therapeutic strategy for the Autism Spectrum Disorder (ASD)-related signs and symptoms.Methods: We reviewed different articles reporting the effect of PUFAS on neurite elongation, membrane expansion, cytoskeleton rearrangement and neurotransmission, considering the ASD-related abnormalities in these processes.Results: In accordance to the reviewed studies, it is clear that ASD is one of the neurological conditions associated with an impairment in neuronal plasticity; therefore, PUFAs-rich diet improvements on cognition and behavioral deficits in individuals with autism, could be involved with the regulation of neuronal processes implicated in the atypical brain plasticity related with this neurodevelopmental disorder.Discussion: The behavioral and cognitive improvement observed in individuals with ASD after PUFAs treatment might underlie, at least in part, in the ability of ω-3 and ω-6 fatty acids to induce neurite outgrowth, probably, through the dynamic regulation of the neuronal cytoskeleton along with the expansion of neuronal membranes. Furthermore, it might also be associated with an enhancement of the efficacy of synaptic transmission and the modulation of neurotransmitters release.
Collapse
Affiliation(s)
- Isabel Barón-Mendoza
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, CDMX, México
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, CDMX, México
| |
Collapse
|
32
|
Lee SH, Lee PH, Liang HJ, Tang CH, Chen TF, Cheng TJ, Lin CY. Brain lipid profiles in the spontaneously hypertensive rat after subchronic real-world exposure to ambient fine particulate matter. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 707:135603. [PMID: 31784156 DOI: 10.1016/j.scitotenv.2019.135603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 06/10/2023]
Abstract
Recent studies have illustrated an association between ambient fine particulate matter (PM2.5) exposure and neuronal toxicity in epidemiological studies and animal models. However, the possible molecular effects on brains under real-world exposure to PM2.5 remain unclear. In this pilot study, male spontaneously hypertensive rats were whole-bodily exposed to ambient air from the outdoor environment of Taipei City for 3 months, while the control rats inhaled HEPA-filtered air. The PM2.5-induced phosphatidylcholine and sphingomyelin profiles in the hippocampus, cortex, medulla, cerebellum, and olfactory bulb were assessed by mass spectrometry (MS)-based lipidomics. Partial least squares discriminant analysis (PLS-DA) and the Wilcoxon rank sum test were used to examine the lipid changes between the exposed and control groups. The PLS-DA models showed that phosphatidylcholine and sphingomyelin profiles of the PM2.5 exposure group were different from those of the control group in each brain region except the cortex. More lipid changes were found in the hippocampus, while fewer lipid changes were observed in the olfactory bulb. The lipid alteration in the hippocampus may strengthen membrane integrity, modulate signaling pathways, and avoid accumulation of lipofuscin to counter the PM2.5-induced stress. The lipid changes in the cortex and medulla may respond to PM2.5-induced injury and inflammation; while the lipid changes in the cerebellum were associated with neuron protection. This study suggests that the MS-based lipidomics is a powerful approach to discriminate the brain lipid profiles even at the environmental level of ambient PM2.5 and has the potential to suggest possible adverse health effects in long-term PM2.5 exposure studies.
Collapse
Affiliation(s)
- Sheng-Han Lee
- Institute of Environmental Health, College of Public Health, National Taiwan University, Taiwan; Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, Taiwan
| | - Pei-Hsuan Lee
- Institute of Environmental Health, College of Public Health, National Taiwan University, Taiwan
| | - Hao-Jan Liang
- Institute of Environmental Health, College of Public Health, National Taiwan University, Taiwan
| | - Chuan-Ho Tang
- National Museum of Marine Biology and Aquarium, Taiwan; Institute of Marine Biodiversity and Evolutionary Biology, National Dong Hwa University, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
| | - Tsun-Jen Cheng
- Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, Taiwan; Department of Public Health, National Taiwan University, Taiwan.
| | - Ching-Yu Lin
- Institute of Environmental Health, College of Public Health, National Taiwan University, Taiwan; Department of Public Health, National Taiwan University, Taiwan.
| |
Collapse
|
33
|
Fatemi F, Siassi F, Qorbani M, Sotoudeh G. Higher dietary fat quality is associated with lower anxiety score in women: a cross-sectional study. Ann Gen Psychiatry 2020; 19:14. [PMID: 32127909 PMCID: PMC7045483 DOI: 10.1186/s12991-020-00264-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/10/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The relationship between anxiety and dietary fat quality (DFQ) has not been well studied. The aim of this study was to investigate the relationship between anxiety disorder and fatty acids' intake in women. METHODS This cross-sectional study included 300 women aged 18-49 attending healthcare centers. Dietary exposure was measured by a 168-item semi-quantitative food frequency questionnaire (FFQ). To determine the status of anxiety, the Depression, Anxiety, and Stress Scale (DASS) questionnaire was used. Based on the total score of anxiety, the participants were divided into two groups of without anxiety (< 8) and with anxiety (≥ 8). The relationship between fatty acids intake and odd ratio (OR) for anxiety was analyzed by simple logistic regression. RESULTS About 37.7% of individuals reported anxiety. After adjustment for covariates, an increase in the OR for anxiety was observed across the quintuples of saturated fatty acids (SFAs) (OR 3.17; 95% CI 1.43-7.00; p-trend = 0.005). In addition, higher intakes of monounsaturated fatty acids (MUFAs) (OR 0.15; 95% CI 0.05-0.44; p-trend = 0.001), oleic acid (OR 0.25; 95% CI 0.09-0.67; p-trend = 0.002), alpha-linolenic acid (ALA) (OR 0.07; 95% CI 0.02-0.23; p-trend < 0.001), and n-3:n-6 poly unsaturated fatty acids (PUFAs) (OR 0.56; 95% CI 0.24-1.03; p-trend = 0.02) were found to be related with lower OR of anxiety. CONCLUSION Intake of SFAs was positively related to anxiety disorder, whereas MUFAs, oleic acid, ALA, and n-3: n-6 PUFAs intake were inversely related to anxiety score. For investigating the association of fat intake and anxiety disorder, DFQ may be a useful measure.
Collapse
Affiliation(s)
- Fatemeh Fatemi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Hojatdost street, Naderi street, Keshavarz Blv., Tehran, Iran
| | - Fereydoun Siassi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Hojatdost street, Naderi street, Keshavarz Blv., Tehran, Iran
| | - Mostafa Qorbani
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gity Sotoudeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Hojatdost street, Naderi street, Keshavarz Blv., Tehran, Iran
| |
Collapse
|
34
|
Talamonti E, Sasso V, To H, Haslam RP, Napier JA, Ulfhake B, Pernold K, Asadi A, Hessa T, Jacobsson A, Chiurchiù V, Viscomi MT. Impairment of DHA synthesis alters the expression of neuronal plasticity markers and the brain inflammatory status in mice. FASEB J 2020; 34:2024-2040. [PMID: 31909582 PMCID: PMC7384056 DOI: 10.1096/fj.201901890rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022]
Abstract
Docosahexaenoic acid (DHA) is a ω-3 fatty acid typically obtained from the diet or endogenously synthesized through the action of elongases (ELOVLs) and desaturases. DHA is a key central nervous system constituent and the precursor of several molecules that regulate the resolution of inflammation. In the present study, we questioned whether the impaired synthesis of DHA affected neural plasticity and inflammatory status in the adult brain. To address this question, we investigated neural and inflammatory markers from mice deficient for ELOVL2 (Elovl2-/- ), the key enzyme in DHA synthesis. From our findings, Elovl2-/- mice showed an altered expression of markers involved in synaptic plasticity, learning, and memory formation such as Egr-1, Arc1, and BDNF specifically in the cerebral cortex, impacting behavioral functions only marginally. In parallel, we also found that DHA-deficient mice were characterized by an increased expression of pro-inflammatory molecules, namely TNF, IL-1β, iNOS, caspase-1 as well as the activation and morphologic changes of microglia in the absence of any brain injury or disease. Reintroducing DHA in the diet of Elovl2-/- mice reversed such alterations in brain plasticity and inflammation. Hence, impairment of systemic DHA synthesis can modify the brain inflammatory and neural plasticity status, supporting the view that DHA is an essential fatty acid with an important role in keeping inflammation within its physiologic boundary and in shaping neuronal functions in the central nervous system.
Collapse
Affiliation(s)
- Emanuela Talamonti
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Valeria Sasso
- Laboratory of Experimental NeurorehabilitationIRCCS Santa Lucia FoundationRomeItaly
| | - Hoi To
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | | | | | - Brun Ulfhake
- Department of NeuroscienceKarolinska InstituteStockholmSweden
| | - Karin Pernold
- Department of NeuroscienceKarolinska InstituteStockholmSweden
| | - Abolfazl Asadi
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Tara Hessa
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Anders Jacobsson
- Department of Molecular BiosciencesThe Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Valerio Chiurchiù
- Department of MedicineCampus Bio‐Medico University of RomeRomeItaly
- Laboratory of Resolution of NeuroinflammationIRCCS Santa Lucia FoundationRomeItaly
| | | |
Collapse
|
35
|
Bhatti GK, Reddy AP, Reddy PH, Bhatti JS. Lifestyle Modifications and Nutritional Interventions in Aging-Associated Cognitive Decline and Alzheimer's Disease. Front Aging Neurosci 2020; 11:369. [PMID: 31998117 PMCID: PMC6966236 DOI: 10.3389/fnagi.2019.00369] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a type of incurable neurodegenerative disease that is characterized by the accumulation of amyloid-β (Aβ; plaques) and tau hyperphosphorylation as neurofibrillary tangles (NFTs) in the brain followed by neuronal death, cognitive decline, and memory loss. The high prevalence of AD in the developed world has become a major public health challenge associated with social and economic burdens on individuals and society. Due to there being limited options for early diagnosis and determining the exact pathophysiology of AD, finding effective therapeutic strategies has become a great challenge. Several possible risk factors associated with AD pathology have been identified; however, their roles are still inconclusive. Recent clinical trials of the drugs targeting Aβ and tau have failed to find a cure for the AD pathology. Therefore, effective preventive strategies should be followed to reduce the exponential increase in the prevalence of cognitive decline and dementia, especially AD. Although the search for new therapeutic targets is a great challenge for the scientific community, the roles of lifestyle interventions and nutraceuticals in the prevention of many metabolic and neurodegenerative diseases are highly appreciated in the literature. In this article, we summarize the molecular mechanisms involved in AD pathology and the possible ameliorative action of lifestyle and nutritional interventions including diet, exercise, Calorie restriction (CR), and various bioactive compounds on cognitive decline and dementia. This article will provide insights into the role of non-pharmacologic interventions in the modulation of AD pathology, which may offer the benefit of improving quality of life by reducing cognitive decline and incident AD.
Collapse
Affiliation(s)
- Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Arubala P. Reddy
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - P. Hemachandra Reddy
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Neuroscience and Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Speech, Language and Hearing Sciences Department, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Jasvinder Singh Bhatti
- Department of Biotechnology and Microbial Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India
| |
Collapse
|
36
|
Medina-Ceja L, Villalpando-Vargas F, Girón de la Cruz GI, Lara-Vazquez AM, Flores-Mancilla L, Salazar-Sánchez JC, Morales-Villagrán A. Effect of Chronic Krill Oil Supplement on Seizures Induced by Pentylenetetrazole in the Hippocampus of Adult Rats with Previous Febrile Seizures. J Food Sci 2019; 84:1703-1711. [PMID: 31218711 DOI: 10.1111/1750-3841.14679] [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: 11/10/2018] [Revised: 04/19/2019] [Accepted: 05/05/2019] [Indexed: 11/28/2022]
Abstract
We evaluated the effect of krill oil (KO) supplement on seizures induced by pentylenetetrazole (PTZ) in animals with previous febrile seizures (FSs) induced by hyperthermia to determine its effectiveness in seizure susceptibility and as an anticonvulsant. Male Wistar rats with FS separated into water (W, 1 mL), palm oil (PO, 300 mg/kg, total volume 1 mL), or KO (300 mg/kg, total volume 1 mL) groups. All drugs were administered chronically via the intragastric route. Electrical activity was recorded by intracranial EEG simultaneously with convulsive behavior. All animals' brains were processed by immunofluorescence against GFAP, NeuN, and connexins (Cx); cellular quantification was performed in hippocampus and pyramidal or granular layer thickness was evaluated with cresyl violet (CV) staining. The results showed a significant delay in convulsive behavior and a slight increased survival time after PTZ administration in the group treated with KO compared with PO and W groups. The epileptiform activity showed high amplitude and frequency, with no significant differences between groups, nor were there differences in the number and duration of discharge trains. KO and PO increased the number of astrocytes and the number of neurons compared with the W group. KO and PO decreased the expression of Cx36 without affecting Cx43 expression or the thickness of layers. Based on these data, we consider it important to perform more experiments to determine the anticonvulsant role of KO, taking into account the partial effect found in this study. KO could be used as a coadjuvant of traditional anticonvulsive treatments. PRACTICAL APPLICATION: In this study was evaluated the anticonvulsive effect of a chronic krill oil (KO) supplement in animals with seizures. Results showed that KO had partial anticonvulsive effects measured by EEG activity and convulsive behavior analysis. These data justify further research that looks at KO supplementation as a prospective coadjuvant of pharmacologic management of seizure disorder.
Collapse
Affiliation(s)
- Laura Medina-Ceja
- Laboratory of Neurophysiology, Dept. of Cellular and Molecular Biology, CUCBA, Univ. of Guadalajara, Jalisco, México
| | - Fridha Villalpando-Vargas
- Laboratory of Neurophysiology, Dept. of Cellular and Molecular Biology, CUCBA, Univ. of Guadalajara, Jalisco, México
| | - Gloria I Girón de la Cruz
- Laboratory of Neurophysiology, Dept. of Cellular and Molecular Biology, CUCBA, Univ. of Guadalajara, Jalisco, México
| | - Adriana M Lara-Vazquez
- Laboratory of Neurophysiology, Dept. of Cellular and Molecular Biology, CUCBA, Univ. of Guadalajara, Jalisco, México
| | - Leopoldo Flores-Mancilla
- Laboratory of Neurophysiology and Behavior, Human Medicine and Health Science Academic Unit, Autonomous Univ. of Zacatecas, Zacatecas, México
| | - Juan C Salazar-Sánchez
- Laboratory of Neurophysiology, Dept. of Cellular and Molecular Biology, CUCBA, Univ. of Guadalajara, Jalisco, México
| | | |
Collapse
|
37
|
Dominguez LJ, Barbagallo M. Dietary Strategies and Supplements for the Prevention of Cognitive Decline and Alzheimer’s Disease. OMEGA FATTY ACIDS IN BRAIN AND NEUROLOGICAL HEALTH 2019:231-247. [DOI: 10.1016/b978-0-12-815238-6.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
38
|
Lee S, Kang S, Ang MJ, Kim J, Kim JC, Kim SH, Jeon TI, Jung C, Im SS, Moon C. Deficiency of sterol regulatory element-binding protein-1c induces schizophrenia-like behavior in mice. GENES BRAIN AND BEHAVIOR 2018; 18:e12540. [PMID: 30430717 DOI: 10.1111/gbb.12540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/26/2018] [Accepted: 11/11/2018] [Indexed: 01/12/2023]
Abstract
Schizophrenia is a hereditary disease that approximately 1% of the worldwide population develops. Many studies have investigated possible underlying genes related to schizophrenia. Recently, clinical studies suggested sterol regulatory element-binding protein (SREBP) as a susceptibility gene in patients with schizophrenia. SREBP controls cellular lipid homeostasis by three isoforms: SREBP-1a, SREBP-1c and SREBP-2. This study used SREBP-1c knockout (KO) mice to examine whether a deficiency in SREBP-1c would affect their emotional and psychiatric behaviors. Altered mRNA expression in genes downstream from SREBP-1c was confirmed in the brains of SREBP-1c KO mice. Schizophrenia-like behavior, including hyperactivity during the dark phase, depressive-like behavior, aggressive behavior and deficits in social interaction and prepulse inhibition, was observed in SREBP-1c KO mice. Furthermore, increased volume of the lateral ventricle was detected in SREBP-1c KO mice. The mRNA levels of several γ-aminobutyric acid (GABA)-receptor subtypes and/or glutamic acid decarboxylase 65/67 decreased in the hippocampus and medial prefrontal cortex of SREBP-1c KO mice. Thus, SREBP-1c deficiency may contribute to enlargement of the lateral ventricle and development of schizophrenia-like behaviors and be associated with altered GABAergic transmission.
Collapse
Affiliation(s)
- Sueun Lee
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Juhwan Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Jong Choon Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Sung-Ho Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Tae-Il Jeon
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju, South Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
39
|
Pan X, Green BD. Temporal Effects of Neuron-specific beta-secretase 1 (BACE1) Knock-in on the Mouse Brain Metabolome: Implications for Alzheimer's Disease. Neuroscience 2018; 397:138-146. [PMID: 30496823 DOI: 10.1016/j.neuroscience.2018.11.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 10/22/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022]
Abstract
Beta secretase 1 (BACE1) is an enzyme involved in the pathogenesis of Alzheimer's disease (AD). PLB4 mice are a neuron-specific human BACE1 knock-in mouse model characterized by the accumulation of extracellular Aβ and an AD-like phenotype. In this investigation brain hemispheres from 'young' (4-6 months) and 'old' (8 months) female PLB4 mice and age-matched wild-type littermates underwent targeted LC-MS/MS metabolomic profiling. Powdered lyophilized brain tissue was extracted in ethanol:PBS 85%:15% (v/v)) and a total of 187 metabolites were quantified using a targeted metabolomics methodology. Multivariate statistical analysis produced models distinguished PLB4 from wild type (WT) mice regardless of their age group. Univariate analysis (t-test) found that more brain metabolites were perturbed in 'old' PLB4 mice than 'young'. Carnosine and 8 phosphatidylcholine species were significantly decreased (p < 0.05) in 'young' PLB4 mouse brain. In 'old' PLB4 mice a total of 21 metabolites were perturbed including: leucine, creatinine, putrescine and species of acylcarnitines, lysophosphatidylcholines, phosphatidylcholines and sphingomyelin. Within the PLB4 genotype there were a range of age-dependent increases in metabolites. This study indicates that gender-specific responses occur in models of AD-like pathology, but importantly, when changes in PLB4 mice (where Aβ oligomers predominate) are compared with APP/PS1 mice (where Aβ plaques predominate) there are consistent and also divergent effects on the brain metabolome.
Collapse
Affiliation(s)
- Xiaobei Pan
- Advanced Asset Technology Centre, Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - Brian D Green
- Advanced Asset Technology Centre, Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK; Core Techology Unit for Mass Spectrometry, Faculty of Medicine, Health and Life Sciences, Queen's University Belfast, UK.
| |
Collapse
|
40
|
Tayebati SK. Phospholipid and Lipid Derivatives as Potential Neuroprotective Compounds. Molecules 2018; 23:molecules23092257. [PMID: 30189584 PMCID: PMC6225353 DOI: 10.3390/molecules23092257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022] Open
Abstract
The worldwide demographical trend is changing towards a more elderly population. In particular, this phenomenon is increasing the number of neurodegenerative disease cases (e.g., Alzheimer’s disease) in advanced countries. Therefore, there is a fertile field for neuroprotective approaches to address this problem. A useful strategy to protect the membrane integrity of cells and reduce inflammatory processes. In this context, the neurons represent particularly vulnerable cells. Thus, a protection strategy should include their membrane preservation and improved anti-inflammatory processes. The contribution of phospholipid derivatives to this issue is crucial and many articles evidence their role in both health and disease. On the other hand, some lipids containing choline actively participate to increase the choline levels in the nervous system. It is acknowledged that the cholinergic system plays a pivotal role both in the central and in the peripheral nervous system. Neurons cannot synthesize choline, which is provided by the diet. The reuptake of ACh and its hydrolysis represent the principal source of choline. Therefore, to cover choline needs, choline-containing lipids may be used. There are different works which demonstrate their neuroprotective features This review article analyzes phospholipid and lipid derivatives that through different mechanisms are involved in these protective processes, although, sometimes the same molecules may behave as neurotoxic elements, therefore, their protective machinery should be detailed better.
Collapse
|
41
|
Dominguez LJ, Barbagallo M. Nutritional prevention of cognitive decline and dementia. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:276-290. [PMID: 29957766 PMCID: PMC6179018 DOI: 10.23750/abm.v89i2.7401] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 04/28/2018] [Indexed: 12/28/2022]
Abstract
Cognitive impairment results from a complex interplay of many factors. The most important independent predictor of cognitive decline is age but other contributing factors include demographic, genetic, socio-economic, and environmental parameters, including nutrition. The number of persons with cognitive decline and dementia will increase in the next decades in parallel with aging of the world population. Effective pharmaceutical treatments for age-related cognitive decline are lacking, emphasizing the importance of prevention strategies. There is extensive evidence supporting a relationship between diet and cognitive functions. Thus, nutritional approaches to prevent or slow cognitive decline could have a remarkable public health impact. Several dietary components and supplements have been examined in relation to their association with the development of cognitive decline. A number of studies have examined the role of dietary patterns on late-life cognition, with accumulating evidence that combinations of foods and nutrients may act synergistically to provide stronger benefit than those conferred by individual dietary components. Higher adherence to the Mediterranean dietary pattern has been associated with decreased cognitive decline and incident AD. Another dietary pattern with neuroprotective actions is the Dietary Approach to Stop Hypertension (DASH). The combination of these two dietary patterns has been associated with slower rates of cognitive decline and significant reduction in incident AD. This review evaluates the evidence for the effects of some dietary components, supplements, and dietary patterns as neuroprotective, with potential to delay cognitive decline and the onset of dementia.
Collapse
Affiliation(s)
- Ligia J Dominguez
- Geriatric Unit, Dept. of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy.
| | - Mario Barbagallo
- Geriatric Unit, Dept. of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy.
| |
Collapse
|
42
|
Gogou M, Kolios G. The effect of dietary supplements on clinical aspects of autism spectrum disorder: A systematic review of the literature. Brain Dev 2017; 39:656-664. [PMID: 28438367 DOI: 10.1016/j.braindev.2017.03.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/27/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Autism spectrum disorder is associated with significant social and financial burden and no definite treatment for this entity has been identified, yet. In recent years there has been an increasing interest in the use of dietary interventions as a complementary therapeutic option for these patients. OBJECTIVE The aim of this systematic review is to provide high evidence level literature data about the effect of dietary supplements on clinical aspects of children with autism. METHODS A comprehensive literature search was conducted using Pubmed as the medical database source. Randomized controlled trials conducted in pediatric populations and including measures of clinical outcomes were considered. RESULTS A total of 17 eligible prospective studies were selected. Types of dietary supplements evaluated in these studies included amino acids, fatty acids and vitamins/minerals. N-acetylcysteine was shown to exert a beneficial effect on symptoms of irritability. On the other hand, literature data about the efficacy of d-cycloserine and pyridoxine-magnesium supplements was controversial. No significant effect was identified for fatty acids, N,N-dimethylglycine and inositol. Literature data about ascorbic acid and methyl B12 was few, although some encouraging results were found. No serious adverse events were reported in the vast majority of the studies, while the prevalence of adverse reactions was similar between treatment and placebo groups. CONCLUSIONS The use of dietary supplements in children with autism seems to be a safe practice with encouraging data about their clinical efficacy. More studies are needed to further investigate this issue.
Collapse
Affiliation(s)
- Maria Gogou
- 2nd Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, University General Hospital AHEPA, Thessaloniki, Greece.
| | - George Kolios
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
43
|
Parga JA, García-Garrote M, Martínez S, Raya Á, Labandeira-García JL, Rodríguez-Pallares J. Prostaglandin EP2 Receptors Mediate Mesenchymal Stromal Cell-Neuroprotective Effects on Dopaminergic Neurons. Mol Neurobiol 2017; 55:4763-4776. [DOI: 10.1007/s12035-017-0681-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022]
|
44
|
Cutuli D. Functional and Structural Benefits Induced by Omega-3 Polyunsaturated Fatty Acids During Aging. Curr Neuropharmacol 2017; 15:534-542. [PMID: 27306037 PMCID: PMC5543674 DOI: 10.2174/1570159x14666160614091311] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/16/2016] [Accepted: 05/31/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids (n-3 PUFA) are structural components of the brain and are indispensable for neuronal membrane synthesis. Along with decline in cognition, decreased synaptic density and neuronal loss, normal aging is accompanied by a reduction in n-3 PUFA concentration in the brain in both humans and rodents. Recently, many clinical and experimental studies have demonstrated the importance of n-3 PUFA in counteracting neurodegeneration and agerelated dysfunctions. METHODS This review will focus on the neuroprotective effects of n-3 PUFA on cognitive impairment, neuroinflammation and neurodegeneration during normal aging. Multiple pathways of n-3 PUFA preventive action will be examined. RESULTS Namely, n-3 PUFA have been shown to increase the levels of several signaling factors involved in synaptic plasticity, thus leading to the increase of dendritic spines and synapses as well as the enhancement of hippocampal neurogenesis even at old age. In elderly subjects n-3 PUFA exert anti-inflammatory effects associated with improved cognitive functions. Interestingly, growing evidence highlights n-3 PUFA efficacy in preventing the loss of both gray and white matter volume and integrity. CONCLUSION This review shows that n-3 PUFA are essential for a successful aging and appear as ideal cognitive enhancers to be implemented in nutritional interventions for the promotion of healthy aging.
Collapse
Affiliation(s)
- Debora Cutuli
- Fondazione Santa Lucia of Rome, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| |
Collapse
|
45
|
Sparvero LJ, Amoscato AA, Fink AB, Anthonymuthu T, New L, Kochanek P, Watkins S, Kagan V, Bayır H. Imaging mass spectrometry reveals loss of polyunsaturated cardiolipins in the cortical contusion, hippocampus, and thalamus after traumatic brain injury. J Neurochem 2016; 139:659-675. [PMID: 27591733 PMCID: PMC5323070 DOI: 10.1111/jnc.13840] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 02/03/2023]
Abstract
Traumatic brain injury (TBI) leads to changes in ion fluxes, alterations in mitochondrial function, and increased generation of reactive oxygen species, resulting in secondary tissue damage. Mitochondria play important signaling roles in coordination of multiple metabolic platforms in addition to their well-known role in bioenergetics. Mitochondrial signaling strongly depends on cardiolipin (CL), a mitochondria-specific structurally unusual anionic phospholipid containing four fatty acyl chains. While our previous reports indicated that CL is selectively oxidized and presents itself as a target for the redox therapy following TBI, the topography of changes of CL in the injured brain remained to be defined. Here, we present a matrix-assisted laser desorption/ionization imaging study which reports regio-specific changes in CL, in a controlled cortical impact model of TBI in rats. Matrix-assisted laser desorption/ionization imaging revealed that TBI caused early decreases in CL in the contusional cortex, ipsilateral hippocampus, and thalamus with the most highly unsaturated CL species being most susceptible to loss. Phosphatidylinositol was the only other lipid species that exhibited a significant decrease, albeit to a lesser extent than CL. Signals for other lipids remained unchanged. This is the first study evaluating the spatial distribution of CL loss after acute brain injury. We propose that the CL loss may constitute an upstream mechanism for CL-driven signaling in different brain regions as an early response mechanism and may also underlie the bioenergetic changes that occur in hippocampal, cortical, and thalamic mitochondria after TBI.
Collapse
Affiliation(s)
- L. J. Sparvero
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - A. A. Amoscato
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - A. B. Fink
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - T. Anthonymuthu
- Department of Critical Care Medicine, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - L.E. New
- Department of Critical Care Medicine, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - P.M. Kochanek
- Department of Critical Care Medicine, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - S. Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - V.E. Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - H. Bayır
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
46
|
Musto AE, Rosencrans RF, Walker CP, Bhattacharjee S, Raulji CM, Belayev L, Fang Z, Gordon WC, Bazan NG. Dysfunctional epileptic neuronal circuits and dysmorphic dendritic spines are mitigated by platelet-activating factor receptor antagonism. Sci Rep 2016; 6:30298. [PMID: 27444269 PMCID: PMC4957208 DOI: 10.1038/srep30298] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/30/2016] [Indexed: 01/28/2023] Open
Abstract
Temporal lobe epilepsy or limbic epilepsy lacks effective therapies due to a void in understanding the cellular and molecular mechanisms that set in motion aberrant neuronal network formations during the course of limbic epileptogenesis (LE). Here we show in in vivo rodent models of LE that the phospholipid mediator platelet-activating factor (PAF) increases in LE and that PAF receptor (PAF-r) ablation mitigates its progression. Synthetic PAF-r antagonists, when administered intraperitoneally in LE, re-establish hippocampal dendritic spine density and prevent formation of dysmorphic dendritic spines. Concomitantly, hippocampal interictal spikes, aberrant oscillations, and neuronal hyper-excitability, evaluated 15–16 weeks after LE using multi-array silicon probe electrodes implanted in the dorsal hippocampus, are reduced in PAF-r antagonist-treated mice. We suggest that over-activation of PAF-r signaling induces aberrant neuronal plasticity in LE and leads to chronic dysfunctional neuronal circuitry that mediates epilepsy.
Collapse
Affiliation(s)
- Alberto E Musto
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Robert F Rosencrans
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Chelsey P Walker
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Chittalsinh M Raulji
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA.,Department of Pediatrics, Hematology-Oncology, Louisiana State University Health Sciences Center and Children's Hospital of New Orleans, New Orleans, Louisiana 70118, USA
| | - Ludmila Belayev
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Zhide Fang
- Biostatistics, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - William C Gordon
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, Louisiana 70112, USA
| |
Collapse
|
47
|
Asirvatham-Jeyaraj N, Fink GD. Possible role for brain prostanoid pathways in the development of angiotensin II-salt hypertension in rats. Am J Physiol Regul Integr Comp Physiol 2016; 311:R232-42. [PMID: 27225954 DOI: 10.1152/ajpregu.00535.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/19/2016] [Indexed: 11/22/2022]
Abstract
Prostanoids generated by the cyclooxygenase (COX) pathway appear to contribute to the neurogenic hypertension (HTN) in rats. The first goal of this study was to establish the time frame during which prostanoids participate in ANG II-salt HTN. We induced HTN using ANG II (150 ng·kg(-1)·min(-1) sc) infusion for 14 days in rats on a high-salt (2% NaCl) diet. When ketoprofen pretreatment was combined with treatment during the first 7 days of ANG II infusion, development of HTN and increased neurogenic pressor activity (indexed by the depressor response to ganglion blockade) were significantly attenuated for the entire ANG II infusion period. This suggests that prostanoid generation caused by administration of ANG II and salt leads to an increase in neurogenic pressor activity and blood pressure (BP) via a mechanism that persists without the need for continuing prostanoid input. The second goal of this study was to determine whether prostanoid products specifically in the brain contribute to HTN development. Expression of prostanoid pathway genes was measured in brain regions known to affect neurogenic BP regulation. ANG II-treated rats exhibited changes in gene expression of phospholipase A2 (upregulated in organum vasculosum of the lamina terminalis, paraventricular nucleus, nucleus of the solitary tract, and middle cerebral artery) and lipocalin-type prostaglandin D synthase (upregulated in the organum vasculosum of the lamina terminalis). On the basis of our results, we propose that activation of the brain prostanoid synthesis pathway both upstream and downstream from COX at early stages plays an important role in the development of the neurogenic component of ANG II-salt HTN.
Collapse
Affiliation(s)
| | - Gregory D Fink
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
48
|
Du SQ, Wang XR, Xiao LY, Tu JF, Zhu W, He T, Liu CZ. Molecular Mechanisms of Vascular Dementia: What Can Be Learned from Animal Models of Chronic Cerebral Hypoperfusion? Mol Neurobiol 2016; 54:3670-3682. [PMID: 27206432 DOI: 10.1007/s12035-016-9915-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/03/2016] [Indexed: 01/06/2023]
Abstract
Vascular dementia (VD) is defined as a progressive neurodegenerative disease of cognitive decline, attributable to cerebrovascular factors. Numerous studies have demonstrated that chronic cerebral hypoperfusion (CCH) is associated with the initiation and progression of VD and Alzheimer's disease (AD). Suitable animal models were established to replicate such pathological condition in experimental research, which contributes largely to comprehending causal relationships between CCH and cognitive impairment. The most widely used experimental model of VD and CCH is permanent bilateral common carotid artery occlusion in rats. In CCH models, changes of learning and memory, cerebral blood flow (CBF), energy metabolism, and neuropathology initiated by ischemia were revealed. However, in order to achieve potential therapeutic targets, particular mechanisms in cognitive and neuropathological changes from CCH to dementia should be investigated. Recent studies have shown that hypoperfusion resulted in a chain of disruption of homeostatic interactions, including oxidative stress, neuroinflammation, neurotransmitter system dysfunction, mitochondrial dysfunction, disturbance of lipid metabolism, and alterations of growth factors. Evidence from experimental studies that elucidate the damaging effects of such imbalances suggests their critical roles in the pathogenesis of VD. The present review provides a summary of the achievements in mechanisms made with the CCH models, permits an understanding of the causative role played by CCH in VD, and highlights preventative and therapeutic prospects.
Collapse
Affiliation(s)
- Si-Qi Du
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Ling-Yong Xiao
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Jian-Feng Tu
- Beijing University of Chinese Medicine, 11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Wen Zhu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Tian He
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China
| | - Cun-Zhi Liu
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, 23 Meishuguanhou Street, Dongcheng District, Beijing, 100010, China.
| |
Collapse
|
49
|
Bošković M, Vovk T, Koprivšek J, Plesničar BK, Grabnar I. Vitamin E and essential polyunsaturated fatty acids supplementation in schizophrenia patients treated with haloperidol. Nutr Neurosci 2016; 19:156-61. [DOI: 10.1179/1476830514y.0000000139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Marija Bošković
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Tomaž Vovk
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Koprivšek
- Department of Psychiatry, University Clinical Centre Maribor, Maribor, Slovenia
| | | | - Iztok Grabnar
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
50
|
Berger GE, Smesny S, Schäfer MR, Milleit B, Langbein K, Hipler UC, Milleit C, Klier CM, Schlögelhofer M, Holub M, Holzer I, Berk M, McGorry PD, Sauer H, Amminger GP. Niacin Skin Sensitivity Is Increased in Adolescents at Ultra-High Risk for Psychosis. PLoS One 2016; 11:e0148429. [PMID: 26894921 PMCID: PMC4764507 DOI: 10.1371/journal.pone.0148429] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/18/2016] [Indexed: 12/14/2022] Open
Abstract
Background Most studies provide evidence that the skin flush response to nicotinic acid (niacin) stimulation is impaired in schizophrenia. However, only little is known about niacin sensitivity in the ultra-high risk (UHR) phase of psychotic disorders. Methods We compared visual ratings of niacin sensitivity between adolescents at UHR for psychosis according to the one year transition outcome (UHR-T n = 11; UHR-NT n = 55) with healthy controls (HC n = 25) and first episode schizophrenia patients (FEP n = 25) treated with atypical antipsychotics. Results Contrary to our hypothesis niacin sensitivity of the entire UHR group was not attenuated, but significantly increased compared to the HC group, whereas no difference could be found between the UHR-T and UHR-NT groups. As expected, niacin sensitivity of FEP was attenuated compared to HC group. In UHR individuals niacin sensitivity was inversely correlated with omega-6 and -9 fatty acids (FA), but positively correlated with phospholipase A2 (inPLA2) activity, a marker of membrane lipid repair/remodelling. Conclusions Increased niacin sensitivity in UHR states likely indicates an impaired balance of eicosanoids and omega-6/-9 FA at a membrane level. Our findings suggest that the emergence of psychosis is associated with an increased mobilisation of eicosanoids prior to the transition to psychosis possibly reflecting a “pro-inflammatory state”, whereas thereafter eicosanoid mobilisation seems to be attenuated. Potential treatment implications for the UHR state should be further investigated.
Collapse
Affiliation(s)
- Gregor E. Berger
- University Hospital of Child and Adolescent Psychiatry, University of Zurich, Neumünsterallee 9, 8032 Zurich, Switzerland
- Orygen Youth Health Research Centre, The University of Melbourne, Locked Bag 10, 35 Poplar Road Parkville, Victoria 3052, Melbourne, Australia
| | - Stefan Smesny
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
- * E-mail:
| | - Miriam R. Schäfer
- Orygen Youth Health Research Centre, The University of Melbourne, Locked Bag 10, 35 Poplar Road Parkville, Victoria 3052, Melbourne, Australia
- Department of Child and Adolescent Psychiatry, Medical University of Vienna, Währingergürtel 18–20, A–1090 Vienna, Austria
| | - Berko Milleit
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - Kerstin Langbein
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - Uta-Christina Hipler
- Department of Dermatology, University Hospital Jena, Erfurter Straße 35, D-07743 Jena, Germany
| | - Christine Milleit
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - Claudia M. Klier
- Department of Child and Adolescent Psychiatry, Medical University of Vienna, Währingergürtel 18–20, A–1090 Vienna, Austria
| | - Monika Schlögelhofer
- Department of Child and Adolescent Psychiatry, Medical University of Vienna, Währingergürtel 18–20, A–1090 Vienna, Austria
| | - Magdalena Holub
- Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Ingrid Holzer
- Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Michael Berk
- Deakin University of Melbourne, School of Medicine, Barwon Health, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, Parkville, Australia
| | - Patrick D. McGorry
- Orygen Youth Health Research Centre, The University of Melbourne, Locked Bag 10, 35 Poplar Road Parkville, Victoria 3052, Melbourne, Australia
| | - Heinrich Sauer
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - G. Paul Amminger
- Orygen Youth Health Research Centre, The University of Melbourne, Locked Bag 10, 35 Poplar Road Parkville, Victoria 3052, Melbourne, Australia
- Department of Child and Adolescent Psychiatry, Medical University of Vienna, Währingergürtel 18–20, A–1090 Vienna, Austria
| |
Collapse
|