1
|
Moschonis G, Sarapis K, Resciniti S, Hall R, Yim K, Tonkovic M, Fitzgerald C, Anixiadis F, Vinh A, Dinh QN, Cronin RA, Hale MW, Wright BJ, Pane M, Tuck CJ, Biesiekierski JR. Evaluation of a probiotic blend on psychosocial health and biomarkers of inflammatory, immune and stress response in adults with subthreshold depression: a double-blind, randomised, placebo-controlled trial. Br J Nutr 2024:1-15. [PMID: 39468832 DOI: 10.1017/s0007114524001703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
This study examined the efficacy of a probiotic in reducing depressive symptom severity in people with subthreshold depression. In a double-blind, randomised, placebo-controlled trial, a probiotic (1 × 10^9 live cells per strain: Limosilactobacillus fermentum LF16 (DSM26956), Lacticaseibacillus rhamnosus LR06 (DSM21981), Lactiplantibacillus plantarum LP01 (LMG P-21021) and Bifidobacterium longum 04 (DSM23233)) or placebo was taken daily for 12 weeks. Data were collected at baseline, 6 and 12 weeks including psychological symptom severity (Beck Depression Inventory, BDI; Patient Health Questionnaire, PHQ; Hospital Anxiety Depression Scale, HADS; and Depression Anxiety and Stress Scale, DASS). Biomarkers of glycaemia, inflammation (high-sensitivity C-reactive protein, hs-CRP), antioxidant status (total glutathione (GSH)) and stress (cortisol awakening response, CAR) were also measured. Thirty-nine participants (nineteen probiotic; twenty placebo) were enrolled. There were no significant between-group differences in the examined psychological symptom severity scores, despite certain significant within-group changes observed in both groups from baseline to 6 and/or 12 weeks of follow-up. Regarding biomarkers, the probiotic group showed reduced hs-CRP (-1520; 95 % CI -273·7, -2766·2 ng/dl) and CAR (-0·28; 95 % CI -0·05, -0·51 μg/dl) at 12 weeks, but increased total GSH (3·9; 95 % CI 0·1, 7·5 ng/dl) at 6 weeks, compared with the placebo. The current study reported favourable decreases in depressive symptoms in both groups. Although the within-group changes observed in the probiotic group were supported by favourable inflammatory, antioxidant status and stress biomarker changes compared with the placebo, further research is required to shed more light on the role of gut microbiota modulation on emotional regulation.
Collapse
Affiliation(s)
- George Moschonis
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
- La Trobe Institute for Sustainable Agriculture & Food (LISAF), La Trobe University, VIC3086, Australia
| | - Katerina Sarapis
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Stephanie Resciniti
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Renate Hall
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Kanny Yim
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Matilda Tonkovic
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Clare Fitzgerald
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Fay Anixiadis
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC3086, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC3086, Australia
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC3086, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC3086, Australia
| | - Rachael A Cronin
- Department of Psychology, Counselling and Therapy, La Trobe University, Albury Wodonga, VIC3690, Australia
| | - Matthew W Hale
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne, VIC3086, Australia
| | - Bradley J Wright
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne, VIC3086, Australia
| | - Marco Pane
- Probiotical Research srl, Novara, 28100, Italy
| | - Caroline J Tuck
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
- Department of Nursing and Allied Health, Swinburne University, Melbourne, VIC3122, Australia
| | - Jessica R Biesiekierski
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC3086, Australia
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, VIC3168, Australia
| |
Collapse
|
2
|
Petersen KA, Zong W, Depoy LM, Scott MR, Shankar VG, Burns JN, Cerwensky AJ, Kim SM, Ketchesin KD, Tseng GC, McClung CA. Comparative rhythmic transcriptome profiling of human and mouse striatal subregions. Neuropsychopharmacology 2024; 49:796-805. [PMID: 38182777 PMCID: PMC10948754 DOI: 10.1038/s41386-023-01788-w] [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: 08/24/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/07/2024]
Abstract
The human striatum can be subdivided into the caudate, putamen, and nucleus accumbens (NAc). In mice, this roughly corresponds to the dorsal medial striatum (DMS), dorsal lateral striatum (DLS), and ventral striatum (NAc). Each of these structures have some overlapping and distinct functions related to motor control, cognitive processing, motivation, and reward. Previously, we used a "time-of-death" approach to identify diurnal rhythms in RNA transcripts in these three human striatal subregions. Here, we identify molecular rhythms across similar striatal subregions collected from C57BL/6J mice across 6 times of day and compare results to the human striatum. Pathway analysis indicates a large degree of overlap between species in rhythmic transcripts involved in processes like cellular stress, energy metabolism, and translation. Notably, a striking finding in humans is that small nucleolar RNAs (snoRNAs) and long non-coding RNAs (lncRNAs) are among the most highly rhythmic transcripts in the NAc and this is not conserved in mice, suggesting the rhythmicity of RNA processing in this region could be uniquely human. Furthermore, the peak timing of overlapping rhythmic genes is altered between species, but not consistently in one direction. Taken together, these studies reveal conserved as well as distinct transcriptome rhythms across the human and mouse striatum and are an important step in understanding the normal function of diurnal rhythms in humans and model organisms in these regions and how disruption could lead to pathology.
Collapse
Affiliation(s)
- Kaitlyn A Petersen
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wei Zong
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lauren M Depoy
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline R Scott
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vaishnavi G Shankar
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer N Burns
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison J Cerwensky
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sam-Moon Kim
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kyle D Ketchesin
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Colleen A McClung
- Department of Psychiatry, Translational Neuroscience Program, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Yamanishi K, Hata M, Gamachi N, Watanabe Y, Yamanishi C, Okamura H, Matsunaga H. Molecular Mechanisms of IL18 in Disease. Int J Mol Sci 2023; 24:17170. [PMID: 38139000 PMCID: PMC10743479 DOI: 10.3390/ijms242417170] [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: 09/25/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Interleukin 18 (IL18) was originally identified as an inflammation-induced cytokine that is secreted by immune cells. An increasing number of studies have focused on its non-immunological functions, with demonstrated functions for IL18 in energy homeostasis and neural stability. IL18 is reportedly required for lipid metabolism in the liver and brown adipose tissue. Furthermore, IL18 (Il18) deficiency in mice leads to mitochondrial dysfunction in hippocampal cells, resulting in depressive-like symptoms and cognitive impairment. Microarray analyses of Il18-/- mice have revealed a set of genes with differential expression in liver, brown adipose tissue, and brain; however, the impact of IL18 deficiency in these tissues remains uncertain. In this review article, we discuss these genes, with a focus on their relationships with the phenotypic disease traits of Il18-/- mice.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Masaki Hata
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Naomi Gamachi
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, Hirakata 573-0122, Osaka, Japan; (Y.W.); (C.Y.)
| | - Chiaki Yamanishi
- Hirakata General Hospital for Developmental Disorders, Hirakata 573-0122, Osaka, Japan; (Y.W.); (C.Y.)
| | - Haruki Okamura
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| |
Collapse
|
4
|
Stankiewicz AM, Jaszczyk A, Goscik J, Juszczak GR. Stress and the brain transcriptome: Identifying commonalities and clusters in standardized data from published experiments. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110558. [PMID: 35405299 DOI: 10.1016/j.pnpbp.2022.110558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022]
Abstract
Interpretation of transcriptomic experiments is hindered by many problems including false positives/negatives inherent to big-data methods and changes in gene nomenclature. To find the most consistent effect of stress on brain transcriptome, we retrieved data from 79 studies applying animal models and 3 human studies investigating post-traumatic stress disorder (PTSD). The analyzed data were obtained either with microarrays or RNA sequencing applied to samples collected from more than 1887 laboratory animals and from 121 human subjects. Based on the initial database containing a quarter million differential expression effect sizes representing transcripts in three species, we identified the most frequently reported genes in 223 stress-control comparisons. Additionally, the analysis considers sex, individual vulnerability and contribution of glucocorticoids. We also found an overlap between gene expression in PTSD patients and animals which indicates relevance of laboratory models for human stress response. Our analysis points to genes that, as far as we know, were not specifically tested for their role in stress response (Pllp, Arrdc2, Midn, Mfsd2a, Ccn1, Htra1, Csrnp1, Tenm4, Tnfrsf25, Sema3b, Fmo2, Adamts4, Gjb1, Errfi1, Fgf18, Galnt6, Slc25a42, Ifi30, Slc4a1, Cemip, Klf10, Tom1, Dcdc2c, Fancd2, Luzp2, Trpm1, Abcc12, Osbpl1a, Ptp4a2). Provided transcriptomic resource will be useful for guiding the new research.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Bialystok, Poland
| | - Grzegorz R Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland.
| |
Collapse
|
5
|
Kang J, Oteiza PI, Milenkovic D. (-)-Epicatechin exerts positive effects on anxiety in high fat diet-induced obese mice through multi-genomic modifications in the hippocampus. Food Funct 2022; 13:10623-10641. [PMID: 36168829 DOI: 10.1039/d2fo01897g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Obesity is associated with increased occurrence of cognitive and mood disorders. While consumption of high-fat diets (HFD) and associated obesity could have a detrimental impact on the brain, dietary bioactives may mitigate these harmful effects. We previously observed that (-)-epicatechin (EC) can mitigate HFD-induced anxiety-associated behaviors in mice. The aim of our study is to investigate the molecular mechanisms of EC actions in the hippocampus which underlies its anti-anxiety effects in HFD-fed mice using a multi-genomic approach. Healthy eight-week old male C57BL/6J mice were fed for 24 weeks either: (A) a control diet containing 10% total calories from fat; (B) a HFD containing 45% total calories from fat; or (C) the HFD supplemented with 20 mg EC per kg body weight. Hippocampi were isolated for genomic analysis using Affymetrix arrays, followed by in-depth bioinformatic analyses. Genomic analysis demonstrated that EC induced significant changes in mouse hippocampal global gene expression. We observed changes in the expression of 1001 protein-coding genes, 241 miRNAs, and 167 long non-coding RNAs. Opposite gene expression profiles were observed when the gene expression profile obtained upon EC supplementation was compared to the profile obtained after consumption of the HFD. Functionality analysis revealed that the differentially expressed genes regulate processes involved in neurofunction, inflammation, endothelial function, cell-cell adhesion, and cell signaling. In summary, the capacity of EC to mitigate anxiety-related behaviors in HFD-induced obese mice can be in part explained by its capacity to exert complex genomic modifications in the hippocampus, counteracting changes driven by consumption of the HFD and/or associated obesity.
Collapse
Affiliation(s)
- Jiye Kang
- Department of Nutrition, University of California, Davis, One Shields Avenue, 95616 Davis, California, USA. .,Department of Environmental Toxicology, University of California, Davis, One Shields Avenue, 95616 Davis, California, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, One Shields Avenue, 95616 Davis, California, USA. .,Department of Environmental Toxicology, University of California, Davis, One Shields Avenue, 95616 Davis, California, USA
| | - Dragan Milenkovic
- Department of Nutrition, University of California, Davis, One Shields Avenue, 95616 Davis, California, USA.
| |
Collapse
|
6
|
Acute stress induces severe neural inflammation and overactivation of glucocorticoid signaling in interleukin-18-deficient mice. Transl Psychiatry 2022; 12:404. [PMID: 36151082 PMCID: PMC9508168 DOI: 10.1038/s41398-022-02175-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022] Open
Abstract
Interleukin-18 (IL18) is an inflammatory cytokine that is related to psychiatric disorders such as depression and cognitive impairment. We previously found that IL18 deficiency may cause hippocampal impairment, resulting in depression-like behavioral changes. However, the potential role of IL18 in stressful conditions remains uncertain. In the present study, we examined the effect of IL18 on neural inflammation and stress tolerance during acute stress. Littermate Il18+/+ and Il18-/- mice were exposed to a single restraint stress for 6 h, and all assessments were performed 18 h after the mice were released from the restraint. In Il18-/- mice exposed to acute stress, the immobility times in both the forced swim test and tail suspension test were decreased, although no difference was observed in Il18+/+ mice. Il1β, Il6, and Tnfα expression levels in the hippocampus of stressed Il18-/- mice were significantly higher than those in the other groups. Moreover, the numbers of astrocytes and microglia, including those in the active form, were also increased compared with those in other groups. Regarding the molecular mechanism, the HSF5 and TTR genes were specifically expressed in stressed Il18-/- mice. As a potential treatment, intracerebral administration of IL18 to Il18-/- mice resulted in partial recovery of changes in behavioral assessments. Our results revealed that IL18-deficient mice were more sensitive and had a longer response to acute stress than that in normal mice. In addition, neural inflammation and augmentation of glucocorticoid signals caused by stress were more intense and remained longer in Il18-/- mice, resulting in behavioral changes. In conclusion, IL18 might be an indispensable factor that modulates the stress response and maintains balance between neural inflammation and glucocorticoid signaling.
Collapse
|
7
|
Exploring Molecular Mechanisms Involved in the Development of the Depression-Like Phenotype in Interleukin-18-Deficient Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9975865. [PMID: 34708129 PMCID: PMC8545524 DOI: 10.1155/2021/9975865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/18/2021] [Indexed: 11/18/2022]
Abstract
Interleukin-18 (IL-18) is an inflammatory cytokine that has been linked to energy homeostasis and psychiatric symptoms such as depression and cognitive impairment. We previously revealed that deficiency in IL-18 led to hippocampal abnormalities and resulted in depression-like symptoms. However, the impact of IL-18 deficiency on other brain regions remains to be clarified. In this study, we first sought to confirm that IL-18 expression in neural cells can be found in human brain tissue. Subsequently, we examined the expression of genes in the prefrontal cortex of Il18−/− mice and compared it with gene expression in mice subjected to a chronic mild stress model of depression. Extracted genes were further analyzed using Ingenuity® Pathway Analysis, in which 18 genes common to both the chronic mild stressed model and Il18−/− mice were identified. Of those, 16 were significantly differentially expressed between Il18+/+ and Il18−/− mice. We additionally measured protein expression of α-2-HS-glycoprotein (AHSG) and transthyretin (TTR) in serum and the brain. In the prefrontal cortex of Il18−/− mice, TTR but not AHSG was significantly decreased. Conversely, in the serum of Il18−/− mice, AHSG was significantly increased but not TTR. Therefore, our results suggest that in IL-18-deficit conditions, TTR in the brain is one of the mediators causally related to depression, and AHSG in peripheral organs is one of the regulators inducing energy imbalance. Moreover, this study suggests a possible “signpost” to clarify the molecular mechanisms commonly underlying the immune system, energy metabolism, neural function, and depressive disorders.
Collapse
|
8
|
Barthelson K, Pederson SM, Newman M, Lardelli M. Brain Transcriptome Analysis of a Protein-Truncating Mutation in Sortilin-Related Receptor 1 Associated With Early-Onset Familial Alzheimer's Disease Indicates Early Effects on Mitochondrial and Ribosome Function. J Alzheimers Dis 2021; 79:1105-1119. [PMID: 33386808 DOI: 10.3233/jad-201383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The early cellular stresses leading to Alzheimer's disease (AD) remain poorly understood because we cannot access living, asymptomatic human AD brains for detailed molecular analyses. Sortilin-related receptor 1 (SORL1) encodes a multi-domain receptor protein genetically associated with both rare, early-onset familial AD (EOfAD) and common, sporadic, late-onset AD (LOAD). SORL1 protein has been shown to act in the trafficking of the amyloid β A4 precursor protein (AβPP) that is proteolysed to form one of the pathological hallmarks of AD, amyloid-β (Aβ) peptide. However, other functions of SORL1 in AD are less well understood. OBJECTIVE To investigate the effects of heterozygosity for an EOfAD-like mutation in SORL1 on the brain transcriptome of young-adult mutation carriers using zebrafish as a model organism. METHODS We performed targeted mutagenesis to generate an EOfAD-like mutation in the zebrafish orthologue of SORL1 and performed RNA-sequencing on mRNA isolated from the young adult brains of siblings in a family of fish either wild type (non-mutant) or heterozygous for the EOfAD-like mutation. RESULTS We identified subtle differences in gene expression indicating changes in mitochondrial and ribosomal function in the mutant fish. These changes appear to be independent of changes in mitochondrial content or the expression of AβPP-related proteins in zebrafish. CONCLUSION These findings provided evidence supporting that EOfAD mutations in SORL1 affect mitochondrial and ribosomal function and provide the basis for future investigation elucidating the nature of these effects.
Collapse
Affiliation(s)
- Karissa Barthelson
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Stephen Martin Pederson
- Bioinformatics Hub, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Morgan Newman
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| | - Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA, Australia
| |
Collapse
|
9
|
Xu E, Wang B, Lu S, Zhang C, Zhu L, Liu X, Bai M, Li Y. Tandem mass tag-based quantitative proteomic analysis of the liver reveals potential protein targets of Xiaochaihutang in CUMS model of depression. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1181:122898. [PMID: 34479180 DOI: 10.1016/j.jchromb.2021.122898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022]
Abstract
Depression is a global mental disorder disease and greatly threatened human health. Xiaochaihutang (XCHT) has been used successfully in treatment of depression for many years in China, but the mechanism is unclear. Using the chronic unpredictable mild stress (CUMS) mice model of depression, the present study aimed to reveal possible antidepressant mechanisms of XCHT from the perspective of liver by analyzing hepatic proteomics in mice. Bioinformatics analysis identified 31 differentially expressed proteins (DEPs), including 5 upregulated and 26 downregulated proteins, between the CUMS model and XCHT groups. The bile secretion pathway was found by KEGG pathway analysis of these DEPs. Four of the 31 differentially expressed proteins, including 2 active proteins involved in bile secretion, carbonic anhydrase 2 (CA2) and cystic fibrosis transmembrane conductance regulator (CFTR), were selected to verify their genes. Four genes (Cyp7a1, Fxr, Shp and Ntcp) related to bile acid synthesis and transport were further investigated by quantitative real-time polymerase chain reaction (qRT-PCR). Both biochemical tests and gene studies demonstrated that CUMS affected bile acid synthesis and transport, while XCHT regulated this pathway. The results indicated that there may be a potential relationship between CUMS induced depression and hepatic injury caused by increased bile acid, and also provide a novel insight to understand the underlying anti-depression mechanisms of XCHT.
Collapse
Affiliation(s)
- Erping Xu
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Baoying Wang
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shuaifei Lu
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Changjing Zhang
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Leilei Zhu
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xueying Liu
- Basic Medical School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ming Bai
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China.
| | - Yucheng Li
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
10
|
Transthyretin as a Biomarker to Predict and Monitor Major Depressive Disorder Identified by Whole-Genome Transcriptomic Analysis in Mouse Models. Biomedicines 2021; 9:biomedicines9091124. [PMID: 34572310 PMCID: PMC8469805 DOI: 10.3390/biomedicines9091124] [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: 08/11/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Accumulations of stressful life events result in the onset of major depressive disorder (MDD). Comprehensive genomic analysis is required to elucidate pathophysiological changes and identify applicable biomarkers. Methods: Transcriptomic analysis was performed on different brain parts of a chronic mild stress (CMS)-induced MDD mouse model followed by systemic analysis. QPCR and ELISA were utilized for validation in mice and patients. Results: The highest numbers of genes with significant changes induced by CMS were 505 in the amygdala followed by 272 in the hippocampus (twofold changes; FDR, p < 0.05). Enrichment analysis indicated that the core-enriched genes in CMS-treated mice were positively enriched for IFN-γ response genes in the amygdala, and hedgehog signaling in the hippocampus. Transthyretin (TTR) was severely reduced in CMS-treated mice. In patients with diagnosed MDD, serum concentrations of TTR were reduced by 48.7% compared to controls (p = 0.0102). Paired samples from patients with MDD demonstrated a further 66.3% increase in TTR at remission compared to the acute phase (p = 0.0339). Conclusions: This study provides comprehensive information on molecular networks related to MDD as a basis for further investigation and identifies TTR for MDD monitoring and management. A clinical trial with bigger patient cohort should be conducted to validate this translational study.
Collapse
|
11
|
Wang D, Wang L, Han J, Zhang Z, Fang B, Chen F. Bioinformatics-Based Analysis of the lncRNA-miRNA-mRNA Network and TF Regulatory Network to Explore the Regulation Mechanism in Spinal Cord Ischemia/Reperfusion Injury. Front Genet 2021; 12:650180. [PMID: 33986769 PMCID: PMC8110913 DOI: 10.3389/fgene.2021.650180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/29/2021] [Indexed: 11/28/2022] Open
Abstract
Background Spinal cord ischemia/reperfusion injury (SCII) is a catastrophic complication involved with cardiovascular, spine, and thoracic surgeries and can lead to paraplegia. Nevertheless, the molecular mechanism of SCII remain ill-defined. Methods Expression profiling (GSE138966) data were obtained from GEO database. Then, differentially expressed (DE) lncRNAs and DEmRNAs were screened out with p < 0.05, and | fold change| > 1.5. Aberrant miRNAs expression in SCII was obtained from PubMed. Functional enrichment analysis of overlapping DEmRNAs between predicted mRNAs in miRDB database and DEmRNAs obtained from GSE138966 was performed using cluster Profiler R package. The lncRNA-miRNA-mRNA competitive endogenous RNA (ceRNA) network was established in light of ceRNA theory. The key lncRNAs in the ceRNA network were identified by topological analysis. Subsequently, key lncRNAs related ceRNA-pathway network and transcription factors (TFs)-mRNAs network were constructed. Simultaneously, the expression levels of hub genes were measured via qRT-PCR. Results The results in this study indicated that 76 miRNAs, 1373 lncRNAs, and 4813 mRNAs were differentially expressed in SCII. A SCII-related ceRNA network was constructed with 154 ncRNAs, 139 mRNAs, and 51 miRNAs. According topological analysis, six lncRNAs (NONRATT019236.2, NONRATT009530.2, NONRATT026999.2, TCONS_00032391, NONRATT023112.2, and NONRATT021956.2) were selected to establish the ceRNA-pathway network, and then two candidate hub lncRNAs (NONRATT009530.2 and NONRATT026999.2) were identified. Subsequently, two lncRNA-miRNA-mRNA regulatory axes were identified. NONRATT026999.2 and NONRATT009530.2 might involve SCII via miR-20b-5p/Map3k8 axis based on the complex ceRNA network. SP1 and Hnf4a acting as important TFs might regulate Map3k8. Furthermore, qRT-PCR results showed that the NONRATT009530.2, NONRATT026999.2, Map3k8, Hfn4a, and SP1 were significantly upregulated in SCII of rats, while the miR-20b-5p was downregulated. Conclusion Our results offer a new insight to understand the ceRNA regulation mechanism in SCII and identify highlighted lncRNA-miRNA-mRNA axes and two key TFs as potential targets for prevention and treatment of SCII.
Collapse
Affiliation(s)
- Dan Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Limei Wang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Jie Han
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Zaili Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Fengshou Chen
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Nakajima K, Miranda A, Craig DW, Shekhtman T, Kmoch S, Bleyer A, Szelinger S, Kato T, Kelsoe JR. Ntrk1 mutation co-segregating with bipolar disorder and inherited kidney disease in a multiplex family causes defects in neuronal growth and depression-like behavior in mice. Transl Psychiatry 2020; 10:407. [PMID: 33235206 PMCID: PMC7687911 DOI: 10.1038/s41398-020-01087-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/03/2022] Open
Abstract
Previously, we reported a family in which bipolar disorder (BD) co-segregates with a Mendelian kidney disorder linked to 1q22. The causative renal gene was later identified as MUC1. Genome-wide linkage analysis of BD in the family yielded a peak at 1q22 that encompassed the NTRK1 and MUC1 genes. NTRK1 codes for TrkA (Tropomyosin-related kinase A) which is essential for development of the cholinergic nervous system. Whole genome sequencing of the proband identified a damaging missense mutation, E492K, in NTRK1. Induced pluripotent stem cells were generated from family members, and then differentiated to neural stem cells (NSCs). E492K NSCs had reduced neurite outgrowth. A conditional knock-in mouse line, harboring the point mutation in the brain, showed depression-like behavior in the tail suspension test following challenge by physostigmine, a cholinesterase inhibitor. These results are consistent with the cholinergic hypothesis of depression. They imply that the NTRK1 E492K mutation, impairs cholinergic neurotransmission, and may convey susceptibility to bipolar disorder.
Collapse
Affiliation(s)
- Kazuo Nakajima
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Alannah Miranda
- Department of Psychiatry, University of California San Diego, San Diego, USA
| | - David W Craig
- Department of Genetics, University of Southern California, Los Angeles, USA
| | - Tatyana Shekhtman
- Department of Psychiatry, University of California San Diego, San Diego, USA
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Anthony Bleyer
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Szabolcs Szelinger
- Neurogenomics Division, Translational Genomics Research Institute, Arizona, USA
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, Saitama, Japan.
- Department of Psychiatry, Juntendo University, Tokyo, Japan.
| | - John R Kelsoe
- Department of Psychiatry, University of California San Diego, San Diego, USA.
- Institute for Genomic Medicine, University of California San Diego, San Diego, USA.
| |
Collapse
|
13
|
Hu VW, Bi C. Phenotypic Subtyping and Re-analyses of Existing Transcriptomic Data from Autistic Probands in Simplex Families Reveal Differentially Expressed and ASD Trait-Associated Genes. Front Neurol 2020; 11:578972. [PMID: 33281715 PMCID: PMC7689346 DOI: 10.3389/fneur.2020.578972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorder (ASD) describes a collection of neurodevelopmental disorders characterized by core symptoms that include social communication deficits and repetitive, stereotyped behaviors often coupled with restricted interests. Primary challenges to understanding and treating ASD are the genetic and phenotypic heterogeneity of cases that complicates all omics analyses as well as a lack of information on relationships among genes, pathways, and autistic traits. In this study, we re-analyze existing transcriptomic data from simplex families by subtyping individuals with ASD according to multivariate cluster analyses of clinical ADI-R scores that encompass a broad range of behavioral symptoms. We also correlate multiple ASD traits, such as deficits in verbal and non-verbal communication, play and social skills, ritualistic behaviors, and savant skills, with expression profiles using Weighted Gene Correlation Network Analyses (WGCNA). Our results show that subtyping greatly enhances the ability to identify differentially expressed genes involved in specific canonical pathways and biological functions associated with ASD within each phenotypic subgroup. Moreover, using WGCNA, we identify gene modules that correlate significantly with specific ASD traits. Network prediction analyses of the genes in these modules reveal canonical pathways as well as neurological functions and disorders relevant to the pathobiology of ASD. Finally, we compare the WGCNA-derived data on autistic traits in simplex families with analogous data from multiplex families using transcriptomic data from our previous studies. The comparison reveals overlapping trait-associated pathways as well as upstream regulators of the module-associated genes that may serve as useful targets for a precision medicine approach to ASD.
Collapse
Affiliation(s)
- Valerie W Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Chongfeng Bi
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
14
|
Nuthikattu S, Milenkovic D, Rutledge JC, Villablanca AC. Lipotoxic Injury Differentially Regulates Brain Microvascular Gene Expression in Male Mice. Nutrients 2020; 12:E1771. [PMID: 32545722 PMCID: PMC7353447 DOI: 10.3390/nu12061771] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022] Open
Abstract
The Western diet (WD) and hyperlipidemia are risk factors for vascular disease, dementia, and cognitive impairment. However, the molecular mechanisms are poorly understood. This pilot study investigated the genomic pathways by which the WD and hyperlipidemia regulate gene expression in brain microvessels. Five-week-old C57BL/6J wild type (WT) control and low-density lipoprotein receptor deficient (LDL-R-/-) male mice were fed the WD for eight weeks. Differential gene expression, gene networks and pathways, transcription factors, and non-protein coding RNAs were evaluated by a genome-wide microarray and bioinformatics analysis of laser-captured hippocampal microvessels. The WD resulted in the differential expression of 1972 genes. Much of the differentially expressed gene (DEG) was attributable to the differential regulation of cell signaling proteins and their transcription factors, approximately 4% was attributable to the differential expression of miRNAs, and 10% was due to other non-protein coding RNAs, primarily long non-coding RNAs (lncRNAs) and small nucleolar RNAs (snoRNAs) not previously described to be modified by the WD. Lipotoxic injury resulted in complex and multilevel molecular regulation of the hippocampal microvasculature involving transcriptional and post-transcriptional regulation and may provide a molecular basis for a better understanding of hyperlipidemia-associated dementia risk.
Collapse
Affiliation(s)
- Saivageethi Nuthikattu
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, One Shields Ave., The Grove, Rm 1159, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Dragan Milenkovic
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, One Shields Ave., The Grove, Rm 1159, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
- INRA, UNH, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - John C. Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, One Shields Ave., The Grove, Rm 1159, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Amparo C. Villablanca
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, One Shields Ave., The Grove, Rm 1159, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| |
Collapse
|
15
|
Zhu LL, Shen JD, Wang BY, Lu SF, Ming B, Xu EP, Li YC, Fang XY. Depressive-like behaviors are induced by chronic liver injury in male and female mice. Neurosci Lett 2020; 718:134750. [PMID: 31926175 DOI: 10.1016/j.neulet.2020.134750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/26/2019] [Accepted: 01/07/2020] [Indexed: 10/25/2022]
Abstract
Depression is a highly prevalent mental disease and increasingly become a global public health problem. Recent studies have shown that the dysfunction of liver was associated with depression. However, the previous studies have not been fully explained the relationship between depression and liver injury. The present study was aimed to investigate whether chronic liver injury could induce depressive-like behavior. Chronic liver injury was induced by intraperitoneal injection of carbon (CCl4), D-galactosamine (D-GalN) and thioacetamide (TAA), respectively. And the results showed that the serum activities of ALT in CCl4, D-GalN and TAA groups were significantly increased in both male and female mice compared with the control group, while the activities of AST increased only in CCl4 group. Meanwhile, H&E staining showed that CCl4, D-GalN and TAA induced hepatocytes injury in both male and female mice. Moreover, the sucrose preference was significantly decreased and the immobility time in forced swimming test and tail suspension test were significantly prolonged in CCl4 and D-GalN group compared with control group. Our findings demonstrated that chronic liver injury induced by CCl4 and D-GalN could induce depressive-like behaviors in mice.
Collapse
Affiliation(s)
- Lei-Lei Zhu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Ji-Duo Shen
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Bao-Ying Wang
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Scientific Research and Experiment Center, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Shuai-Fei Lu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Bai Ming
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Scientific Research and Experiment Center, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Er-Ping Xu
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Scientific Research and Experiment Center, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Yu-Cheng Li
- Henan Key Laboratory for Modern Research on Zhongjing's Herbal Formulae, Scientific Research and Experiment Center, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Xiao-Yan Fang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| |
Collapse
|
16
|
Yamanishi K, Hashimoto T, Miyauchi M, Mukai K, Ikubo K, Uwa N, Watanabe Y, Ikawa T, Okuzaki D, Okamura H, Yamanishi H, Matsunaga H. Analysis of genes linked to depressive-like behaviors in interleukin-18-deficient mice: Gene expression profiles in the brain. Biomed Rep 2019; 12:3-10. [PMID: 31839943 DOI: 10.3892/br.2019.1259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/23/2019] [Indexed: 01/21/2023] Open
Abstract
Interleukin (IL)-18 is an interferon γ-inducing inflammatory cytokine associated with function of the immune system and other physiological functions. IL-18-deficient (Il18 -/-) mice exhibit obesity, dyslipidemia, non-alcoholic steatohepatitis and depressive-like behavioral changes. Therefore, IL-18 has a number of important roles associated with immunity, energy homeostasis and psychiatric conditions. In the present study, gene expression in the brains of Il18 -/- mice was analyzed to identify genes associated with the depressive-like behaviors and other impairments displayed by Il18 -/- mice. Using whole genome microarray analysis, gene expression patterns in the brains of Il18 +/+ and Il18 -/- mice at 6 and 12 weeks of age were examined and compared. Subsequently, genes were categorized using Ingenuity® Pathway Analysis (IPA). At 12 weeks of age, 2,805 genes were identified using microarray analysis. Genes related to 'Major depression' and 'Depressive disorders' were identified by IPA core analysis, and 13 genes associated with depression were isolated. Among these genes, fibroblast growth factor receptor 1 (Fgfr1); protein tyrosine phosphatase, non-receptor type 1 (Ptpn1); and urocortin 3 (Ucn3) were classed as depression-inducing and the other genes were considered depression-suppressing genes. Subsequently, the interactions between the microarray results at 6 weeks of age and the above three depression-inducing genes were analyzed to search for effector genes of depression at 12 weeks of age. This analysis identified cyclin D1 (Ccnd1) and NADPH oxidase 4 (Nox4). The microarray analysis results were correlated with the results of reverse transcription-quantitative PCR (RT-qPCR). Overall, the results suggest that Fgfr1, Ptpn1 and Ucn3 may be involved in depression-like changes and Ccnd1 and Nox4 regulate these three genes in IL-18-deficient mice.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Psychoimmunology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan.,Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Takuya Hashimoto
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Masahiro Miyauchi
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Keiichiro Mukai
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Kaoru Ikubo
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Noriko Uwa
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| | - Takashi Ikawa
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Haruki Okamura
- Department of Psychoimmunology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan.,Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| | - Hiromichi Yamanishi
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| | - Hisato Matsunaga
- Department of Psychoimmunology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan.,Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
17
|
Kir6.1 Heterozygous Mice Exhibit Aberrant Amygdala-Dependent Cued Fear Memory. Mol Neurobiol 2019; 57:1622-1635. [PMID: 31808063 DOI: 10.1007/s12035-019-01840-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/18/2019] [Indexed: 10/25/2022]
Abstract
ATP-sensitive K+ (KATP) channels are predominantly expressed in the brain and consist of four identical inward-rectifier potassium ion channel subunits (Kir6.1 or Kir6.2) and four identical high-affinity sulfonylurea receptor subunits (SUR1, SUR2A, or SUR2B). We previously observed that chronic corticosterone-treated (CORT) mice exhibited enhanced anxiety-like behaviors and cued fear memory. In the present study, the protein and mRNA expression levels of Kir6.1, but not Kir6.2, were decreased in the lateral amygdala (LA) of CORT mice. Heterozygous Kir6.1-null (Kir6.1+/-) mice also showed enhanced tone (cued) fear memory and long-term potentiation (LTP) in the cortico-LA pathway compared to those in wild-type mice. However, LTP was not enhanced in the hippocampal CA1 regions of Kir6.1+/- mice. Consistent with increased cued fear memory, both Ca2+/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) activities were significantly elevated in the LAs of Kir6.1+/- mice after tone stimulation. Our results indicate that increased CaMKII and ERK activities may induce LTP in the LA in Kir6.1+/- mice, leading to aberrant cued fear memory. The changes in neural plasticity in the LA of Kir6.1+/- mice were associated with anxiety-like behaviors and may be related to the pathogenic mechanisms of anxiety disorders in human patients.
Collapse
|
18
|
Interleukin-18-deficient mice develop hippocampal abnormalities related to possible depressive-like behaviors. Neuroscience 2019; 408:147-160. [PMID: 30981863 DOI: 10.1016/j.neuroscience.2019.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/31/2022]
Abstract
Interleukin-18 (IL-18) is an inflammatory cytokine linked to major depressive disorder (MDD). MDD is closely related to metabolic disorders, such as diabetes mellitus (DM) and obesity. Moreover, DM is associated with cognitive impairment and promotes apoptosis of hippocampal cells by activating pro-apoptotic and inhibiting anti-apoptotic factors. IL-18-deficient (Il18-/-) mice are obese and have DM. Therefore, we hypothesized a close relationship between IL-18 and death of hippocampal cells, affecting neurogenesis related to behavioral changes such as MDD. Il18-/- male mice were generated on the C57Bl/6 background and Il18+/+ mice were used as controls. Behavioral, histopathological, and molecular responses, as well as responses to intracerebral recombinant IL-18 administration, were examined. Compared with Il18+/+ mice, Il18-/- mice had impaired learning and memory and exhibited lower motivation. In the Il18-/- mice, degenerated mitochondria were detected in synaptic terminals in the molecular layer, the polymorphic layer, and in mossy fibers in the dentate gyrus, suggesting mitochondrial abnormalities. Because of the degeneration of mitochondria in the dentate gyrus, in which pro-apoptotic molecules were upregulated and anti-apoptotic factors were decreased, apoptosis inducers were not cleaved, indicating inhibition of apoptosis. In addition, neurogenesis in the dentate gyrus and the maturity of neuronal cells were decreased in the Il18-/- mice, while intracerebral administration of recombinant IL-18 promoted significant recovery of neurogenesis. Our findings suggested that IL-18 was indispensable for mitochondrial homeostasis, sustaining clearance of degenerative neural cells, and supporting neurogenesis, normal neuronal maturation and hippocampal function.
Collapse
|
19
|
Walker DM, Cates HM, Loh YHE, Purushothaman I, Ramakrishnan A, Cahill KM, Lardner CK, Godino A, Kronman HG, Rabkin J, Lorsch ZS, Mews P, Doyle MA, Feng J, Labonté B, Koo JW, Bagot RC, Logan RW, Seney ML, Calipari ES, Shen L, Nestler EJ. Cocaine Self-administration Alters Transcriptome-wide Responses in the Brain's Reward Circuitry. Biol Psychiatry 2018; 84:867-880. [PMID: 29861096 PMCID: PMC6202276 DOI: 10.1016/j.biopsych.2018.04.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/15/2018] [Accepted: 04/17/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Global changes in gene expression underlying circuit and behavioral dysregulation associated with cocaine addiction remain incompletely understood. Here, we show how a history of cocaine self-administration (SA) reprograms transcriptome-wide responses throughout the brain's reward circuitry at baseline and in response to context and/or cocaine re-exposure after prolonged withdrawal (WD). METHODS We assigned male mice to one of six groups: saline/cocaine SA + 24-hour WD or saline/cocaine SA + 30-day WD + an acute saline/cocaine challenge within the previous drug-paired context. RNA sequencing was conducted on six interconnected brain reward regions. Using pattern analysis of gene expression and factor analysis of behavior, we identified genes that are strongly associated with addiction-related behaviors and uniquely altered by a history of cocaine SA. We then identified potential upstream regulators of these genes. RESULTS We focused on three patterns of gene expression that reflect responses to 1) acute cocaine, 2) context re-exposure, and 3) drug + context re-exposure. These patterns revealed region-specific regulation of gene expression. Further analysis revealed that each of these gene expression patterns correlated with an addiction index-a composite score of several addiction-like behaviors during cocaine SA-in a region-specific manner. Cyclic adenosine monophosphate response element binding protein and nuclear receptor families were identified as key upstream regulators of genes associated with such behaviors. CONCLUSIONS This comprehensive picture of transcriptome-wide regulation in the brain's reward circuitry by cocaine SA and prolonged WD provides new insight into the molecular basis of cocaine addiction, which will guide future studies of the key molecular pathways involved.
Collapse
Affiliation(s)
- Deena M Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hannah M Cates
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yong-Hwee E Loh
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aarthi Ramakrishnan
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kelly M Cahill
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Casey K Lardner
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Arthur Godino
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hope G Kronman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jacqui Rabkin
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zachary S Lorsch
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Philipp Mews
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marie A Doyle
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jian Feng
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Benoit Labonté
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ja Wook Koo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rosemary C Bagot
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ryan W Logan
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
20
|
Yamanishi K, Maeda S, Kuwahara-Otani S, Hashimoto T, Ikubo K, Mukai K, Nakasho K, Gamachi N, El-Darawish Y, Li W, Okuzaki D, Watanabe Y, Yamanishi H, Okamura H, Matsunaga H. Deficiency in interleukin-18 promotes differentiation of brown adipose tissue resulting in fat accumulation despite dyslipidemia. J Transl Med 2018; 16:314. [PMID: 30453990 PMCID: PMC6245626 DOI: 10.1186/s12967-018-1684-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/09/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The cytokine, interleukin-18 (IL-18), was originally identified as an interferon-γ-inducing proinflammatory factor; however, there is increasing evidence suggesting that it has non-immunological effects on physiological functions. We have previously investigated the potential pathophysiological relationship between IL-18 and dyslipidemia, non-alcoholic fatty liver disease and non-alcoholic steatohepatitis, which were mediated by lipid energy imbalance. Therefore, herein we focused on brown adipocytes (BAs) and brown adipose tissue (BAT) related to energy consumption as non-shivering thermogenesis. METHODS Il18-/- male mice were generated on the C57Bl/6 background, and littermate C57Bl/6 Il18+/+ male mice were used as controls. To reveal the direct effect of IL-18, primary cell cultures derived from both mice were established. Moreover, for molecular analysis, microarray, quantitative reverse transcription PCR and western blotting were performed using 6 and 12 weeks old mice. To evaluate the short- and long-term effects of IL-18 on BAT, recombinant IL-18 was administered for 2 and 12 weeks, respectively. RESULTS Compared with Il18+/+ mice, BAT of Il18-/- mice showed earlier differentiation and lipid accumulation. To examine the direct effect of IL-18 on BAT, BA cell cultures were established. Myogenic factor 5-expressing adipose precursor cells were extracted from Il18+/+ and Il18-/- mice. PR domain containing 16 (PRDM16), a differentiation inducer, was strongly expressed in Il18-/- BAs, and uncoupling protein 1, a thermogenic and differentiation marker, was upregulated, resulting in the promotion of BA differentiation. Moreover, PRDM16-dependent and independent molecules related to BAT function, such as fibroblast growth factor 21, were activated. These findings were confirmed by comparing Il18+/+ and Il18-/- mice at 6 and 12 weeks of age. Additional analyses of the molecular mechanisms influencing the 'Quantity of adipocytes' identified three associated genes, apolipoprotein C3 (Apoc3), insulin-induced gene 1 (Insig1) and vitamin D (1,25-dihydroxyvitamin D3) receptor (Vdr). Intravenous administration of IL-18 not only significantly improved the expression of some of these genes, but it also significantly decreased the adipocytes' size. CONCLUSIONS This study demonstrated the critical function of IL-18 in differentiation and lipid metabolism in BAs. Furthermore, IL-18 may contribute to novel treatments by improving the energy imbalance.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Seishi Maeda
- Department of Anatomy and Cell Biology, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Sachi Kuwahara-Otani
- Department of Anatomy and Cell Biology, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takuya Hashimoto
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kaoru Ikubo
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Keiichiro Mukai
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Naomi Gamachi
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, 565-0871, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka, 573-0122, Japan
| | - Hiromichi Yamanishi
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka, 573-0122, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
21
|
Yamanishi K, Mukai K, Hashimoto T, Ikubo K, Nakasho K, El-Darawish Y, Li W, Okuzaki D, Watanabe Y, Hayakawa T, Nojima H, Yamanishi H, Okamura H, Matsunaga H. Physiological and molecular effects of interleukin-18 administration on the mouse kidney. J Transl Med 2018. [PMID: 29514661 PMCID: PMC5842592 DOI: 10.1186/s12967-018-1426-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The cytokine interleukin-18 was originally identified as an interferon-γ-inducing proinflammatory factor; however, there is increasing evidence to suggest that it has non-immunological effects on physiological functions. We previously investigated the potential pathophysiological relationship between interleukin-18 and dyslipidemia, non-alcoholic fatty liver disease, and non-alcoholic steatohepatitis, and suggested interleukin-18 as a possible novel treatment for not only these diseases but also for cancer immunotherapy. Before clinical application, the effects of interleukin-18 on the kidney need to be determined. In the current study, we examined the kidney of interleukin-18 knockout (Il18-/-) mice and the effects of interleukin-18 on the kidney following intravenous administration of recombinant interleukin-18. METHODS Il18-/- male mice were generated on the C57Bl/6 background and littermate C57Bl/6 Il18+/+ male mice were used as controls. To assess kidney damage, serum creatinine and blood urea nitrogen levels were measured and histopathological analysis was performed. For molecular analysis, microarray and quantitative reverse transcription PCR was performed using mice 6 and 12 weeks old. To evaluate the short- and long-term effects of interleukin-18 on the kidney, recombinant interleukin-18 was administered for 2 and 12 weeks, respectively. RESULTS Compared with Il18+/+ mice, Il18-/- mice developed kidney failure in their youth-6 weeks of age, but the condition was observed to improve as the mice aged, even though dyslipidemia, arteriosclerosis, and higher insulin resistance occurred. Analyses of potential molecular mechanisms involved in the onset of early kidney failure in Il18-/- mice identified a number of associated genes, such as Itgam, Nov, and Ppard. Intravenous administration of recombinant interleukin-18 over both the short and long term showed no effects on the kidney despite significant improvement in metabolic diseases. CONCLUSIONS Short- and long-term administration of interleukin-18 appeared to have no adverse effects on the kidney in these mice, suggesting that administration may be a safe and novel treatment for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Keiichiro Mukai
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takuya Hashimoto
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kaoru Ikubo
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Daisuke Okuzaki
- DNA-Chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, 565-0871, Japan.,Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, 565-0871, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka, 573-0122, Japan
| | - Tetsu Hayakawa
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hiroshi Nojima
- DNA-Chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, 565-0871, Japan.,Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, 565-0871, Japan
| | - Hiromichi Yamanishi
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka, 573-0122, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
22
|
Abstract
Gut microbiomes may have a significant impact on mood and cognition, which is leading experts towards a new frontier in neuroscience. Studies have shown that increase in the amount of good bacteria in the gut can curb inflammation and cortisol level, reduces symptoms of depression and anxiety, lowers stress reactivity, improves memory and even lessens neuroticism and social anxiety. This shows that, probably the beneficial gut bacteria or probiotics function mechanistically as delivery vehicles for neuroactive compounds. Thus, a psychobiotic is a live organism, when ingested in adequate amounts, produces a health benefit in patients suffering from psychiatric illness. Study of these novel class of probiotics may open up the possibility of rearrangement of intestinal microbiota for effective management of various psychiatric disorders.
Collapse
Affiliation(s)
- Snigdha Misra
- a Department of Nutrition and Dietetics, School of Health Sciences , International Medical University , Kuala Lumpur , Malaysia
| | - Debapriya Mohanty
- b Department of Microbiology , Centre for Post Graduate Studies, Orissa University of Agriculture and Technology , Bhubaneswar , Odisha , India
| |
Collapse
|
23
|
Arriazu E, Ge X, Leung TM, Magdaleno F, Lopategi A, Lu Y, Kitamura N, Urtasun R, Theise N, Antoine DJ, Nieto N. Signalling via the osteopontin and high mobility group box-1 axis drives the fibrogenic response to liver injury. Gut 2017; 66:1123-1137. [PMID: 26818617 PMCID: PMC5532463 DOI: 10.1136/gutjnl-2015-310752] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/17/2015] [Accepted: 12/28/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Liver fibrosis is associated with significant collagen-I deposition largely produced by activated hepatic stellate cells (HSCs); yet, the link between hepatocyte damage and the HSC profibrogenic response remains unclear. Here we show significant induction of osteopontin (OPN) and high-mobility group box-1 (HMGB1) in liver fibrosis. Since OPN was identified as upstream of HMGB1, we hypothesised that OPN could participate in the pathogenesis of liver fibrosis by increasing HMGB1 to upregulate collagen-I expression. DESIGN AND RESULTS Patients with long-term hepatitis C virus (HCV) progressing in disease stage displayed enhanced hepatic OPN and HMGB1 immunostaining, which correlated with fibrosis stage, whereas it remained similar in non-progressors. Hepatocyte cytoplasmic OPN and HMGB1 expression was significant while loss of nuclear HMGB1 occurred in patients with HCV-induced fibrosis compared with healthy explants. Well-established liver fibrosis along with marked induction of HMGB1 occurred in CCl4-injected OpnHep transgenic yet it was less in wild type and almost absent in Opn-/- mice. Hmgb1 ablation in hepatocytes (Hmgb1ΔHep) protected mice from CCl4-induced liver fibrosis. Coculture with hepatocytes that secrete OPN plus HMGB1 and challenge with recombinant OPN (rOPN) or HMGB1 (rHMGB1) enhanced collagen-I expression in HSCs, which was blunted by neutralising antibodies (Abs) and by Opn or Hmgb1 ablation. rOPN induced acetylation of HMGB1 in HSCs due to increased NADPH oxidase activity and the associated decrease in histone deacetylases 1/2 leading to upregulation of collagen-I. Last, rHMGB1 signalled via receptor for advanced glycation end-products and activated the PI3K-pAkt1/2/3 pathway to upregulate collagen-I. CONCLUSIONS During liver fibrosis, the increase in OPN induces HMGB1, which acts as a downstream alarmin driving collagen-I synthesis in HSCs.
Collapse
Affiliation(s)
- Elena Arriazu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Xiaodong Ge
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tung-Ming Leung
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fernando Magdaleno
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Aritz Lopategi
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yongke Lu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Naoto Kitamura
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Raquel Urtasun
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Neil Theise
- Division of Digestive Diseases, Mount Sinai Beth Israel Medical Center, New York, New York, USA
| | - Daniel J Antoine
- Medical Research Council Centre for Drug Safety Science, Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Natalia Nieto
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
24
|
Ikubo K, Yamanishi K, Doe N, Hashimoto T, Sumida M, Watanabe Y, El-Darawish Y, Li W, Okamura H, Yamanishi H, Matsunaga H. Molecular analysis of the mouse brain exposed to chronic mild stress: The influence of hepatocyte nuclear factor 4α on physiological homeostasis. Mol Med Rep 2017; 16:301-309. [PMID: 28498421 DOI: 10.3892/mmr.2017.6577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/27/2017] [Indexed: 11/05/2022] Open
Abstract
Major depressive disorder (MDD) is a prevalent disorder that causes considerable disability in social functioning and is a risk factor for physical diseases. Recent clinical reports have demonstrated a marked association between MDD and physiological dyshomeostasis induced by metabolic disorders, including diabetes, hormone abnormalities and autoimmune diseases. The authors of the present study have previously analyzed comparative gene expression profiles in the prefrontal cortex (PFC) of a chronic mild stress (CMS) animal model of MDD. Hepatocyte nuclear factor 4α (Hnf4α) was identified as a central regulator that exerted significant influence on genes associated with physiological homeostasis. The aim of the present study was to investigate: i) the molecular mechanism of the depressive state in the PFC, and ii) the involvement of genes extracted from the comparative gene expression profiles, particularly those applicable to MDD in clinical practice. Core analysis of the previous PFC microarray results was performed using Ingenuity Pathway Analysis (IPA). Subsequently, IPA was used to search for molecules that are regulated by Hnf4α, and exist in the PFC and serum. From the core analysis, 5 genes that are associated with cell death and are expressed in the cortex were selected. Four of the extracted genes, insulin‑like growth factor 1, transthyretin, serpin family A member 3 and plasminogen, were markedly affected by Hnf4α. S100 calcium‑binding protein A9 (S100a9) and α2-HS-glycoprotein (Ahsg) were also chosen as they exist in serum and are also affected by Hnf4α. A significant group difference in the expression of these two genes was detected in the PFC, thalamus and hippocampus. The protein levels of AHSG and S100A9 in the PFC and hippocampus of the CMS group increased significantly when compared with the control group. These findings support the close association of Hnf4α (through genes such as S100a9 and Ahsg) with the development of various diseases induced by deregulation of physiological homeostasis during the progression of MDD.
Collapse
Affiliation(s)
- Kaoru Ikubo
- Department of Neuropsychiatry, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Nobutaka Doe
- Laboratory of Neurogenesis and CNS Repair, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Takuya Hashimoto
- Department of Neuropsychiatry, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Miho Sumida
- Department of Psychology, Hyōgo University of Health Sciences, Kobe, Hyōgo 650‑8530, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573‑0122, Japan
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell Therapy, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| | - Hiromichi Yamanishi
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573‑0122, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyōgo College of Medicine, Nishinomiya, Hyōgo 663‑8501, Japan
| |
Collapse
|
25
|
Yamanishi K, Maeda S, Kuwahara-Otani S, Watanabe Y, Yoshida M, Ikubo K, Okuzaki D, El-Darawish Y, Li W, Nakasho K, Nojima H, Yamanishi H, Hayakawa T, Okamura H, Matsunaga H. Interleukin-18-deficient mice develop dyslipidemia resulting in nonalcoholic fatty liver disease and steatohepatitis. Transl Res 2016; 173:101-114.e7. [PMID: 27063959 DOI: 10.1016/j.trsl.2016.03.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/23/2015] [Accepted: 03/12/2016] [Indexed: 12/11/2022]
Abstract
We investigated potential pathophysiological relationships between interleukin 18 (IL-18) and dyslipidemia, nonalcoholic fatty liver disease (NAFLD) or nonalcoholic steatohepatitis (NASH). Compared with Il18(+/+) mice, IL-18 knockout (Il18(-/-)) mice developed hypercholesterolemia and hyper-high-density-lipoprotein-cholesterolemia as well as hypertriglyceridemia as they aged, and these disorders occurred before the manifestation of obesity and might cause secondary NASH. The analyses of molecular mechanisms involved in the onset of dyslipidemia, NAFLD, and NASH in Il18(-/-) mice identified a number of genes associated with these metabolic diseases. In addition, molecules related to circadian rhythm might affect these extracted genes. The intravenous administration of recombinant IL-18 significantly improved dyslipidemia, inhibited the body weight gain of Il18(+/+) mice, and prevented the onset of NASH. The expression of genes related to these dysfunctions was also affected by recombinant IL-18 administration. In conclusion, this study demonstrated the critical function of IL-18 in lipid metabolism and these findings might contribute to the progress of novel treatments for NAFLD or NASH.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan; Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka 573-0122, Japan
| | - Seishi Maeda
- Department of Anatomy and Cell Biology, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Sachi Kuwahara-Otani
- Department of Anatomy and Cell Biology, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka 573-0122, Japan
| | - Momoko Yoshida
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka 573-0122, Japan; Department of Genome Informatics, Osaka University, 3-1, Yamadaoka, Suita 565-0871, Japan
| | - Kaoru Ikubo
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Daisuke Okuzaki
- DNA-Chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1, Yamadaoka, Suita 565-0871, Japan; Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3-1, Yamadaoka, Suita 565-0871, Japan
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Wen Li
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroshi Nojima
- DNA-Chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1, Yamadaoka, Suita 565-0871, Japan; Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3-1, Yamadaoka, Suita 565-0871, Japan
| | - Hiromichi Yamanishi
- Hirakata General Hospital for Developmental Disorders, 2-1-1 Tsudahigashi, Hirakata, Osaka 573-0122, Japan
| | - Tetsu Hayakawa
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1, Mukogawa, Nishinomiya, Hyogo 663-8501, Japan.
| |
Collapse
|
26
|
Quantitative proteomics analysis of the liver reveals immune regulation and lipid metabolism dysregulation in a mouse model of depression. Behav Brain Res 2016; 311:330-339. [PMID: 27247144 DOI: 10.1016/j.bbr.2016.05.057] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/20/2022]
Abstract
Major depressive disorder (MDD) is a highly prevalent and debilitating mental illness with substantial impairments in quality of life and functioning. However, the pathophysiology of major depression remains poorly understood. Combining the brain and body should provide a comprehensive understanding of the etiology of MDD. As the largest internal organ of the human body, the liver has an important function, yet no proteomic study has assessed liver protein expression in a preclinical model of depression. Using the chronic unpredictable mild stress (CUMS) mouse model of depression, differential protein expression between CUMS and control (CON) mice was examined in the liver proteome using isobaric tag for relative and absolute quantitation (iTRAQ) coupled with tandem mass spectrometry. More than 4000 proteins were identified and 66 most significantly differentiated proteins were used for further bioinformatic analysis. According to the ingenuity pathway analysis (IPA), we found that proteins related to the inflammation response, immune regulation, lipid metabolism and NFκB signaling network were altered by CUMS. Moreover, four proteins closely associated with these processes, hemopexin, haptoglobin, cytochrome P450 2A4 (CYP2A4) and bile salt sulfotransferase 1 (SULT2A1), were validated by western blotting. In conclusion, we report, for the first time, the liver protein expression profile in the CUMS mouse model of depression. Our findings provide novel insight (liver-brain axis) into the multifaceted mechanisms of major depressive disorder.
Collapse
|
27
|
Stankiewicz AM, Goscik J, Dyr W, Juszczak GR, Ryglewicz D, Swiergiel AH, Wieczorek M, Stefanski R. Novel candidate genes for alcoholism--transcriptomic analysis of prefrontal medial cortex, hippocampus and nucleus accumbens of Warsaw alcohol-preferring and non-preferring rats. Pharmacol Biochem Behav 2015; 139:27-38. [PMID: 26455281 DOI: 10.1016/j.pbb.2015.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/06/2015] [Accepted: 10/06/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Animal models provide opportunity to study neurobiological aspects of human alcoholism. Changes in gene expression have been implicated in mediating brain functions, including reward system and addiction. The current study aimed to identify genes that may underlie differential ethanol preference in Warsaw High Preferring (WHP) and Warsaw Low Preferring (WLP) rats. METHODS Microarray analysis comparing gene expression in nucleus accumbens (NAc), hippocampus (HP) and medial prefrontal cortex (mPFC) was performed in male WHP and WLP rats bred for differences in ethanol preference. RESULTS Differential and stable between biological repeats expression of 345, 254 and 129 transcripts in NAc, HP and mPFC was detected. Identified genes and processes included known mediators of ethanol response (Mx2, Fam111a, Itpr1, Gabra4, Agtr1a, LTP/LTD, renin-angiotensin signaling pathway), toxicity (Sult1c2a, Ces1, inflammatory response), as well as genes involved in regulation of important addiction-related brain systems such as dopamine, tachykinin or acetylcholine (Gng7, Tac4, Slc5a7). CONCLUSIONS The identified candidate genes may underlie differential ethanol preference in an animal model of alcoholism. COMMENT Names of genes are written in italics, while names of proteins are written in standard font. Names of human genes/proteins are written in all capital letters. Names of rodent genes/proteins are written in capital letter followed by small letters.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Animal Behaviour, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Joanna Goscik
- Software Department, Faculty of Computer Science, Bialystok University of Technology, 15-351 Bialystok, Poland
| | - Wanda Dyr
- Department of Pharmacology and Physiology of the Nervous System, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Grzegorz R Juszczak
- Department of Animal Behaviour, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland
| | - Danuta Ryglewicz
- First Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Artur H Swiergiel
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland; Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA71130, USA.
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Roman Stefanski
- Department of Pharmacology and Physiology of the Nervous System, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| |
Collapse
|
28
|
Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, Jafari P, Akbari H, Taghizadeh M, Memarzadeh MR, Asemi Z, Esmaillzadeh A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2015; 32:315-20. [PMID: 26706022 DOI: 10.1016/j.nut.2015.09.003] [Citation(s) in RCA: 477] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/15/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We are aware of no study examining the effects of probiotic supplementation on symptoms of depression, metabolic profiles, serum high-sensitivity C-reactive protein (hs-CRP), and biomarkers of oxidative stress in patients with major depressive disorder (MDD). The present study was designed to determine the effects of probiotic intake on symptoms of depression and metabolic status in patients with MDD. METHODS This randomized, double-blind, placebo-controlled clinical trial included 40 patients with a diagnosis of MDD based on DSM-IV criteria whose age ranged between 20 and 55 y. Patients were randomly allocated into two groups to receive either probiotic supplements (n = 20) or placebo (n = 20) for 8 wk. Probiotic capsule consisted of three viable and freeze-dried strains: Lactobacillus acidophilus (2 × 10(9) CFU/g), Lactobacillus casei (2 × 10(9) CFU/g), and Bifidobacterium bifidum (2 × 10(9) CFU/g). Fasting blood samples were taken at the beginning and end of the trial to quantify the relevant variables. All participants provided three dietary records (two weekdays and one weekend) and three physical activity records during the intervention. RESULTS Dietary intake of study participants was not significantly different between the two groups. After 8 wk of intervention, patients who received probiotic supplements had significantly decreased Beck Depression Inventory total scores (-5.7 ± 6.4 vs. -1.5 ± 4.8, P = 0.001) compared with the placebo. In addition, significant decreases in serum insulin levels (-2.3 ± 4.1 vs. 2.6 ± 9.3 μIU/mL, P = 0.03), homeostasis model assessment of insulin resistance (-0.6 ± 1.2 vs. 0.6 ± 2.1, P = 0.03), and serum hs-CRP concentrations (-1138.7 ± 2274.9 vs. 188.4 ± 1455.5 ng/mL, P = 0.03) were observed after the probiotic supplementation compared with the placebo. Additionally, taking probiotics resulted in a significant rise in plasma total glutathione levels (1.8 ± 83.1 vs. -106.8 ± 190.7 μmol/L, P = 0.02) compared with the placebo. We did not find any significant change in fasting plasma glucose, homeostatic model assessment of beta cell function, quantitative insulin sensitivity check index, lipid profiles, and total antioxidant capacity levels. CONCLUSIONS Probiotic administration in patients with MDD for 8 wk had beneficial effects on Beck Depression Inventory, insulin, homeostasis model assessment of insulin resistance, hs-CRP concentrations, and glutathione concentrations, but did not influence fasting plasma glucose, homeostatic model assessment of beta cell function, quantitative insulin sensitivity check index, lipid profiles, and total antioxidant capacity levels.
Collapse
Affiliation(s)
- Ghodarz Akkasheh
- Department of Psychiatry, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Kashani-Poor
- Department of Psychiatry, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Tajabadi-Ebrahimi
- Science Department, Science Faculty, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | - Parvaneh Jafari
- Department of Microbiology, Science Faculty, Arak Branch, Islamic Azad University, Arak, Iran
| | - Hossein Akbari
- Department of Biostatistics, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
| | - Ahmad Esmaillzadeh
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Biological basis of “depression with liver-qi stagnation and spleen deficiency syndrome”: A digital gene expression profiling study. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2015. [DOI: 10.1016/j.jtcms.2016.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|