1
|
Fu Q, Luo Y, Li J, Li H, Liu X, Chen Z, Ni G, Wang T. Caerin 1.1 and 1.9 peptides halt B16 melanoma metastatic tumours via expanding cDC1 and reprogramming tumour macrophages. J Transl Med 2024; 22:973. [PMID: 39468595 PMCID: PMC11514859 DOI: 10.1186/s12967-024-05763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cancer immunotherapy, particularly immune checkpoint inhibitors (ICBs) such as anti-PD-1 antibodies, has revolutionised cancer treatment, although response rates vary among patients. Previous studies have demonstrated that caerin 1.1 and 1.9, host-defence peptides from the Australian tree frog, enhance the effectiveness of anti-PD-1 and therapeutic vaccines in a murine TC-1 model by activating tumour-associated macrophages intratumorally. METHODS We employed a murine B16 melanoma model to investigate the therapeutic potential of caerin 1.1 and 1.9 in combination with anti-CD47 and a therapeutic vaccine (triple therapy, TT). Tumour growth of caerin-injected primary tumours and distant metastatic tumours was assessed, and survival analysis conducted. Single-cell RNA sequencing (scRNAseq) of CD45+ cells isolated from distant tumours was performed to elucidate changes in the tumour microenvironment induced by TT. RESULTS The TT treatment significantly reduced tumour volumes on the treated side compared to untreated and control groups, with notable effects observed by Day 21. Survival analysis indicated extended survival in mice receiving TT, both on the treated and distant sides. scRNAseq revealed a notable expansion of conventional type 1 dendritic cells (cDC1s) and CD4+CD8+ T cells in the TT group. Tumour-associated macrophages in the TT group shifted toward a more immune-responsive M1 phenotype, with enhanced communication observed between cDC1s and CD8+ and CD4+CD25+ T cells. Additionally, TT downregulated M2-like macrophage marker genes, particularly in MHCIIhi and tissue-resident macrophages, suppressing Cd68 and Arg1 expression across all macrophage types. Differential gene expression analysis highlighted pathway alterations, including upregulation of oxidative phosphorylation and MYC target V1 in Arg1hi macrophages, and activation of pro-inflammatory pathways in MHCIIhi and tissue-resident macrophages. CONCLUSION Our findings suggest that caerin 1.1 and 1.9, combined with immunotherapy, effectively modulate the tumour microenvironment in primary and secondary tumours, leading to reduced tumour growth and enhanced systemic immunity. Further investigation into these mechanisms could pave the way for improved combination therapies in advanced melanoma treatment.
Collapse
Affiliation(s)
- Quanlan Fu
- Medical School of Guizhou University, Guiyang, Guizhou, 550000, China
| | - Yuandong Luo
- Medical School of Guizhou University, Guiyang, Guizhou, 550000, China
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Junjie Li
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Zhongao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, 528403, China
| | - Hejie Li
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, BC, QLD 4558, Australia
| | - Xiaosong Liu
- Medical School of Guizhou University, Guiyang, Guizhou, 550000, China
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Zhu Chen
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550004, China.
| | - Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China.
- The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Tianfang Wang
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, BC, QLD 4558, Australia.
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore, BC, QLD 4558, Australia.
| |
Collapse
|
2
|
Zhang Y, Li D, Han Y, Wu M, Zhang S, Ma H, Liu L, Ju X. Intraovarian injection of 3D-MSC-EVs-ECM gel significantly improved rat ovarian function after chemotherapy. Reprod Biol Endocrinol 2024; 22:125. [PMID: 39415205 PMCID: PMC11481453 DOI: 10.1186/s12958-024-01299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Restoring the function of the ovary is important for chemotherapy-induced ovarian failure (COF) patients. Stem cell and extracellular vesicles (EVs) therapy show promise but need further improvement. METHODS Human umbilical cord mesenchymal stem cells (hUC-MSCs) were primarily cultured and further three-dimensional (3D) cultured using an ultra-low attachment surface method. The expression levels of nutritional cytokines and immunomodulatory and stemness-related genes of 3D-cultured hUC-MSCs were analyzed. EVs were isolated by ultracentrifugation and characterized. Ovaries were decellularized with sodium dodecyl sulfate to obtain extracellular matrix (ECM). Lyophilized EVs from three-dimensional (2D) or 3D hUC-MSCs were mixed with ECM to prepare the 2D/3D-MSC-EVs-ECM gels. The therapeutic effect of the MSC-EVs-ECM gel on cyclophosphamide (CTX) -treated rats was analyzed through various tests. RNA sequencing was used to analyze the expression changes of genes before and after treatment. RESULTS After culturing in ultra-low attachment dishes, hUC-MSCs aggregated into spheroids and significantly upregulated the expression levels of immunomodulatory and stemness-related genes. The total EVs yield was also upregulated (5.6-fold) after 3D culture. The cell viability of CTX-treated ovarian granulosa cells (OGCs) was significantly rescued by coculture with the 3D-MSC-EVs-ECM gel. Hormones indicative of ovarian function, AMH, E2, and FSH, were recovered in both the CTX + 2D-MSC-EVs-ECM gel group and the CTX + 3D-MSC-EVs-ECM gel group, while the apoptosis-related protein Bax was significantly downregulated. The 3D-MSC-EVs-ECM gel was more effective than the 2D-MSC-EVs-ECM gel. Significantly differentially expressed genes, such as Hbb-b1, Gpd1, and Sirpa, were detected by RNA sequencing. Hbb-b1 was increased in the ovaries of CTX-treated rats, and this increase was attenuated by injecting the 2D/3D-MSC-EVs-ECM gel. Gpd1 was increased after CTX treatment, and this increase was reversed by the 3D-MSC-EVs-ECM gel. Sirpa was decreased in the ovaries of CTX-treated rats, and this decrease was attenuated by injecting the 3D-MSC-EVs-ECM gel. CONCLUSIONS Our study demonstrated that the 3D-MSC-EVs-ECM gel is an efficient strategy for the recovery of ovarian function in CTX-induced ovarian failure.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Department of Anesthesiology, Shanghai Jiaotong University First People's Hospital (Shanghai General Hospital), Shanghai, China
| | - Dong Li
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Yi Han
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Min Wu
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Shule Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Huixian Ma
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Linghong Liu
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan, Shandong Province, 250012, China.
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
3
|
Robertson KV, Rodriguez AS, Cartailler JP, Shrestha S, Schleh MW, Schroeder KR, Valenti AM, Kramer AT, Harrison FE, Hasty AH. Knockdown of microglial iron import gene, Slc11a2, worsens cognitive function and alters microglial transcriptional landscape in a sex-specific manner in the APP/PS1 model of Alzheimer's disease. J Neuroinflammation 2024; 21:238. [PMID: 39334471 PMCID: PMC11438269 DOI: 10.1186/s12974-024-03238-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] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Microglial cell iron load and inflammatory activation are significant hallmarks of late-stage Alzheimer's disease (AD). In vitro, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and excess iron can augment cellular inflammation, suggesting a feed-forward loop between iron import mechanisms and inflammatory signaling. However, it is not understood whether microglial iron import mechanisms directly contribute to inflammatory signaling and chronic disease in vivo. These studies determined the effects of microglial-specific knockdown of Slc11a2 on AD-related cognitive decline and microglial transcriptional phenotype. METHODS In vitro experiments and RT-qPCR were used to assess a role for DMT1 in amyloid-β-associated inflammation. To determine the effects of microglial Slc11a2 knockdown on AD-related phenotypes in vivo, triple-transgenic Cx3cr1Cre-ERT2;Slc11a2flfl;APP/PS1+or - mice were generated and administered corn oil or tamoxifen to induce knockdown at 5-6 months of age. Both sexes underwent behavioral analyses to assess cognition and memory (12-15 months of age). Hippocampal CD11b+ microglia were magnetically isolated from female mice (15-17 months) and bulk RNA-sequencing analysis was conducted. RESULTS DMT1 inhibition in vitro robustly decreased Aβ-induced inflammatory gene expression and cellular iron levels in conditions of excess iron. In vivo, Slc11a2KD APP/PS1 female, but not male, mice displayed a significant worsening of memory function in Morris water maze and a fear conditioning assay, along with significant hyperactivity compared to control WT and APP/PS1 mice. Hippocampal microglia from Slc11a2KD APP/PS1 females displayed significant increases in Enpp2, Ttr, and the iron-export gene, Slc40a1, compared to control APP/PS1 cells. Slc11a2KD cells from APP/PS1 females also exhibited decreased expression of markers associated with subsets of disease-associated microglia (DAMs), such as Apoe, Ctsb, Ly9, Csf1, and Hif1α. CONCLUSIONS This work suggests a sex-specific role for microglial iron import gene Slc11a2 in propagating behavioral and cognitive phenotypes in the APP/PS1 model of AD. These data also highlight an association between loss of a DAM-like phenotype in microglia and cognitive deficits in Slc11a2KD APP/PS1 female mice. Overall, this work illuminates an iron-related pathway in microglia that may serve a protective role during disease and offers insight into mechanisms behind disease-related sex differences.
Collapse
Affiliation(s)
- Katrina Volk Robertson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Alec S Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | | | - Shristi Shrestha
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, TN, USA
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Kyle R Schroeder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Arianna M Valenti
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Alec T Kramer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, 7465 Medical Research Building IV, 2213 Garland Avenue, Nashville, TN, 37232, USA.
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA.
- VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
4
|
Masson BA, Kiridena P, Lu D, Kleeman EA, Reisinger SN, Qin W, Davies WJ, Muralitharan RR, Jama HA, Antonacci S, Marques FZ, Gubert C, Hannan AJ. Depletion of the paternal gut microbiome alters sperm small RNAs and impacts offspring physiology and behavior in mice. Brain Behav Immun 2024; 123:290-305. [PMID: 39293692 DOI: 10.1016/j.bbi.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
The paternal environment prior to conception has been demonstrated to influence offspring physiology and behavior, with the sperm epigenome (including noncoding RNAs) proposed as a potential facilitator of non-genetic inheritance. Whilst the maternal gut microbiome has been established as an important influence on offspring development, the impact of the paternal gut microbiome on offspring development, health and behavior is largely unknown. Gut microbiota have major influences on immunity, and thus we hypothesized that they may be relevant to paternal immune activation (PIA) modulating epigenetic inheritance in mice. Therefore, male C57BL/6J mice (F0) were orally administered non-absorbable antibiotics via drinking water in order to substantially deplete their gut microbiome. Four weeks after administration of the antibiotics (gut microbiome depletion), F0 male mice were then mated with naïve female mice. The F1 offspring of the microbiome-depleted males had reduced body weight as well as altered gut morphology (shortened colon length). F1 females showed significant alterations in affective behaviors, including measures of anxiety and depressive-like behaviors, indicating altered development. Analysis of small noncoding RNAs in the sperm of F0 mice revealed that gut microbiome depletion is associated with differential expression of 8 different PIWI-interacting RNAs (piRNAs), each of which has the potential to modulate the expression of multiple downstream gene targets, and thus influence epigenetic inheritance and offspring development. This study demonstrates that the gut-germline axis influences sperm small RNA profiles and offspring physiology, with specific impacts on offspring affective and/or coping behaviors. These findings may have broader implications for other animal species with comparable gut microbiota, intergenerational epigenetics and developmental biology, including humans.
Collapse
Affiliation(s)
- Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pamudika Kiridena
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - William J Davies
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Simona Antonacci
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
5
|
Kwon D, Kim Y, Cho SH. Antidepressant Effects of Ginsenoside Rc on L-Alpha-Aminoadipic Acid-Induced Astrocytic Ablation and Neuroinflammation in Mice. Int J Mol Sci 2024; 25:9673. [PMID: 39273621 PMCID: PMC11396248 DOI: 10.3390/ijms25179673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Depression is a prevalent and debilitating mental disorder that affects millions worldwide. Current treatments, such as antidepressants targeting the serotonergic system, have limitations, including delayed onset of action and high rates of treatment resistance, necessitating novel therapeutic strategies. Ginsenoside Rc (G-Rc) has shown potential anti-inflammatory and neuroprotective effects, but its antidepressant properties remain unexplored. This study investigated the antidepressant effects of G-Rc in an L-alpha-aminoadipic acid (L-AAA)-induced mouse model of depression, which mimics the astrocytic pathology and neuroinflammation observed in major depressive disorder. Mice were administered G-Rc, vehicle, or imipramine orally after L-AAA injection into the prefrontal cortex. G-Rc significantly reduced the immobility time in forced swimming and tail suspension tests compared to vehicle treatment, with more pronounced effects than imipramine. It also attenuated the expression of pro-inflammatory cytokines (TNF-α, IL-6, TGF-β, lipocalin-2) and alleviated astrocytic degeneration, as indicated by increased GFAP and decreased IBA-1 levels. Additionally, G-Rc modulated apoptosis-related proteins, decreasing caspase-3 and increasing Bcl-2 levels compared to the L-AAA-treated group. These findings suggest that G-Rc exerts antidepressant effects by regulating neuroinflammation, astrocyte-microglia crosstalk, and apoptotic pathways in the prefrontal cortex, highlighting its potential as a novel therapeutic agent for depression.
Collapse
Affiliation(s)
- Dohyung Kwon
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yunna Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung-Hun Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Jellinger AL, Suthard RL, Yuan B, Surets M, Ruesch EA, Caban AJ, Liu S, Shpokayte M, Ramirez S. Chronic activation of a negative engram induces behavioral and cellular abnormalities. eLife 2024; 13:RP96281. [PMID: 38990919 PMCID: PMC11239178 DOI: 10.7554/elife.96281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
Negative memories engage a brain and body-wide stress response in humans that can alter cognition and behavior. Prolonged stress responses induce maladaptive cellular, circuit, and systems-level changes that can lead to pathological brain states and corresponding disorders in which mood and memory are affected. However, it is unclear if repeated activation of cells processing negative memories induces similar phenotypes in mice. In this study, we used an activity-dependent tagging method to access neuronal ensembles and assess their molecular characteristics. Sequencing memory engrams in mice revealed that positive (male-to-female exposure) and negative (foot shock) cells upregulated genes linked to anti- and pro-inflammatory responses, respectively. To investigate the impact of persistent activation of negative engrams, we chemogenetically activated them in the ventral hippocampus over 3 months and conducted anxiety and memory-related tests. Negative engram activation increased anxiety behaviors in both 6- and 14-month-old mice, reduced spatial working memory in older mice, impaired fear extinction in younger mice, and heightened fear generalization in both age groups. Immunohistochemistry revealed changes in microglial and astrocytic structure and number in the hippocampus. In summary, repeated activation of negative memories induces lasting cellular and behavioral abnormalities in mice, offering insights into the negative effects of chronic negative thinking-like behaviors on human health.
Collapse
Affiliation(s)
- Alexandra L Jellinger
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
| | - Rebecca L Suthard
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
- Graduate Program for Neuroscience, Boston UniversityBostonUnited States
| | - Bingbing Yuan
- Whitehead Institute for Biomedical Research, MITCambridgeUnited States
| | - Michelle Surets
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
| | - Evan A Ruesch
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
| | - Albit J Caban
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
- Graduate Program for Neuroscience, Boston UniversityBostonUnited States
| | - Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University Medical CenterNew YorkUnited States
| | - Monika Shpokayte
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
- Graduate Program for Neuroscience, Boston UniversityBostonUnited States
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston UniversityBostonUnited States
- Neurophotonics Center, and Photonics Center, Boston UniversityBostonUnited States
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
| |
Collapse
|
7
|
Popova EY, Kawasawa YI, Leung M, Barnstable CJ. Temporal changes in mouse hippocampus transcriptome after pilocarpine-induced seizures. Front Neurosci 2024; 18:1384805. [PMID: 39040630 PMCID: PMC11260795 DOI: 10.3389/fnins.2024.1384805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Status epilepticus (SE) is a seizure lasting more than 5 min that can have lethal consequences or lead to various neurological disorders, including epilepsy. Using a pilocarpine-induced SE model in mice we investigated temporal changes in the hippocampal transcriptome. Methods We performed mRNA-seq and microRNA-seq analyses at various times after drug treatment. Results At 1 h after the start of seizures, hippocampal cells upregulated transcription of immediate early genes and genes involved in the IGF-1, ERK/MAPK and RNA-PolII/transcription pathways. At 8 h, we observed changes in the expression of genes associated with oxidative stress, overall transcription downregulation, particularly for genes related to mitochondrial structure and function, initiation of a stress response through regulation of ribosome and translation/EIF2 signaling, and upregulation of an inflammatory response. During the middle of the latent period, 36 h, we identified upregulation of membrane components, cholesterol synthesis enzymes, channels, and extracellular matrix (ECM), as well as an increased inflammatory response. At the end of the latent period, 120 h, most changes in expression were in genes involved in ion transport, membrane channels, and synapses. Notably, we also elucidated the involvement of novel pathways, such as cholesterol biosynthesis pathways, iron/BMP/ferroptosis pathways, and circadian rhythms signaling in SE and epileptogenesis. Discussion These temporal changes in metabolic reactions indicate an immediate response to injury followed by recovery and regeneration. CREB was identified as the main upstream regulator. Overall, our data provide new insights into molecular functions and cellular processes involved at different stages of seizures and offer potential avenues for effective therapeutic strategies.
Collapse
Affiliation(s)
- Evgenya Y. Popova
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
- Penn State Hershey Eye Center, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, United States
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, NC, United States
| | - Ming Leung
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, NC, United States
| | - Colin J. Barnstable
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
- Penn State Hershey Eye Center, Hershey, PA, United States
| |
Collapse
|
8
|
Robertson KV, Rodriguez AS, Cartailler JP, Shrestha S, Schroeder KR, Valenti AM, Harrison FE, Hasty AH. Knockdown of microglial iron import gene, DMT1, worsens cognitive function and alters microglial transcriptional landscape in a sex-specific manner in the APP/PS1 model of Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-4559940. [PMID: 38978579 PMCID: PMC11230470 DOI: 10.21203/rs.3.rs-4559940/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background Microglial cell iron load and inflammatory activation are significant hallmarks of late-stage Alzheimer's disease (AD). In vitro, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and excess iron can augment cellular inflammation, suggesting a feed-forward loop between iron import mechanisms and inflammatory signaling. However, it is not understood whether microglial iron import mechanisms directly contribute to inflammatory signaling and chronic disease in vivo. These studies determined the effects of microglial-specific knockdown of Slc11a2 on AD-related cognitive decline and microglial transcriptional phenotype. Methods In vitro experiments and RT-qPCR were used to assess a role for DMT1 in amyloid-β-associated inflammation. To determine the effects of microglial Slc11a2 knockdown on AD-related phenotypes in vivo, triple-transgenic Cx3cr1 Cre - ERT2 ;Slc11a2 flfl;APP/PS1 + or - mice were generated and administered corn oil or tamoxifen to induce knockdown at 5-6 months of age. Both sexes underwent behavioral analyses to assess cognition and memory (12-15 months of age). Hippocampal CD11b + microglia were magnetically isolated from female mice (15-17 months) and bulk RNA-sequencing analysis was conducted. Results DMT1 inhibition in vitro robustly decreased Aβ-induced inflammatory gene expression and cellular iron levels in conditions of excess iron. In vivo, Slc11a2 KD APP/PS1 female, but not male, mice displayed a significant worsening of memory function in Morris water maze and a fear conditioning assay, along with significant hyperactivity compared to control WT and APP/PS1 mice. Hippocampal microglia from Slc11a2 KD APP/PS1 females displayed significant increases in Enpp2, Ttr, and the iron-export gene, Slc40a1, compared to control APP/PS1 cells. Slc11a2 KD cells from APP/PS1 females also exhibited decreased expression of markers associated with disease-associated microglia (DAMs), such as Apoe, Ctsb, Csf1, and Hif1α. Conclusions This work suggests a sex-specific role for microglial iron import gene Slc11a2 in propagating behavioral and cognitive phenotypes in the APP/PS1 model of AD. These data also highlight an association between loss of a DAM-like phenotype in microglia and cognitive deficits in Slc11a2 KD APP/PS1 female mice. Overall, this work illuminates an iron-related pathway in microglia that may serve a protective role during disease and offers insight into mechanisms behind disease-related sex differences.
Collapse
|
9
|
Yang G, Shan H, Wu D, Li S, Lai Z, Zheng F, Xiong Z, Xiong Z, Diao Y, Shan Y, Chen Y, Wang A, Liang W, Yin Y. COVID-19 increases extracorporeal coagulation during hemodialysis associated with upregulation of vWF/FBLN5 signaling in patients with severe/critical symptoms. BMC Infect Dis 2024; 24:427. [PMID: 38649864 PMCID: PMC11036607 DOI: 10.1186/s12879-024-09245-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND COVID-19 has been shown to increase the risk of extracorporeal coagulation during hemodialysis in patients, but the underlying mechanism remains unclear. This study aimed to investigate the effect and mechanism of COVID-19 on the risk of extracorporeal coagulation in patients with chronic kidney disease undergoing hemodialysis. METHODS A retrospective analysis of the extracorporeal coagulation status of 339 hemodialysis patients at our center before and after COVID-19 infection was performed, including subgroup analyses. Post-infection blood composition was analyzed by protein spectrometry and ELISA. RESULTS Compared to the pre-COVID-19 infection period, COVID-19-induced extracorporeal coagulation predominantly occurred in patients with severe/critical symptoms. Further proteomic analysis demonstrated that in patients with severe/critical symptoms, the coagulation cascade reaction, platelet activation, inflammation, and oxidative stress-related pathways were significantly amplified compared to those in patients with no/mild symptoms. Notably, the vWF/FBLN5 pathway, which is associated with inflammation, vascular injury, and coagulation, was significantly upregulated. CONCLUSIONS Patients with severe/critical COVID-19 symptoms are at a higher risk of extracorporeal coagulation during hemodialysis, which is associated with the upregulation of the vWF/FBLN5 signaling pathway. These findings highlight the importance of early anticoagulant therapy initiation in COVID-19 patients with severe/critical symptoms, particularly those undergoing hemodialysis. Additionally, vWF/FBLN5 upregulation may be a novel mechanism for virus-associated thrombosis/coagulation.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
- Shenzhen Clinical Research Centre for Urology and Nephrology, Shenzhen, 518036, China.
- Institute of Nephrology, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518036, China.
| | - Hui Shan
- Precision Medicine Research Institute, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Dibin Wu
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Sanmu Li
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Zhiwei Lai
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Fengping Zheng
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Zibo Xiong
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
- Shenzhen Clinical Research Centre for Urology and Nephrology, Shenzhen, 518036, China
| | - Zuying Xiong
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
- Shenzhen Clinical Research Centre for Urology and Nephrology, Shenzhen, 518036, China
- Institute of Nephrology, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518036, China
| | - Yuhan Diao
- Department of Medical Records & Statistics, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Ying Shan
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China
| | - Yun Chen
- Institute of Ultrasound Medicine, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518036, China
| | - Aihong Wang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| | - Wei Liang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| | - Yuxin Yin
- Precision Medicine Research Institute, Peking University Shenzhen Hospital, Peking University, Shenzhen, 518036, China.
| |
Collapse
|
10
|
Privitera M, von Ziegler LM, Floriou-Servou A, Duss SN, Zhang R, Waag R, Leimbacher S, Sturman O, Roessler FK, Heylen A, Vermeiren Y, Van Dam D, De Deyn PP, Germain PL, Bohacek J. Noradrenaline release from the locus coeruleus shapes stress-induced hippocampal gene expression. eLife 2024; 12:RP88559. [PMID: 38477670 DOI: 10.7554/elife.88559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024] Open
Abstract
Exposure to an acute stressor triggers a complex cascade of neurochemical events in the brain. However, deciphering their individual impact on stress-induced molecular changes remains a major challenge. Here, we combine RNA sequencing with selective pharmacological, chemogenetic, and optogenetic manipulations to isolate the contribution of the locus coeruleus-noradrenaline (LC-NA) system to the acute stress response in mice. We reveal that NA release during stress exposure regulates a large and reproducible set of genes in the dorsal and ventral hippocampus via β-adrenergic receptors. For a smaller subset of these genes, we show that NA release triggered by LC stimulation is sufficient to mimic the stress-induced transcriptional response. We observe these effects in both sexes, and independent of the pattern and frequency of LC activation. Using a retrograde optogenetic approach, we demonstrate that hippocampus-projecting LC neurons directly regulate hippocampal gene expression. Overall, a highly selective set of astrocyte-enriched genes emerges as key targets of LC-NA activation, most prominently several subunits of protein phosphatase 1 (Ppp1r3c, Ppp1r3d, Ppp1r3g) and type II iodothyronine deiodinase (Dio2). These results highlight the importance of astrocytic energy metabolism and thyroid hormone signaling in LC-mediated hippocampal function and offer new molecular targets for understanding how NA impacts brain function in health and disease.
Collapse
Affiliation(s)
- Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Lukas M von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Sian N Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Runzhong Zhang
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Sebastian Leimbacher
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| | - Fabienne K Roessler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Annelies Heylen
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yannick Vermeiren
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University & Research (WUR), Wageningen, Netherlands
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Peter P De Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
- Department of Neurology, Memory Clinic of Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Pierre-Luc Germain
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
- Laboratory of Statistical Bioinformatics, University of Zürich, Zürich, Switzerland
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland, Zurich, Switzerland
| |
Collapse
|
11
|
Bhuiyan P, Sun Z, Khan MA, Hossain MA, Rahman MH, Qian Y. System biology approaches to identify hub genes linked with ECM organization and inflammatory signaling pathways in schizophrenia pathogenesis. Heliyon 2024; 10:e25191. [PMID: 38322840 PMCID: PMC10844262 DOI: 10.1016/j.heliyon.2024.e25191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 12/18/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Schizophrenia (SZ) is a chronic and devastating mental illness that affects around 20 million individuals worldwide. Cognitive deficits and structural and functional changes of the brain, abnormalities of brain ECM components, chronic neuroinflammation, and devastating clinical manifestation during SZ are likely etiological factors shown by affected individuals. However, the pathophysiological events associated with multiple regulatory pathways involved in the brain of this complex disorder are still unclear. This study aimed to develop a pipeline based on bioinformatics and systems biology approaches for identifying potential therapeutic targets involving possible biological mechanisms from SZ patients and healthy volunteers. About 420 overlapping differentially expressed genes (DEGs) from three RNA-seq datasets were identified. Gene ontology (GO), and pathways analysis showed several biological mechanisms enriched by the commonly shared DEGs, including extracellular matrix organization (ECM) organization, collagen fibril organization, integrin signaling pathway, inflammation mediated by chemokines and cytokines signaling pathway, and GABA-B receptor II and IL4 mediated signaling. Besides, 15 hub genes (FN1, COL1A1, COL3A1, COL1A2, COL5A1, COL2A1, COL6A2, COL6A3, MMP2, THBS1, DCN, LUM, HLA-A, HLA-C, and FBN1) were discovered by comprehensive analysis, which was mainly involved in the ECM organization and inflammatory signaling pathway. Furthermore, the miRNA target of the hub genes was analyzed with the random-forest-based approach software miRTarBase. In addition, the transcriptional factors and protein kinases regulating overlapping DEGs in SZ, namely, SUZ12, EZH2, TRIM28, TP53, EGR1, CSNK2A1, GSK3B, CDK1, and MAPK14, were also identified. The results point to a new understanding that the hub genes (fibronectin 1, collagen, matrix metalloproteinase-2, and lumican) in the ECM organization and inflammatory signaling pathways may be involved in the SZ occurrence and pathogenesis.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, 1209, Bangladesh
| | - Zhaochu Sun
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | - Md Arif Khan
- Department of Biotechnology and Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka, 1209, Bangladesh
- Bio-Bio-1 Bioinformatics Research Foundation, Dhaka, Bangladesh
| | - Md Arju Hossain
- Department of Microbiology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, Islamic University, Kushtia-7003, Bangladesh
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| |
Collapse
|
12
|
Gamboa-Sánchez C, Becerril-Villanueva E, Alvarez-Herrera S, Leyva-Mascareño G, González-López SL, Estudillo E, Fernández-Molina AE, Elizalde-Contreras JM, Ruiz-May E, Segura-Cabrera A, Jiménez-Genchi J, Pavón L, Zamudio SR, Pérez-Sánchez G. Upregulation of S100A8 in peripheral blood mononuclear cells from patients with depression treated with SSRIs: a pilot study. Proteome Sci 2023; 21:23. [PMID: 38049858 PMCID: PMC10694904 DOI: 10.1186/s12953-023-00224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/17/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Major depressive disorder (MDD) affects more than 350 million people worldwide, and there is currently no laboratory test to diagnose it. This pilot study aimed to identify potential biomarkers in peripheral blood mononuclear cells (PBMCs) from MDD patients. METHODS We used tandem mass tagging coupled to synchronous precursor selection (mass spectrometry) to obtain the differential proteomic profile from a pool of PBMCs from MDD patients and healthy subjects, and quantitative PCR to assess gene expression of differentially expressed proteins (DEPs) of our interest. RESULTS We identified 247 proteins, of which 133 had a fold change ≥ 2.0 compared to healthy volunteers. Using pathway enrichment analysis, we found that some processes, such as platelet degranulation, coagulation, and the inflammatory response, are perturbed in MDD patients. The gene-disease association analysis showed that molecular alterations in PBMCs from MDD patients are associated with cerebral ischemia, vascular disease, thrombosis, acute coronary syndrome, and myocardial ischemia, in addition to other conditions such as inflammation and diabetic retinopathy. CONCLUSIONS We confirmed by qRT-PCR that S100A8 is upregulated in PBMCs from MDD patients and thus could be an emerging biomarker of this disorder. This report lays the groundwork for future studies in a broader and more diverse population and contributes to a deeper characterization of MDD.
Collapse
Affiliation(s)
- Concepción Gamboa-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, 07738, Ciudad de México, México
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Gabriela Leyva-Mascareño
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Sandra L González-López
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Av. Insurgentes Sur 3877 Del. Tlalpan, 14269. Col. La Fama., Ciudad de México, México
| | - Alberto E Fernández-Molina
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - José Miguel Elizalde-Contreras
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A. C, Cluster BioMimic®, Carretera Antigua a Coatepec 351, Congregación El Haya, 91073, Xalapa, Veracruz, México
| | - Eliel Ruiz-May
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A. C, Cluster BioMimic®, Carretera Antigua a Coatepec 351, Congregación El Haya, 91073, Xalapa, Veracruz, México
| | - Aldo Segura-Cabrera
- Red de Estudios Moleculares Avanzados, Instituto de Ecología A. C, Cluster BioMimic®, Carretera Antigua a Coatepec 351, Congregación El Haya, 91073, Xalapa, Veracruz, México
- Genomic Sciences, GSK, Stevenage, UK
| | - Janeth Jiménez-Genchi
- Hospital Psiquiátrico Fray Bernardino Álvarez. Av, Niño Jesús, San Buenaventura 214000, Tlalpan, Ciudad de Mexico, México
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México
| | - Sergio Roberto Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, 07738, Ciudad de México, México.
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de La Fuente Muñiz, Colonia San Lorenzo Huipulco, Calzada México-Xochimilco 101, Tlalpan, 14370, Ciudad de Mexico, México.
| |
Collapse
|
13
|
Wu A, Zhang J. Neuroinflammation, memory, and depression: new approaches to hippocampal neurogenesis. J Neuroinflammation 2023; 20:283. [PMID: 38012702 PMCID: PMC10683283 DOI: 10.1186/s12974-023-02964-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
As one of most common and severe mental disorders, major depressive disorder (MDD) significantly increases the risks of premature death and other medical conditions for patients. Neuroinflammation is the abnormal immune response in the brain, and its correlation with MDD is receiving increasing attention. Neuroinflammation has been reported to be involved in MDD through distinct neurobiological mechanisms, among which the dysregulation of neurogenesis in the dentate gyrus (DG) of the hippocampus (HPC) is receiving increasing attention. The DG of the hippocampus is one of two niches for neurogenesis in the adult mammalian brain, and neurotrophic factors are fundamental regulators of this neurogenesis process. The reported cell types involved in mediating neuroinflammation include microglia, astrocytes, oligodendrocytes, meningeal leukocytes, and peripheral immune cells which selectively penetrate the blood-brain barrier and infiltrate into inflammatory regions. This review summarizes the functions of the hippocampus affected by neuroinflammation during MDD progression and the corresponding influences on the memory of MDD patients and model animals.
Collapse
Affiliation(s)
- Anbiao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
14
|
Moshfegh CM, Elkhatib SK, Watson GF, Drake J, Taylor ZN, Reed EC, Lauten TH, Clopp AJ, Vladimirov VI, Case AJ. S100a9 Protects Against the Effects of Repeated Social Defeat Stress. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:919-929. [PMID: 37881565 PMCID: PMC10593888 DOI: 10.1016/j.bpsgos.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/07/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Background Posttraumatic stress disorder, a consequence of psychological trauma, is associated with increased inflammation and an elevated risk of developing comorbid inflammatory diseases. However, the mechanistic link between this mental health disorder and inflammation remains elusive. We previously found that S100a8 and S100a9 messenger RNA, genes that encode the protein calprotectin, were significantly upregulated in T lymphocytes and positively correlated with inflammatory gene expression and the mitochondrial redox environment in these cells. Therefore, we hypothesized that genetic deletion of calprotectin would attenuate the inflammatory and redox phenotype displayed after psychological trauma. Methods We used a preclinical mouse model of posttraumatic stress disorder known as repeated social defeat stress (RSDS) combined with pharmacological and genetic manipulation of S100a9 (which functionally eliminates calprotectin). A total of 186 animals (93 control, 93 RSDS) were used in these studies. Results Unexpectedly, we observed worsening of behavioral pathology, inflammation, and the mitochondrial redox environment in mice after RSDS compared with wild-type animals. Furthermore, loss of calprotectin significantly enhanced the metabolic demand on T lymphocytes, suggesting that this protein may play an undescribed role in mitochondrial regulation. This was further supported by single-cell RNA sequencing analysis demonstrating that RSDS and loss of S100a9 primarily altered genes associated with mitochondrial function and oxidative phosphorylation. Conclusions These data demonstrate that the loss of calprotectin potentiates the RSDS-induced phenotype, which suggests that its observed upregulation after psychological trauma may provide previously unexplored protective functions.
Collapse
Affiliation(s)
- Cassandra M. Moshfegh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Safwan K. Elkhatib
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gabrielle F. Watson
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - John Drake
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
| | - Zachary N. Taylor
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
| | - Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| | - Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| | - Amelia J. Clopp
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| | - Vladimir I. Vladimirov
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Psychiatry, University of Arizona, Phoenix, Arizona
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, Maryland
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| |
Collapse
|
15
|
Gerstner JR, Flores CC, Lefton M, Rogers B, Davis CJ. FABP7: a glial integrator of sleep, circadian rhythms, plasticity, and metabolic function. Front Syst Neurosci 2023; 17:1212213. [PMID: 37404868 PMCID: PMC10315501 DOI: 10.3389/fnsys.2023.1212213] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Sleep and circadian rhythms are observed broadly throughout animal phyla and influence neural plasticity and cognitive function. However, the few phylogenetically conserved cellular and molecular pathways that are implicated in these processes are largely focused on neuronal cells. Research on these topics has traditionally segregated sleep homeostatic behavior from circadian rest-activity rhythms. Here we posit an alternative perspective, whereby mechanisms underlying the integration of sleep and circadian rhythms that affect behavioral state, plasticity, and cognition reside within glial cells. The brain-type fatty acid binding protein, FABP7, is part of a larger family of lipid chaperone proteins that regulate the subcellular trafficking of fatty acids for a wide range of cellular functions, including gene expression, growth, survival, inflammation, and metabolism. FABP7 is enriched in glial cells of the central nervous system and has been shown to be a clock-controlled gene implicated in sleep/wake regulation and cognitive processing. FABP7 is known to affect gene transcription, cellular outgrowth, and its subcellular localization in the fine perisynaptic astrocytic processes (PAPs) varies based on time-of-day. Future studies determining the effects of FABP7 on behavioral state- and circadian-dependent plasticity and cognitive processes, in addition to functional consequences on cellular and molecular mechanisms related to neural-glial interactions, lipid storage, and blood brain barrier integrity will be important for our knowledge of basic sleep function. Given the comorbidity of sleep disturbance with neurological disorders, these studies will also be important for our understanding of the etiology and pathophysiology of how these diseases affect or are affected by sleep.
Collapse
Affiliation(s)
- Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Micah Lefton
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Brooke Rogers
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
16
|
Kambe J, Miyata S, Li C, Yamamoto Y, Nagaoka K. Xanthine-induced deficits in hippocampal behavior and abnormal expression of hemoglobin genes. Behav Brain Res 2023; 449:114476. [PMID: 37148916 DOI: 10.1016/j.bbr.2023.114476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
The prevalence of mental disorders such as depression and anxiety is high and often comorbid with other diseases. Chronic stress is a common risk factor for these disorders, but the mechanisms behind their development are not yet fully understood. Metabolomics has revealed a close association between purine and pyrimidine metabolism and depression and anxiety, with increased levels of serum xanthine observed in both humans and mice. Xanthine is known as purine metabolism, and this compound shows several biological activities, but the impact of xanthine on our brain function is still unclear. The hippocampus, which plays a crucial role in memory and learning, is also implicated in the pathophysiology of depression and anxiety. Here, we investigated the effects of xanthine intraperitoneal administration on spatial memory and anxiety-like behavior in mice. The findings indicated that xanthine administration induced a deficit of hippocampus-dependent spatial memory and a tendency to anxiety-like behavior in mice. RNA-seq analysis showed that xanthine administration upregulated hemoglobin (Hb) genes involved in oxygen transport in the hippocampus. The upregulated Hb genes occurred in the neuronal cells, and in vitro experiments revealed that both Hba-a1 derived from mice and HBA2 derived from humans were upregulated by xanthine treatment. These observations suggest that the xanthine-induced Hb in the hippocampus could be related to spatial memory deficit and anxiety. This study sheds light on the direct effects of xanthine on the brain and its potential role in the development of depression and anxiety symptoms caused by chronic stress.
Collapse
Affiliation(s)
- Jun Kambe
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shiho Miyata
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Chunmei Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuki Yamamoto
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Tokyo University of Agriculture and Technology, Tokyo, Japan.
| |
Collapse
|
17
|
Edelmann S, Wiegand A, Hentrich T, Pasche S, Schulze-Hentrich JM, Munk MHJ, Fallgatter AJ, Kreifelts B, Nieratschker V. Blood transcriptome analysis suggests an indirect molecular association of early life adversities and adult social anxiety disorder by immune-related signal transduction. Front Psychiatry 2023; 14:1125553. [PMID: 37181876 PMCID: PMC10168183 DOI: 10.3389/fpsyt.2023.1125553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/29/2023] [Indexed: 05/16/2023] Open
Abstract
Social anxiety disorder (SAD) is a psychiatric disorder characterized by severe fear in social situations and avoidance of these. Multiple genetic as well as environmental factors contribute to the etiopathology of SAD. One of the main risk factors for SAD is stress, especially during early periods of life (early life adversity; ELA). ELA leads to structural and regulatory alterations contributing to disease vulnerability. This includes the dysregulation of the immune response. However, the molecular link between ELA and the risk for SAD in adulthood remains largely unclear. Evidence is emerging that long-lasting changes of gene expression patterns play an important role in the biological mechanisms linking ELA and SAD. Therefore, we conducted a transcriptome study of SAD and ELA performing RNA sequencing in peripheral blood samples. Analyzing differential gene expression between individuals suffering from SAD with high or low levels of ELA and healthy individuals with high or low levels of ELA, 13 significantly differentially expressed genes (DEGs) were identified with respect to SAD while no significant differences in expression were identified with respect to ELA. The most significantly expressed gene was MAPK3 (p = 0.003) being upregulated in the SAD group compared to control individuals. In contrary, weighted gene co-expression network analysis (WGCNA) identified only modules significantly associated with ELA (p ≤ 0.05), not with SAD. Furthermore, analyzing interaction networks of the genes from the ELA-associated modules and the SAD-related MAPK3 revealed complex interactions of those genes. Gene functional enrichment analyses indicate a role of signal transduction pathways as well as inflammatory responses supporting an involvement of the immune system in the association of ELA and SAD. In conclusion, we did not identify a direct molecular link between ELA and adult SAD by transcriptional changes. However, our data indicate an indirect association of ELA and SAD mediated by the interaction of genes involved in immune-related signal transduction.
Collapse
Affiliation(s)
- Susanne Edelmann
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Ariane Wiegand
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Max Planck Fellow Group Precision Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Thomas Hentrich
- Institute for Medical Genetics and Applied Genomics, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Department of Genetics and Epigenetics, Faculty NT, Saarland University, Saarbrücken, Germany
| | - Sarah Pasche
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Julia Maria Schulze-Hentrich
- Institute for Medical Genetics and Applied Genomics, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Department of Genetics and Epigenetics, Faculty NT, Saarland University, Saarbrücken, Germany
| | - Matthias H. J. Munk
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Andreas J. Fallgatter
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Benjamin Kreifelts
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Vanessa Nieratschker
- Department of Psychiatry and Psychotherapy, University Hospital of Tuebingen, Tuebingen Center for Mental Health (TüCMH), Eberhard Karls University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
18
|
Lee H, Park J, Kim S. Metabolic and Transcriptomic Signatures of the Acute Psychological Stress Response in the Mouse Brain. Metabolites 2023; 13:metabo13030453. [PMID: 36984893 PMCID: PMC10052811 DOI: 10.3390/metabo13030453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Acute stress response triggers various physiological responses such as energy mobilization to meet metabolic demands. However, the underlying molecular changes in the brain remain largely obscure. Here, we used a brief water avoidance stress (WAS) to elicit an acute stress response in mice. By employing RNA-sequencing and metabolomics profiling, we investigated the acute stress-induced molecular changes in the mouse whole brain. The aberrant expression of 60 genes was detected in the brain tissues of WAS-exposed mice. Functional analyses showed that the aberrantly expressed genes were enriched in various processes such as superoxide metabolism. In our global metabolomic profiling, a total of 43 brain metabolites were significantly altered by acute WAS. Metabolic pathways upregulated from WAS-exposed brain tissues relative to control samples included lipolysis, eicosanoid biosynthesis, and endocannabinoid synthesis. Acute WAS also elevated the levels of branched-chain amino acids, 5-aminovalerates, 4-hydroxy-nonenal-glutathione as well as mannose, suggesting complex metabolic changes in the brain. The observed molecular events in the present study provide a valuable resource that can help us better understand how acute psychological stress impacts neural functions.
Collapse
Affiliation(s)
- Haein Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jina Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
19
|
Martini P, Mingardi J, Carini G, Mattevi S, Ndoj E, La Via L, Magri C, Gennarelli M, Russo I, Popoli M, Musazzi L, Barbon A. Transcriptional Profiling of Rat Prefrontal Cortex after Acute Inescapable Footshock Stress. Genes (Basel) 2023; 14:genes14030740. [PMID: 36981011 PMCID: PMC10048409 DOI: 10.3390/genes14030740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Stress is a primary risk factor for psychiatric disorders such as Major Depressive Disorder (MDD) and Post Traumatic Stress Disorder (PTSD). The response to stress involves the regulation of transcriptional programs, which is supposed to play a role in coping with stress. To evaluate transcriptional processes implemented after exposure to unavoidable traumatic stress, we applied microarray expression analysis to the PFC of rats exposed to acute footshock (FS) stress that were sacrificed immediately after the 40 min session or 2 h or 24 h after. While no substantial changes were observed at the single gene level immediately after the stress session, gene set enrichment analysis showed alterations in neuronal pathways associated with glia development, glia-neuron networking, and synaptic function. Furthermore, we found alterations in the expression of gene sets regulated by specific transcription factors that could represent master regulators of the acute stress response. Of note, these pathways and transcriptional programs are activated during the early stress response (immediately after FS) and are already turned off after 2 h-while at 24 h, the transcriptional profile is largely unaffected. Overall, our analysis provided a transcriptional landscape of the early changes triggered by acute unavoidable FS stress in the PFC of rats, suggesting that the transcriptional wave is fast and mild, but probably enough to activate a cellular response to acute stress.
Collapse
Affiliation(s)
- Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Jessica Mingardi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Giulia Carini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25123 Brescia, Italy
| | - Stefania Mattevi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Elona Ndoj
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Chiara Magri
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25123 Brescia, Italy
| | - Isabella Russo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25123 Brescia, Italy
| | - Maurizio Popoli
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Laura Musazzi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
20
|
Saito-Takatsuji H, Yoshitomi Y, Yamamoto R, Furuyama T, Ishigaki Y, Kato N, Yonekura H, Ikeda T. Transthyretin Is Commonly Upregulated in the Hippocampus of Two Stress-Induced Depression Mouse Models. Int J Mol Sci 2023; 24:ijms24043736. [PMID: 36835151 PMCID: PMC9964880 DOI: 10.3390/ijms24043736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/28/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Chronic stress can affect gene expression in the hippocampus, which alters neural and cerebrovascular functions, thereby contributing to the development of mental disorders such as depression. Although several differentially expressed genes in the depressed brain have been reported, gene expression changes in the stressed brain remain underexplored. Therefore, this study examines hippocampal gene expression in two mouse models of depression induced by forced swim stress (FSS) and repeated social defeat stress (R-SDS). Transthyretin (Ttr) was commonly upregulated in the hippocampus of both mouse models, as determined by microarray, RT-qPCR, and Western blot analyses. Evaluation of the effects of overexpressed Ttr in the hippocampus using adeno-associated virus-mediated gene transfer revealed that TTR overexpression induced depression-like behavior and upregulation of Lcn2 and several proinflammatory genes (Icam1 and Vcam1) in the hippocampus. Upregulation of these inflammation-related genes was confirmed in the hippocampus obtained from mice vulnerable to R-SDS. These results suggest that chronic stress upregulates Ttr expression in the hippocampus and that Ttr upregulation may be involved in the induction of depression-like behavior.
Collapse
Affiliation(s)
- Hidehito Saito-Takatsuji
- Department of Biochemistry, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Yasuo Yoshitomi
- Department of Biochemistry, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Takafumi Furuyama
- Department of Physiology, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Yasuhito Ishigaki
- Division of Molecular and Cell Biology, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Hideto Yonekura
- Department of Biochemistry, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Takayuki Ikeda
- Department of Biochemistry, Kanazawa Medical University School of Medicine, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
- Correspondence: ; Tel.: +81-76-218-8111
| |
Collapse
|
21
|
Overnight Corticosterone and Gene Expression in Mouse Hippocampus: Time Course during Resting Period. Int J Mol Sci 2023; 24:ijms24032828. [PMID: 36769150 PMCID: PMC9917930 DOI: 10.3390/ijms24032828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The aim of the experiment was to test the effect of an elevated level of glucocorticoids on the mouse hippocampal transcriptome after 12 h of treatment with corticosterone that was administered during an active phase of the circadian cycle. Additionally, we also tested the circadian changes in gene expression and the decay time of transcriptomic response to corticosterone. Gene expression was analyzed using microarrays. Obtained results show that transcriptomic responses to glucocorticoids are heterogeneous in terms of the decay time with some genes displaying persistent changes in expression during 9 h of rest. We have also found a considerable overlap between genes regulated by corticosterone and genes implicated previously in stress response. The examples of such genes are Acer2, Agt, Apod, Aqp4, Etnppl, Fabp7, Fam107a, Fjx1, Fmo2, Galnt15, Gjc2, Heph, Hes5, Htra1, Jdp2, Kif5a, Lfng, Lrg1, Mgp, Mt1, Pglyrp1, Pla2g3, Plin4, Pllp, Ptgds, Ptn, Slc2a1, Slco1c1, Sult1a1, Thbd and Txnip. This indicates that the applied model is a useful tool for the investigation of mechanisms underlying the stress response.
Collapse
|
22
|
Klotho Regulated by Estrogen Plays a Key Role in Sex Differences in Stress Resilience in Rats. Int J Mol Sci 2023; 24:ijms24021206. [PMID: 36674721 PMCID: PMC9862442 DOI: 10.3390/ijms24021206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Klotho (KL) is a glycosyl hydrolase and aging-suppressor gene. Stress is a risk factor for depression and anxiety, which are highly comorbid with each other. The aim of this study is to determine whether KL is regulated by estrogen and plays an important role in sex differences in stress resilience. Our results showed that KL is regulated by estrogen in rat hippocampal neurons in vivo and in vitro and is essential for the estrogen-mediated increase in the number of presynaptic vesicular glutamate transporter 1 (Vglut1)-positive clusters on the dendrites of hippocampal neurons. The role of KL in sex differences in stress response was examined in rats using 3-week chronic unpredictable mild stress (CUMS). CUMS produced a deficit in spatial learning and memory, anhedonic-like behaviors, and anxiety-like behaviors in male but not female rats, which was accompanied by a reduction in KL protein levels in the hippocampus of male but not female rats. This demonstrated the resilience of female rats to CUMS. Interestingly, the knockdown of KL protein levels in the rat hippocampus of both sexes caused a decrease in stress resilience in both sexes, especially in female rats. These results suggest that the regulation of KL by estrogen plays an important role in estrogen-mediated synapse formation and that KL plays a critical role in the sex differences in cognitive deficit, anhedonic-like behaviors, and anxiety-like behaviors induced by chronic stress in rats, highlighting an important role of KL in sex differences in stress resilience.
Collapse
|
23
|
Chong YS, Wong LW, Gaunt J, Lee YJ, Goh CS, Morris RGM, Ch'ng TH, Sajikumar S. Distinct contributions of ventral CA1/amygdala co-activation to the induction and maintenance of synaptic plasticity. Cereb Cortex 2023; 33:676-690. [PMID: 35253866 DOI: 10.1093/cercor/bhac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 02/03/2023] Open
Abstract
The amygdala is known to modulate hippocampal synaptic plasticity. One role could be an immediate effect of basolateral amygdala (BLA) in priming synaptic plasticity in the hippocampus. Another role could be through associative synaptic co-operation and competition that triggers events involved in the maintenance of synaptic potentiation. We present evidence that the timing and activity level of BLA stimulation are important factors for the induction and maintenance of long-term potentiation (LTP) in ventral hippocampal area CA1. A 100 Hz BLA co-stimulation facilitated the induction of LTP, whereas 200 Hz co-stimulation attenuated induction. A 100 Hz BLA co-stimulation also caused enhanced persistence, sufficient to prevent synaptic competition. This maintenance effect is likely through translational mechanisms, as mRNA expression of primary response genes was unaffected, whereas protein level of plasticity-related products was increased. Further understanding of the neural mechanisms of amygdala modulation on hippocampus could provide insights into the mechanisms of emotional disorders.
Collapse
Affiliation(s)
- Yee Song Chong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Jessica Gaunt
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.,Interdisciplinary Graduate School, Nanyang Technological University, Singapore 637335, Singapore
| | - Cai Shan Goh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Richard G M Morris
- Laboratory for Cognitive Neuroscience, Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh EH8 9JZ, Scotland
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
24
|
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: 1.5] [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
|
25
|
Manganese-dependent microRNA trimming by 3'→5' exonucleases generates 14-nucleotide or shorter tiny RNAs. Proc Natl Acad Sci U S A 2022; 119:e2214335119. [PMID: 36508664 PMCID: PMC9907110 DOI: 10.1073/pnas.2214335119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are about 22-nucleotide (nt) noncoding RNAs forming the effector complexes with Argonaute (AGO) proteins to repress gene expression. Although tiny RNAs (tyRNAs) shorter than 19 nt have been found to bind to plant and vertebrate AGOs, their biogenesis remains a long-standing question. Here, our in vivo and in vitro studies show several 3'→5' exonucleases, such as interferon-stimulated gene 20 kDa (ISG20), three prime repair exonuclease 1 (TREX1), and ERI1 (enhanced RNAi, also known as 3'hExo), capable of trimming AGO-associated full-length miRNAs to 14-nt or shorter tyRNAs. Their guide trimming occurs in a manganese-dependent manner but independently of the guide sequence and the loaded four human AGO paralogs. We also show that ISG20-mediated guide trimming makes Argonaute3 (AGO3) a slicer. Given the high Mn2+ concentrations in stressed cells, virus-infected cells, and neurodegeneration, our study sheds light on the roles of the Mn2+-dependent exonucleases in remodeling gene silencing.
Collapse
|
26
|
Yin W, Swanson SP, Biltz RG, Goodman EJ, Gallagher NR, Sheridan JF, Godbout JP. Unique brain endothelial profiles activated by social stress promote cell adhesion, prostaglandin E2 signaling, hypothalamic-pituitary-adrenal axis modulation, and anxiety. Neuropsychopharmacology 2022; 47:2271-2282. [PMID: 36104533 PMCID: PMC9630498 DOI: 10.1038/s41386-022-01434-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/06/2022] [Accepted: 08/16/2022] [Indexed: 02/05/2023]
Abstract
Chronic stress may precipitate psychiatric disorders including anxiety. We reported that Repeated Social Defeat (RSD) in mice increased accumulation of inflammatory monocytes within the brain vasculature, which corresponded with increased interleukin (IL)-1 Receptor 1-mediated activation of endothelia, and augmented anxiety-like behavior. One unknown, however, is the role of immune-activated endothelia in regulating the physiological and behavioral responses to social stress. Thus, we sought to determine the RNA profile of activated endothelia and delineate the pathways by which these endothelia communicate within the brain to influence key responses to social stress. First, endothelial-specific RiboTag mice were exposed to RSD and brain endothelial mRNA profiles from the whole brain and prefrontal cortex were determined using RNAseq. RSD increased expression of cell adhesion molecules (Icam1), inflammatory genes (Lrg1, Lcn2, Ackr1, Il1r1), and cyclooxygenase-2 (Ptgs2/COX-2). In studies with IL-1R1KO mice, there was clear dependence on IL-1R1 on endothelia-associated transcripts including Lrg1, Icam1, Lcn2. Moreover, prostaglandin (PG)E2 was increased in the brain after RSD and Ptgs2 was localized to endothelia, especially within the hypothalamus. Next, a selective COX-2 inhibitor, Celecoxib (CCB), was used with social stress. RSD increased PGE2 in the brain and this was abrogated by CCB. Moreover, CCB reduced RSD-induced Hypothalamic-Pituitary-Adrenal (HPA) axis activation with attenuation of hypothalamic paraventricular neuron activation, hypothalamic Crh expression, and corticosterone in circulation. Production, release, and accumulation of inflammatory monocytes after RSD was COX-2 independent. Nonetheless, CCB blocked anxiety-like behavior in response to RSD. Collectively, social stress stimulated specific endothelia RNA profiles associated with increased cell adhesion, IL-1 and prostaglandin signaling, HPA axis activation, and anxiety.
Collapse
Affiliation(s)
- Wenyuan Yin
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, 43210, Columbus, OH, USA
| | - Samuel P Swanson
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, 43210, Columbus, OH, USA
| | - Rebecca G Biltz
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, 43210, Columbus, OH, USA
| | - Ethan J Goodman
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, 43210, Columbus, OH, USA
| | - Natalie R Gallagher
- Institute for Behavioral Medicine Research, Wexner Medicine Center, The Ohio State University, 43210, Columbus, OH, USA
- Division of Biosciences, College of Dentistry, The Ohio State University, 43210, Columbus, OH, USA
| | - John F Sheridan
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, 43210, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, Wexner Medicine Center, The Ohio State University, 43210, Columbus, OH, USA.
- Division of Biosciences, College of Dentistry, The Ohio State University, 43210, Columbus, OH, USA.
| | - Jonathan P Godbout
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, 43210, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, Wexner Medicine Center, The Ohio State University, 43210, Columbus, OH, USA.
| |
Collapse
|
27
|
Anxiolytic-like Effect of Inhaled Cinnamon Essential Oil and Its Main Component Cinnamaldehyde in Animal Models. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227997. [PMID: 36432096 PMCID: PMC9693619 DOI: 10.3390/molecules27227997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/19/2022]
Abstract
Aromatherapy is one of the most common safer alternative treatments for psychiatric disorders with fewer side effects than conventional drugs. Here, we investigated the effects of cinnamon essential oil (CIEO) inhalation on mouse behaviors by performing different behavioral tests. CIEO inhalation showed anxiolytic effects in the elevated plus maze test, as inferred from increased time spent in open arms and decreased time spent in closed arms. Moreover, the CIEO treatment enhanced social behavior by increasing the total contact number, time spent in the center, distance traveled in the center, and total distance in the social interaction test. However, CIEO inhalation did not have any effect on performance in the open field test, tail suspension test, forced swimming test, and Y maze tests. The microarray analysis indicated that the CIEO treatment downregulated 17 genes and upregulated 15 genes in the hippocampus. Among them, Dcc, Egr2, and Fos are the most crucial genes that are involved in anxiety-related biological processes and pathways, including the regulation of neuronal death and neuroinflammation. Gas chromatography/mass spectrometry analysis revealed that cinnamaldehyde is the main component of CIEO. Cinnamaldehyde recovered MK-801-induced anxiety-related changes in the electroencephalogram power spectrum in zebrafish. Taken together, our findings suggest that CIEO and its main component cinnamaldehyde have an anxiolytic effect through the regulation of the expression of genes related to neuroinflammatory response and neuronal death.
Collapse
|
28
|
Reshetnikov VV, Kisaretova PE, Bondar NP. Transcriptome Alterations Caused by Social Defeat Stress of Various Durations in Mice and Its Relevance to Depression and Posttraumatic Stress Disorder in Humans: A Meta-Analysis. Int J Mol Sci 2022; 23:ijms232213792. [PMID: 36430271 PMCID: PMC9698544 DOI: 10.3390/ijms232213792] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
The research on molecular causes of stress-associated psychopathologies is becoming highly important because the number of people with depression, generalized anxiety disorder and posttraumatic stress disorders (PTSDs) is steadily increasing every year. Investigation of molecular mechanisms in animal models opens up broad prospects for researchers, but relevant molecular signatures can differ significantly between patients and animal models. In our work, we for the first time carried out a meta-analysis of transcriptome changes in the prefrontal cortex of C57BL/6 mice after 10 and 30 days of social defeat stress (SDS). We then examined possible correlations of these alterations with transcriptome changes found in post-mortem samples from patients with depression or PTSD. Although transcriptional signatures of human psychiatric disorders and SDS did not overlap substantially, our results allowed us to identify the most reproducible changes seen after SDS of various durations. In addition, we were able to identify the genes involved in susceptibility to SDS after 10 days of stress. Taken together, these data help us to elucidate the molecular changes induced by SDS depending on its duration as well as their relevance to the alterations found in depression or PTSD in humans.
Collapse
Affiliation(s)
- Vasiliy V. Reshetnikov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Avenue, Sochi 354340, Russia
- Correspondence: ; Tel.: +7-913-715-0695
| | - Polina E. Kisaretova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia
| | - Natalia P. Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia
| |
Collapse
|
29
|
Sim G, Kehling AC, Park MS, Secor J, Divoky C, Zhang H, Malhotra N, Bhagdikar D, El-wahaband EA, Nakanishi K. Manganese-dependent microRNA trimming by 3’→5’ exonucleases generates 14-nucleotide or shorter tiny RNAs.. [DOI: 10.1101/2022.10.06.511180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractMicroRNAs (miRNAs) are about 22-nucleotide (nt) non-coding RNAs forming the effector complexes with Argonaute (AGO) proteins to repress gene expression. Although tiny RNAs (tyRNAs) shorter than 19 nt have been found to bind to plant and vertebrate AGOs, their biogenesis remains a long-standing question. Here, our in vivo and in vitro studies show several 3’→5’ exonucleases, such as interferon-stimulated gene 20 kDa (ISG20), three prime repair exonuclease 1 (TREX1), and ERI1 (enhanced RNAi, also known as 3’hExo), capable of trimming AGO-associated full-length miRNAs to 14 nt or shorter tyRNAs. Their guide trimming occurs in a manganese-dependent manner but independently of the guide sequence and the loaded four human AGO paralogs. We also show that ISG20-mediated guide trimming makes Argonaute3 (AGO3) a slicer. Given the high Mn2+ concentrations in stressed cells, virus-infected cells, and neurodegeneration, our study sheds light on the roles of the Mn2+-dependent exonucleases in remodeling gene silencing.
Collapse
|
30
|
Helman TJ, Headrick JP, Peart JN, Stapelberg NJC. Central and cardiac stress resiliences consistently linked to integrated immuno-neuroendocrine responses across stress models in male mice. Eur J Neurosci 2022; 56:4333-4362. [PMID: 35763309 DOI: 10.1111/ejn.15747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/29/2022]
Abstract
Stress resilience, and behavioural and cardiovascular impacts of chronic stress, are theorised to involve integrated neuro-endocrine/inflammatory/transmitter/trophin signalling. We tested for this integration, and whether behaviour/emotionality, together with myocardial ischaemic tolerance, are consistently linked to these pathways across diverse conditions in male C57Bl/6 mice. This included: Restraint Stress (RS), 1 hr restraint/day for 14 days; Chronic Unpredictable Mild Stress (CUMS), 7 stressors randomised over 21 days; Social Stress (SS), 35 days social isolation with brief social encounters in final 13 days; and Control conditions (CTRL; un-stressed mice). Behaviour was assessed via open field (OFT) and sucrose preference (SPT) tests, and neurobiology from frontal cortex (FC) and hippocampal transcripts. Endocrine factors, and function and ischaemic tolerance in isolated hearts, were also measured. Model characteristics ranged from no behavioural or myocardial changes with homotypic RS, to increased emotionality and cardiac ischaemic injury (with apparently distinct endocrine/neurobiological profiles) in CUMS and SS models. Highly integrated expression of HPA axis, neuro-inflammatory, BDNF, monoamine, GABA, cannabinoid and opioid signalling genes was confirmed across conditions, and consistent/potentially causal correlations identified for: i) Locomotor activity (noradrenaline, ghrelin; FC Crhr1, Tnfrsf1b, Il33, Nfkb1, Maoa, Gabra1; hippocampal Il33); ii) Thigmotaxis (adrenaline, leptin); iii) Anxiety-like behaviour (adrenaline, leptin; FC Tnfrsf1a; hippocampal Il33); iv) Depressive-like behaviour (ghrelin; FC/hippocampal s100a8); and v) Cardiac stress-resistance (noradrenaline, leptin; FC Il33, Tnfrsf1b, Htr1a, Gabra1, Gabrg2; hippocampal Il33, Tnfrsf1a, Maoa, Drd2). Data support highly integrated pathway responses to stress, and consistent adipokine, sympatho-adrenergic, inflammatory and monoamine involvement in mood and myocardial disturbances across diverse conditions.
Collapse
Affiliation(s)
- Tessa J Helman
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
| | - John P Headrick
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
| | - Jason N Peart
- School of Pharmacy and Medical Science, Griffith University, Southport, Australia
| | - Nicolas J C Stapelberg
- Faculty of Health Sciences and Medicine, Bond University, Robina, Australia.,Gold Coast Hospital and Health Service, Southport, Australia
| |
Collapse
|
31
|
Sun Y, Wang Z, Hou J, Shi J, Tang Z, Wang C, Zhao H. Shuangxinfang Prevents S100A9-Induced Macrophage/Microglial Inflammation to Improve Cardiac Function and Depression-Like Behavior in Rats After Acute Myocardial Infarction. Front Pharmacol 2022; 13:832590. [PMID: 35814253 PMCID: PMC9263923 DOI: 10.3389/fphar.2022.832590] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/06/2022] [Indexed: 01/07/2023] Open
Abstract
Background: Depression is a common complication of cardiovascular disease, which deteriorates cardiac function. Shuangxinfang (psycho-cardiology formula, PCF) was reported to alleviate myocardial ischemia injury and improve depression-like behavior. Interestingly, our previous proteomics study predicted that the protein S100A9 appeared as an important target, and macrophage/microglial inflammation might be involved in the process of PCF improving depression induced by acute myocardial infarction (AMI). This study aims to validate the proteomics results. Methods: AMI rat models were established in vivo, followed by the administration of PCF or ABR-215757 (also named paquinimod, inhibiting S100A9 binding to TLR4) for 5 days. Forced swimming test (FST) and open field test (OFT) were applied to record depression-like behavior, and echocardiography was employed to evaluate cardiac function. Morphological changes of cardiomyocytes were assessed by HE staining and TUNEL staining on day 7 after cardiac surgery, as well as Masson trichrome staining on day 21. Hippocampal neurogenesis was determined by Nissl staining, while 5-hydroxytryptamine (5-HT), tryptophan/kynurenine ratio, and brain-derived neurotrophic factor (BDNF) in the hippocampus were analyzed as biochemical indicators of depression. We employed RT-qPCR, western blotting, and immunofluorescence to detect the expression of pathway-related genes and proteins. Myocardial and hippocampal expression of inflammatory factors were performed by ELISA. The activation of macrophage and microglia was assessed via immunoreaction using CD68 and Iba1, respectively. For in vitro confirmation, BV2 cells were primed with recombinant protein S100A9 and then treated with PCF serum or ferulic acid to determine alterations in microglial inflammation. Results: Rats in the AMI group showed heart function deterioration and depression-like behavior. Coronary ligation not only brought about myocardial inflammation, cell apoptosis, and fibrosis but also reduced the neurogenesis, elevated the tryptophan/kynurenine ratio, and decreased the content of 5-HT. PCF could ameliorate the pathological and phenotypic changes in the heart and brain and inhibit the expression of the S100A9 protein, the activation of the microglial cell, and the secretion of IL-1β and TNF-α raised by AMI. ABR-215757 showed therapeutic effect and molecular biological mechanisms similar to PCF. Treatment with PCF serum or ferulic acid in vitro was proved to efficiently block the hyperactivation of BV2 cells and increment of cytokine contents induced by recombinant protein S100A9. Conclusion: We identify S100A9 as a novel and potent regulator of inflammation in both the heart and brain. Macrophage/microglia inflammation mediated by S100A9 is considered a pivotal pathogenic in depression after AMI and a major pathway for the treatment of PCF, suggesting that PCF is a promising therapeutic candidate for psycho-cardiology disease.
Collapse
Affiliation(s)
- Yize Sun
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zheyi Wang
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiqiu Hou
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinyu Shi
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhuoran Tang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wang
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Chao Wang, ; Haibin Zhao,
| | - Haibin Zhao
- Oriental Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Chao Wang, ; Haibin Zhao,
| |
Collapse
|
32
|
Jaszczyk A, Stankiewicz AM, Juszczak GR. Dissection of Mouse Hippocampus with Its Dorsal, Intermediate and Ventral Subdivisions Combined with Molecular Validation. Brain Sci 2022; 12:brainsci12060799. [PMID: 35741684 PMCID: PMC9221087 DOI: 10.3390/brainsci12060799] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Many research methods applied in molecular neuroscience require the collection of hippocampal samples, but a still poorly recognized problem is contamination with the choroid plexus during brain dissection. Because of a distinct pattern of gene expression, its inclusion in brain samples can obscure or even confound conclusions drawn from molecular studies. Therefore, we tested our dissection method designed for removal of tissue contamination using expression of the transthyretin gene (Ttr) as a marker of the choroid plexus. Additionally, we also validated dissection of the entire hippocampus into its dorsal, intermediate and ventral subdivisions using the expression of Trhr and Lct genes as molecular markers of anatomical subdivisions. The PCR analysis showed that Ttr is expressed at a residual level in hippocampal samples that display an mRNA level several hundred lower than the adjacent control tissue colocalized with the choroid plexus. This indicates that the applied method for dissecting the hippocampus from a fresh brain allows for replicable removal of the majority of choroid plexus from hippocampal samples. In turn, differences in expression of Lct and Trhr confirmed the proper dissection of dorsal, intermediate and ventral subdivisions from fresh brain tissue. Therefore, a special emphasis on the removal of tissue contamination and avoidance of tissue distortions makes our protocol especially suitable for molecular experiments performed either on the entire hippocampus or its subdivisions.
Collapse
Affiliation(s)
- Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland;
| | - Adrian M. Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland;
| | - Grzegorz R. Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzebiec, Poland;
- Correspondence: or ; Tel.: +48-22736-7083
| |
Collapse
|
33
|
Sayson LV, Kim M, Jeon SJ, Custodio RJP, Lee HJ, Ortiz DM, Cheong JH, Kim HJ. Differentially Expressed Genes in Period 2-Overexpressing Mice Striatum May Underlie Their Lower Sensitivity to Methamphetamine Addiction-Like Behavior. Biomol Ther (Seoul) 2022; 30:238-245. [PMID: 35477688 PMCID: PMC9047490 DOI: 10.4062/biomolther.2021.184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022] Open
Abstract
Previous reports have demonstrated that genetic mechanisms greatly mediate responses to drugs of abuse, including methamphetamine (METH). The circadian gene Period 2 (Per2) has been previously associated with differential responses towards METH in mice. While the behavioral consequences of eliminating Per2 have been illustrated previously, Per2 overexpression has not yet been comprehensively described; although, Per2-overexpressing (Per2 OE) mice previously showed reduced sensitivity towards METH-induced addiction-like behaviors. To further elucidate this distinct behavior of Per2 OE mice to METH, we identified possible candidate biomarkers by determining striatal differentially expressed genes (DEGs) in both drug-naïve and METH-treated Per2 OE mice relative to wild-type (WT), through RNA sequencing. Of the several DEGs in drug naïve Per2 OE mice, we identified six genes that were altered after repeated METH treatment in WT mice, but not in Per2 OE mice. These results, validated by quantitative real-time polymerase chain reaction, could suggest that the identified DEGs might underlie the previously reported weaker METH-induced responses of Per2 OE mice compared to WT. Gene network analysis also revealed that Asic3, Hba-a1, and Rnf17 are possibly associated with Per2 through physical interactions and predicted correlations, and might potentially participate in addiction. Inhibiting the functional protein of Asic3 prior to METH administration resulted in the partial reduction of METH-induced conditioned place preference in WT mice, supporting a possible involvement of Asic3 in METH-induced reward. Although encouraging further investigations, our findings suggest that these DEGs, including Asic3, may play significant roles in the lower sensitivity of Per2 OE mice to METH.
Collapse
Affiliation(s)
- Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Mikyung Kim
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | | | - Hyun Jun Lee
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Darlene Mae Ortiz
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Jae Hoon Cheong
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
34
|
von Ziegler LM, Floriou-Servou A, Waag R, Das Gupta RR, Sturman O, Gapp K, Maat CA, Kockmann T, Lin HY, Duss SN, Privitera M, Hinte L, von Meyenn F, Zeilhofer HU, Germain PL, Bohacek J. Multiomic profiling of the acute stress response in the mouse hippocampus. Nat Commun 2022; 13:1824. [PMID: 35383160 PMCID: PMC8983670 DOI: 10.1038/s41467-022-29367-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 03/11/2022] [Indexed: 12/26/2022] Open
Abstract
The acute stress response mobilizes energy to meet situational demands and re-establish homeostasis. However, the underlying molecular cascades are unclear. Here, we use a brief swim exposure to trigger an acute stress response in mice, which transiently increases anxiety, without leading to lasting maladaptive changes. Using multiomic profiling, such as proteomics, phospho-proteomics, bulk mRNA-, single-nuclei mRNA-, small RNA-, and TRAP-sequencing, we characterize the acute stress-induced molecular events in the mouse hippocampus over time. Our results show the complexity and specificity of the response to acute stress, highlighting both the widespread changes in protein phosphorylation and gene transcription, and tightly regulated protein translation. The observed molecular events resolve efficiently within four hours after initiation of stress. We include an interactive app to explore the data, providing a molecular resource that can help us understand how acute stress impacts brain function in response to stress. Acute stress can help individuals to respond to challenging events, although chronic stress leads to maladaptive changes. Here, the authors present a multi omic analysis profiling acute stress-induced changes in the mouse hippocampus, providing a resource for the scientific community.
Collapse
Affiliation(s)
- Lukas M von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Rebecca R Das Gupta
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Katharina Gapp
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Christina A Maat
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Tobias Kockmann
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Han-Yu Lin
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Sian N Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Laura Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Hanns U Zeilhofer
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Pierre-Luc Germain
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.,Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.,Laboratory of Statistical Bioinformatics, Department for Molecular Life Sciences, University of Zürich, Zurich, Switzerland
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland. .,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Gómez-Arnaiz S, Tate RJ, Grant MH. Cobalt Neurotoxicity: Transcriptional Effect of Elevated Cobalt Blood Levels in the Rodent Brain. TOXICS 2022; 10:toxics10020059. [PMID: 35202246 PMCID: PMC8878729 DOI: 10.3390/toxics10020059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/11/2022]
Abstract
Metal-on-metal (MoM) hip implants made of cobalt chromium (CoCr) alloy have shown early failure compared with other bearing materials. A consequence of the abnormal wear produced by these prostheses is elevated levels of cobalt in the blood of patients, which can lead to systemic conditions involving cardiac and neurological symptoms. In order to better understand the implications for patients with these implants, we carried out metal content and RNA-Seq analysis of excised tissue from rats treated intraperitonially for 28 days with low concentrations of cobalt. Cobalt blood levels in dosed rats were found to be similar to those seen in some patients with MoM implants (range: 4–38 μg/L Co in blood). Significant accumulation of cobalt was measured in a range of tissues including kidney, liver, and heart, but also in brain tissue. RNA-Seq analysis of neural tissue revealed that exposure to cobalt induces a transcriptional response in the prefrontal cortex (pref. cortex), cerebellum, and hippocampus. Many of the most up- and downregulated genes appear to correspond to choroid plexus transcripts. These results indicate that the choroid plexus could be the brain tissue most affected by cobalt. More specifically, the differentially expressed genes show a disruption of steroidogenesis and lipid metabolism. Several other transcripts also demonstrate that cobalt induces an immune response. In summary, cobalt exposure induces alterations in the brain transcriptome, more specifically, the choroid plexus, which is in direct contact with neurotoxicants at the blood–cerebrospinal fluid barrier.
Collapse
Affiliation(s)
- Sara Gómez-Arnaiz
- Wolfson Centre, Biomedical Engineering Department, University of Strathclyde, Glasgow G4 0NW, UK;
| | - Rothwelle J. Tate
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
| | - Mary Helen Grant
- Wolfson Centre, Biomedical Engineering Department, University of Strathclyde, Glasgow G4 0NW, UK;
- Correspondence:
| |
Collapse
|
36
|
Camilli C, Hoeh AE, De Rossi G, Moss SE, Greenwood J. LRG1: an emerging player in disease pathogenesis. J Biomed Sci 2022; 29:6. [PMID: 35062948 PMCID: PMC8781713 DOI: 10.1186/s12929-022-00790-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
The secreted glycoprotein leucine-rich α-2 glycoprotein 1 (LRG1) was first described as a key player in pathogenic ocular neovascularization almost a decade ago. Since then, an increasing number of publications have reported the involvement of LRG1 in multiple human conditions including cancer, diabetes, cardiovascular disease, neurological disease, and inflammatory disorders. The purpose of this review is to provide, for the first time, a comprehensive overview of the LRG1 literature considering its role in health and disease. Although LRG1 is constitutively expressed by hepatocytes and neutrophils, Lrg1-/- mice show no overt phenotypic abnormality suggesting that LRG1 is essentially redundant in development and homeostasis. However, emerging data are challenging this view by suggesting a novel role for LRG1 in innate immunity and preservation of tissue integrity. While our understanding of beneficial LRG1 functions in physiology remains limited, a consistent body of evidence shows that, in response to various inflammatory stimuli, LRG1 expression is induced and directly contributes to disease pathogenesis. Its potential role as a biomarker for the diagnosis, prognosis and monitoring of multiple conditions is widely discussed while dissecting the mechanisms underlying LRG1 pathogenic functions. Emphasis is given to the role that LRG1 plays as a vasculopathic factor where it disrupts the cellular interactions normally required for the formation and maintenance of mature vessels, thereby indirectly contributing to the establishment of a highly hypoxic and immunosuppressive microenvironment. In addition, LRG1 has also been reported to affect other cell types (including epithelial, immune, mesenchymal and cancer cells) mostly by modulating the TGFβ signalling pathway in a context-dependent manner. Crucially, animal studies have shown that LRG1 inhibition, through gene deletion or a function-blocking antibody, is sufficient to attenuate disease progression. In view of this, and taking into consideration its role as an upstream modifier of TGFβ signalling, LRG1 is suggested as a potentially important therapeutic target. While further investigations are needed to fill gaps in our current understanding of LRG1 function, the studies reviewed here confirm LRG1 as a pleiotropic and pathogenic signalling molecule providing a strong rationale for its use in the clinic as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Carlotta Camilli
- Institute of Ophthalmology, University College London, London, UK.
| | - Alexandra E Hoeh
- Institute of Ophthalmology, University College London, London, UK
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London, UK
| | - Stephen E Moss
- Institute of Ophthalmology, University College London, London, UK
| | - John Greenwood
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
37
|
Craig CF, Filippone RT, Stavely R, Bornstein JC, Apostolopoulos V, Nurgali K. Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease. J Neuroinflammation 2022; 19:4. [PMID: 34983592 PMCID: PMC8729103 DOI: 10.1186/s12974-021-02354-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with inflammatory bowel disease (IBD) suffer from depression at higher rates than the general population. An etiological trigger of depressive symptoms is theorised to be inflammation within the central nervous system. It is believed that heightened intestinal inflammation and dysfunction of the enteric nervous system (ENS) contribute to impaired intestinal permeability, which facilitates the translocation of intestinal enterotoxins into the blood circulation. Consequently, these may compromise the immunological and physiological functioning of distant non-intestinal tissues such as the brain. In vivo models of colitis provide evidence of increased blood–brain barrier permeability and enhanced central nervous system (CNS) immune activity triggered by intestinal enterotoxins and blood-borne inflammatory mediators. Understanding the immunological, physiological, and structural changes associated with IBD and neuroinflammation may aid in the development of more tailored and suitable pharmaceutical treatment for IBD-associated depression.
Collapse
Affiliation(s)
- Colin F Craig
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhiannon T Filippone
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Joel C Bornstein
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Vasso Apostolopoulos
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia. .,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia. .,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia. .,Institute for Health and Sport, Victoria University, Level 4 Research Labs, Western Centre for Health Research and Education, Sunshine Hospital, 176 Furlong Road, St Albans, VIC, 3021, Australia.
| |
Collapse
|
38
|
Sun Y, Wang Z, Wang C, Tang Z, Zhao H. Psycho-cardiology therapeutic effects of Shuangxinfang in rats with depression-behavior post acute myocardial infarction: Focus on protein S100A9 from proteomics. Biomed Pharmacother 2021; 144:112303. [PMID: 34673424 DOI: 10.1016/j.biopha.2021.112303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Depressive disorders induced by acute myocardial infarction (AMI) play a pivotal role in the deterioration of cardiac function, and Shuangxinfang (Psycho-cardiology Formula, PCF) was reported to alleviate heart function damage and improve depression-like behavior, but the complex mechanism in such process has not been clarified. METHODS AMI models were established and PCF was administered in rats. Subjects were then assessed in open field test (OFT) and forced swimming test (FST) recapitulating symptoms of depressive disorder. Afterward, pharmacoproteomic profiling of the hippocampus and peri-infarct border zone (BZ) was performed using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) technique, to identify contributing proteins and pathways responsible for myocardial ischemia and behavioral allostasis. Bioinformatics analysis was processed for further investigation, while western blotting was employed for testing dominating proteins to validate proteomic results. RESULTS Rats in the AMI group showed depression-like behavior in OFT and FST, which was improved by PCF. There were 131 differentially expressed proteins (DEPs) in BZ and 64 proteins in the hippocampus being detected and quantified shared by the sham group, the AMI group, and the PCF group. Subsequently, pertinent pathways and molecular functions were further identified. Altered molecules were discovered to be enriched in the apoptotic process, innate immune response, and NF-κB transcription factor activity in BZ, as well as chemical synaptic transmission, axon, collagen binding, cell adhesion, response to carbohydrate, laminin binding, and cellular response to nitric oxide in the hippocampus. Groups of signal transducers were also able to select multiple pathways, including innate immunity and arginine biosynthesis in the heart, also integrin signaling in the brain. DEPs were intersected from the myocardium and hippocampus to screen out the protein S100A9, which was up-regulated in the AMI group compared with the sham, and showed a down-regulation trend after treatment with PCF. CONCLUSION Taken together, we present a comprehensive proteomics analysis of rat models with depression post-AMI. Reviewing the literatures concerned, it's hypothesized that macrophage/microglia inflammation mediated by S100A9 might be the pivotal pathogenic process of psycho-cardiology disease, as well as potential mechanisms for the treatment of PCF.
Collapse
Affiliation(s)
- Yize Sun
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zheyi Wang
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Chunguo Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhuoran Tang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haibin Zhao
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
39
|
Talley S, Valiauga R, Anderson L, Cannon AR, Choudhry MA, Campbell EM. DSS-induced inflammation in the colon drives a proinflammatory signature in the brain that is ameliorated by prophylactic treatment with the S100A9 inhibitor paquinimod. J Neuroinflammation 2021; 18:263. [PMID: 34758843 PMCID: PMC8578918 DOI: 10.1186/s12974-021-02317-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/03/2021] [Indexed: 12/23/2022] Open
Abstract
Background Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and neuroinflammation remains unclear and characterization of the neuroinflammatory phenotype in the brain during colitis is ill-defined. Methods Transgenic mice expressing a bioluminescent reporter of active caspase-1 were treated with 2% dextran sodium sulfate (DSS) for 7 days to induce acute colitis, and colonic, systemic and neuroinflammation were assessed. In some experiments, mice were prophylactically treated with paquinimod (ABR-215757) to inhibit S100A9 inflammatory signaling. As a positive control for peripheral-induced neuroinflammation, mice were injected with lipopolysaccharide (LPS). Colonic, systemic and brain inflammatory cytokines and chemokines were measured by cytokine bead array (CBA) and Proteome profiler mouse cytokine array. Bioluminescence was quantified in the brain and caspase activation was confirmed by immunoblot. Immune cell infiltration into the CNS was measured by flow cytometry, while light sheet microscopy was used to monitor changes in resident microglia localization in intact brains during DSS or LPS-induced neuroinflammation. RNA sequencing was performed to identify transcriptomic changes occurring in the CNS of DSS-treated mice. Expression of inflammatory biomarkers were quantified in the brain and serum by qRT-PCR, ELISA and WB. Results DSS-treated mice exhibited clinical hallmarks of colitis, including weight loss, colonic shortening and inflammation in the colon. We also detected a significant increase in inflammatory cytokines in the serum and brain, as well as caspase and microglia activation in the brain of mice with ongoing colitis. RNA sequencing of brains isolated from DSS-treated mice revealed differential expression of genes involved in the regulation of inflammatory responses. This inflammatory phenotype was similar to the signature detected in LPS-treated mice, albeit less robust and transient, as inflammatory gene expression returned to baseline following cessation of DSS. Pharmacological inhibition of S100A9, one of the transcripts identified by RNA sequencing, attenuated colitis severity and systemic and neuroinflammation. Conclusions Our findings suggest that local inflammation in the colon drives systemic inflammation and neuroinflammation, and this can be ameliorated by inhibition of the S100 alarmin, S100A9. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02317-6.
Collapse
Affiliation(s)
- Sarah Talley
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| | - Rasa Valiauga
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Lillian Anderson
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Science Division, Maywood, IL, USA
| | - Abigail R Cannon
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Science Division, Maywood, IL, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Science Division, Maywood, IL, USA
| | - Edward M Campbell
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA. .,Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
40
|
Sturman O, von Ziegler L, Privitera M, Waag R, Duss S, Vermeiren Y, Giovagnoli L, de Deyn P, Bohacek J. Chronic adolescent stress increases exploratory behavior but does not appear to change the acute stress response in adult male C57BL/6 mice. Neurobiol Stress 2021; 15:100388. [PMID: 34527792 PMCID: PMC8430388 DOI: 10.1016/j.ynstr.2021.100388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/26/2022] Open
Abstract
Chronic stress exposure in adolescence can lead to a lasting change in stress responsiveness later in life and is associated with increased mental health issues in adulthood. Here we investigate whether the Chronic Social Instability (CSI) paradigm influences the behavioral and molecular responses to novel acute stressors in mice, and whether it alters physiological responses influenced by the noradrenergic system. Using large cohorts of mice, we show that CSI mice display a persistent increase in exploratory behaviors in the open field test alongside small but widespread transcriptional changes in the ventral hippocampus. However, both the transcriptomic and behavioral responses to novel acute stressors are indistinguishable between groups. In addition, the pupillometric response to a tail shock, known to be mediated by the noradrenergic system, remains unaltered in CSI mice. Ultra-high performance liquid chromatography analysis of monoaminergic neurotransmitter levels in the ventral hippocampus also shows no differences between control or CSI mice at baseline or in response to acute stress. We conclude that CSI exposure during adolescence leads to persistent changes in exploratory behavior and gene expression in the hippocampus, but it does not alter the response to acute stress in adulthood and is unlikely to alter the function of the noradrenergic system.
Collapse
Affiliation(s)
- Oliver Sturman
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Sian Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Yannick Vermeiren
- Department of Biomedical Sciences, Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Antwerp, Belgium
- Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University & Research, Wageningen, Netherlands
- Faculty of Medicine & Health Sciences, Translational Neurosciences, University of Antwerp, Antwerp, Belgium
| | - Letizia Giovagnoli
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| | - Peter de Deyn
- Department of Biomedical Sciences, Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Antwerp, Belgium
- Department of Neurology and Alzheimer Center, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
- Department of Neurology, Memory Clinic of Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Switzerland
| |
Collapse
|
41
|
Montagud-Romero S, Miñarro J, Rodríguez-Arias M. Unravelling the Neuroinflammatory Mechanisms Underlying the Effects of Social Defeat Stress on Use of Drugs of Abuse. Curr Top Behav Neurosci 2021; 54:153-180. [PMID: 34628585 DOI: 10.1007/7854_2021_260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The immune system provides the first line of the organism's defenses, working to maintain homeostasis against external threats and respond also to internal danger signals. There is much evidence to suggest that modifications of inflammatory parameters are related to vulnerability to develop mental illnesses, such as depression, autism, schizophrenia, and substance use disorders. In addition, not only are inflammatory parameters related to these disorders, but stress also induces the activation of the immune system, as recent preclinical research demonstrates. Social stress activates the immune response in the central nervous system through a number of mechanisms; for example, by promoting microglial stimulation, modifying peripheral and brain cytokine levels, and altering the blood brain barrier, which allows monocytes to traffic into the brain. In this chapter, we will first deal with the most important short- and long-term consequences of social defeat (SD) stress on the neuroinflammatory response. SD experiences (brief episodes of social confrontations during adolescence and adulthood) induce functional modifications in the brain, which are accompanied by an increase in proinflammatory markers. Most importantly, inflammatory mechanisms play a significant role in mediating the process of adaptation in the face of adversity (resilience vs susceptibility), allowing us to understand individual differences in stress responses. Secondly, we will address the role of the immune system in the vulnerability and enhanced sensitivity to drugs of abuse after social stress. We will explore in depth the effects seen in the inflammatory system in response to social stress and how they enhance the rewarding effects of drugs such as alcohol or cocaine. To conclude, we will consider pharmacological and environmental interventions that seek to influence the inflammatory response to social stress and diminish increased drug intake, as well as the translational potential and future directions of this exciting new field of research.
Collapse
Affiliation(s)
- S Montagud-Romero
- Department of Psychology and Sociology, University of Zaragoza, Teruel, Spain
| | - J Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - M Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain. .,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
42
|
Chen YH, Xie SY, Chen CW, Lu DY. Electroacupuncture improves repeated social defeat stress-elicited social avoidance and anxiety-like behaviors by reducing Lipocalin-2 in the hippocampus. Mol Brain 2021; 14:150. [PMID: 34565419 PMCID: PMC8474847 DOI: 10.1186/s13041-021-00860-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/13/2021] [Indexed: 12/28/2022] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is a trauma-related disorder that is associated with pro-inflammatory activation and neurobiological impairments in the brain and leads to a series of affective-like behaviors. Electroacupuncture (EA) has been proposed as a clinically useful therapy for several brain diseases. However, the potential role of EA treatment in PTSD and its molecular and cellular mechanisms has rarely been investigated. Methods We used an established preclinical social defeat stress mouse model to study whether EA treatment modulates PTSD-like symptoms and understand its underlying mechanisms. To this end, male C57BL/6 mice were subjected to repeated social defeat stress (RSDS) for 6 consecutive days to induce symptoms of PTSD and treated with EA at Baihui (GV 20) and Dazhui (GV 14) acupoints. Results The stimulation of EA, but not needle insertion at Baihui (GV 20) and Dazhui (GV 14) acupoints effectively improved PTSD-like behaviors such as, social avoidance and anxiety-like behaviors. However, EA stimulation at the bilateral Tianzong (SI11) acupoints did not affect the PTSD-like behaviors obtained by RSDS. EA stimulation also markedly inhibited astrocyte activation in both the dorsal and ventral hippocampi of RSDS-treated mice. Using next-generation sequencing analysis, our results showed that EA stimulation attenuated RSDS-enhanced lipocalin 2 expression in the hippocampus. Importantly, using double-staining immunofluorescence, we observed that the increased lipocalin 2 expression in astrocytes by RSDS was also reduced by EA stimulation. In addition, intracerebroventricular injection of mouse recombinant lipocalin 2 protein in the lateral ventricles provoked social avoidance, anxiety-like behaviors, and the activation of astrocytes in the hippocampus. Interestingly, the overexpression of lipocalin 2 in the brain also altered the expression of stress-related genes, including monoamine oxidase A, monoamine oxidase B, mineralocorticoid receptor, and glucocorticoid receptor in the hippocampus. Conclusions This study suggests that the treatment of EA at Baihui (GV 20) and Dazhui (GV 14) acupoints improves RSDS-induced social avoidance, anxiety-like behaviors, astrocyte activation, and lipocalin 2 expression. Furthermore, our findings also indicate that lipocalin 2 expression in the brain may be an important biomarker for the development of PTSD-related symptoms. Supplementary Information The online version contains supplementary material available at 10.1186/s13041-021-00860-0.
Collapse
Affiliation(s)
- Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Sheng-Yun Xie
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chao-Wei Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan. .,Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| |
Collapse
|
43
|
Glavan D, Gheorman V, Gresita A, Hermann DM, Udristoiu I, Popa-Wagner A. Identification of transcriptome alterations in the prefrontal cortex, hippocampus, amygdala and hippocampus of suicide victims. Sci Rep 2021; 11:18853. [PMID: 34552157 PMCID: PMC8458545 DOI: 10.1038/s41598-021-98210-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 08/30/2021] [Indexed: 11/09/2022] Open
Abstract
Suicide is one of the leading causes of death globally for all ages, and as such presents a very serious problem for clinicians worldwide. However, the underlying neurobiological pathology remains to a large extent unknown. In order to address this gap, we have carried out a genome-wide investigation of the gene expression in the amygdala, hippocampus, prefrontal cortex and thalamus in post-mortem brain samples obtained from 20 suicide completers and 7 control subjects. By KEGG enrichment analysis indicated we identified novel clusters of downregulated pathways involved in antigen neutralization and autoimmune thyroid disease (amygdala, thalamus), decreased axonal plasticity in the hippocampus. Two upregulated pathways were involved in neuronal death in the hippocampus and olfactory transduction in the thalamus and the prefrontal cortex. Autoimmune thyroid disease pathway was downregulated only in females. Metabolic pathways involved in Notch signaling amino acid metabolism and unsaturated lipid synthesis were thalamus-specific. Suicide-associated changes in the expression of several genes and pseudogenes that point to various functional mechanisms possibly implicated in the pathology of suicide. Two genes (SNORA13 and RNU4-2) involved in RNA processing were common to all brain regions analyzed. Most of the identified gene expression changes were related to region-specific dysregulated manifestation of genetic and epigenetic mechanisms underlying neurodevelopmental disorders (SNORD114-10, SUSd1), motivation, addiction and motor disorders (CHRNA6), long-term depression (RAB3B), stress response, major depression and schizophrenia (GFAP), signal transduction at the neurovascular unit (NEXN) and inhibitory neurotransmission in spatial learning, neural plasticity (CALB2; CLIC6, ENPP1). Some of the differentially expressed genes were brain specific non-coding RNAs involved in the regulation of translation (SNORA13). One, (PARM1) is a potential oncogene and prognostic biomarker for colorectal cancer with no known function in the brain. Disturbed gene expression involved in antigen neutralization, autoimmunity, neural plasticity, stress response, signal transduction at the neurovascular unit, dysregulated nuclear RNA processing and translation and epigenetic imprinting signatures is associated with suicide and point to regulatory non-coding RNAs as potential targets of new drugs development.
Collapse
Affiliation(s)
- Daniela Glavan
- Department of Psychiatry, University of Medicine and Pharmacy, Craiova, Romania
| | - Victor Gheorman
- Department of Psychiatry, University of Medicine and Pharmacy, Craiova, Romania
| | - Andrei Gresita
- Griffith University Menzies Health Institute of Queensland, Gold Coast Campus, Brisbane, QLD, 4000, Australia
| | - Dirk M Hermann
- Chair of Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg, Essen, Germany
| | - Ion Udristoiu
- Department of Psychiatry, University of Medicine and Pharmacy, Craiova, Romania.
| | - Aurel Popa-Wagner
- Griffith University Menzies Health Institute of Queensland, Gold Coast Campus, Brisbane, QLD, 4000, Australia. .,Chair of Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg, Essen, Germany.
| |
Collapse
|
44
|
Karunakaran KB, Amemori S, Balakrishnan N, Ganapathiraju MK, Amemori KI. Generalized and social anxiety disorder interactomes show distinctive overlaps with striosome and matrix interactomes. Sci Rep 2021; 11:18392. [PMID: 34526518 PMCID: PMC8443595 DOI: 10.1038/s41598-021-97418-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
Mechanisms underlying anxiety disorders remain elusive despite the discovery of several associated genes. We constructed the protein-protein interaction networks (interactomes) of six anxiety disorders and noted enrichment for striatal expression among common genes in the interactomes. Five of these interactomes shared distinctive overlaps with the interactomes of genes that were differentially expressed in two striatal compartments (striosomes and matrix). Generalized anxiety disorder and social anxiety disorder interactomes showed exclusive and statistically significant overlaps with the striosome and matrix interactomes, respectively. Systematic gene expression analysis with the anxiety disorder interactomes constrained to contain only those genes that were shared with striatal compartment interactomes revealed a bifurcation among the disorders, which was influenced by the anterior cingulate cortex, nucleus accumbens, amygdala and hippocampus, and the dopaminergic signaling pathway. Our results indicate that the functionally distinct striatal pathways constituted by the striosome and the matrix may influence the etiological differentiation of various anxiety disorders.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | - Satoko Amemori
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - N Balakrishnan
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | - Madhavi K Ganapathiraju
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, USA.
- Intelligent Systems Program, School of Computing and Information, University of Pittsburgh, Pittsburgh, USA.
| | - Ken-Ichi Amemori
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan.
| |
Collapse
|
45
|
Dekens DW, Eisel ULM, Gouweleeuw L, Schoemaker RG, De Deyn PP, Naudé PJW. Lipocalin 2 as a link between ageing, risk factor conditions and age-related brain diseases. Ageing Res Rev 2021; 70:101414. [PMID: 34325073 DOI: 10.1016/j.arr.2021.101414] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Chronic (neuro)inflammation plays an important role in many age-related central nervous system (CNS) diseases, including Alzheimer's disease, Parkinson's disease and vascular dementia. Inflammation also characterizes many conditions that form a risk factor for these CNS disorders, such as physical inactivity, obesity and cardiovascular disease. Lipocalin 2 (Lcn2) is an inflammatory protein shown to be involved in different age-related CNS diseases, as well as risk factor conditions thereof. Lcn2 expression is increased in the periphery and the brain in different age-related CNS diseases and also their risk factor conditions. Experimental studies indicate that Lcn2 contributes to various neuropathophysiological processes of age-related CNS diseases, including exacerbated neuroinflammation, cell death and iron dysregulation, which may negatively impact cognitive function. We hypothesize that increased Lcn2 levels as a result of age-related risk factor conditions may sensitize the brain and increase the risk to develop age-related CNS diseases. In this review we first provide a comprehensive overview of the known functions of Lcn2, and its effects in the CNS. Subsequently, this review explores Lcn2 as a potential (neuro)inflammatory link between different risk factor conditions and the development of age-related CNS disorders. Altogether, evidence convincingly indicates Lcn2 as a key constituent in ageing and age-related brain diseases.
Collapse
Affiliation(s)
- Doortje W Dekens
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Leonie Gouweleeuw
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Regien G Schoemaker
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Laboratory of Neurochemistry and Behaviour, Biobank, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Petrus J W Naudé
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands; Department of Psychiatry and Mental Health and Neuroscience Institute, Brain Behaviour Unit, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
46
|
Stott RT, Kritsky O, Tsai LH. Profiling DNA break sites and transcriptional changes in response to contextual fear learning. PLoS One 2021; 16:e0249691. [PMID: 34197463 PMCID: PMC8248687 DOI: 10.1371/journal.pone.0249691] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Neuronal activity generates DNA double-strand breaks (DSBs) at specific loci in vitro and this facilitates the rapid transcriptional induction of early response genes (ERGs). Physiological neuronal activity, including exposure of mice to learning behaviors, also cause the formation of DSBs, yet the distribution of these breaks and their relation to brain function remains unclear. Here, following contextual fear conditioning (CFC) in mice, we profiled the locations of DSBs genome-wide in the medial prefrontal cortex and hippocampus using γH2AX ChIP-Seq. Remarkably, we found that DSB formation is widespread in the brain compared to cultured primary neurons and they are predominately involved in synaptic processes. We observed increased DNA breaks at genes induced by CFC in neuronal and non-neuronal nuclei. Activity-regulated and proteostasis-related transcription factors appear to govern some of these gene expression changes across cell types. Finally, we find that glia but not neurons have a robust transcriptional response to glucocorticoids, and many of these genes are sites of DSBs. Our results indicate that learning behaviors cause widespread DSB formation in the brain that are associated with experience-driven transcriptional changes across both neuronal and glial cells.
Collapse
Affiliation(s)
- Ryan T. Stott
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Oleg Kritsky
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
47
|
Floriou-Servou A, von Ziegler L, Waag R, Schläppi C, Germain PL, Bohacek J. The Acute Stress Response in the Multiomic Era. Biol Psychiatry 2021; 89:1116-1126. [PMID: 33722387 DOI: 10.1016/j.biopsych.2020.12.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/13/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022]
Abstract
Studying the stress response is a major pillar of neuroscience research not only because stress is a daily reality but also because the exquisitely fine-tuned bodily changes triggered by stress are a neuroendocrinological marvel. While the genome-wide changes induced by chronic stress have been extensively studied, we know surprisingly little about the complex molecular cascades triggered by acute stressors, the building blocks of chronic stress. The acute stress (or fight-or-flight) response mobilizes organismal energy resources to meet situational demands. However, successful stress coping also requires the efficient termination of the stress response. Maladaptive coping-particularly in response to severe or repeated stressors-can lead to allostatic (over)load, causing wear and tear on tissues, exhaustion, and disease. We propose that deep molecular profiling of the changes triggered by acute stressors could provide molecular correlates for allostatic load and predict healthy or maladaptive stress responses. We present a theoretical framework to interpret multiomic data in light of energy homeostasis and activity-dependent gene regulation, and we review the signaling cascades and molecular changes rapidly induced by acute stress in different cell types in the brain. In addition, we review and reanalyze recent data from multiomic screens conducted mainly in the rodent hippocampus and amygdala after acute psychophysical stressors. We identify challenges surrounding experimental design and data analysis, and we highlight promising new research directions to better understand the stress response on a multiomic level.
Collapse
Affiliation(s)
- Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Christa Schläppi
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Pierre-Luc Germain
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland; Laboratory of Statistical Bioinformatics, Department for Molecular Life Sciences, University of Zürich, Zürich, Switzerland.
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland.
| |
Collapse
|
48
|
Chae S, Hong J, Kang K, Shin A, Kim DG, Lee S, Kim MY, Jung I, Kim D. Molecular laterality encodes stress susceptibility in the medial prefrontal cortex. Mol Brain 2021; 14:92. [PMID: 34127022 PMCID: PMC8201740 DOI: 10.1186/s13041-021-00802-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/02/2021] [Indexed: 01/11/2023] Open
Abstract
Functional lateralization of the prefrontal cortex has been implicated in stress and emotional disorders, yet underlying gene expression changes remains unknown. Here, we report molecular signatures lateralized by chronic social defeats between the two medial prefrontal cortices (mPFCs). Stressed mice show 526 asymmetrically expressed genes between the mPFCs. This cortical asymmetry selectively occurs in stressed mice with depressed social activity, but not in resilient mice with normal behavior. We have isolated highly asymmetric genes including connective tissue growth factor (CTGF), a molecule that modulates wound healing at the periphery. Knockdown of CTGF gene in the right mPFC by shRNA led to a stress-resistant behavioral phenotype. Overexpression of CTGF in the right mPFC using viral transduction induces social avoidance while the left mPFC thereof prevent stress-induced social avoidance. Our study provides a molecular window into the mechanism of stress-induced socioemotional disorders, which can pave the way for new interventions by targeting cortical asymmetry.
Collapse
Affiliation(s)
- Sujin Chae
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Korea
| | - Jiso Hong
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Keunsoo Kang
- Department of Microbiology, College of Natural Sciences, Dankook University, Chungnam, 31116, Korea
| | - Anna Shin
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Dae-Gun Kim
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Korea
| | - Sinjeong Lee
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Moo-Young Kim
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Inkyung Jung
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Daesoo Kim
- Behavioral Genetics Lab., Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea.
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Korea.
| |
Collapse
|
49
|
Rojas-Colón LA, Dash PK, Morales-Vías FA, Lebrón-Dávila M, Ferchmin PA, Redell JB, Maldonado-Martínez G, Vélez-Torres WI. 4R-cembranoid confers neuroprotection against LPS-induced hippocampal inflammation in mice. J Neuroinflammation 2021; 18:95. [PMID: 33874954 PMCID: PMC8054431 DOI: 10.1186/s12974-021-02136-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/22/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Chronic brain inflammation has been implicated in the pathogenesis of various neurodegenerative diseases and disorders. For example, overexpression of pro-inflammatory cytokines has been associated with impairments in hippocampal-dependent memory. Lipopolysaccharide (LPS) injection is a widely used model to explore the pathobiology of inflammation. LPS injection into mice causes systemic inflammation, neuronal damage, and poor memory outcomes if the inflammation is not controlled. Activation of the alpha-7 nicotinic receptor (α7) plays an anti-inflammatory role in the brain through vagal efferent nerve signaling. 4R-cembranoid (4R) is a natural compound that crosses the blood-brain barrier, induces neuronal survival, and has been shown to modulate the activity of nicotinic receptors. The purpose of this study is to determine whether 4R reduces the deleterious effects of LPS-induced neuroinflammation and whether the α7 receptor plays a role in mediating these beneficial effects. METHODS Ex vivo population spike recordings were performed in C57BL/6J wild-type (WT) and alpha-7-knockout (α7KO) mouse hippocampal slices in the presence of 4R and nicotinic receptor inhibitors. For in vivo studies, WT and α7KO mice were injected with LPS for 2 h, followed by 4R or vehicle for 22 h. Analyses of IL-1β, TNF-α, STAT3, CREB, Akt1, and the long-term novel object recognition test (NORT) were performed for both genotypes. In addition, RNA sequencing and RT-qPCR analyses were carried out for 12 mRNAs related to neuroinflammation and their modification by 4R. RESULTS 4R confers neuroprotection after NMDA-induced neurotoxicity in both WT and α7KO mice. Moreover, hippocampal TNF-α and IL-1β levels were decreased with 4R treatment following LPS exposure in both strains of mice. 4R restored LPS-induced cognitive decline in NORT. There was a significant increase in the phosphorylation of STAT3, CREB, and Akt1 with 4R treatment in the WT mouse hippocampus following LPS exposure. In α7KO mice, only pAkt levels were significantly elevated in the cortex. 4R significantly upregulated mRNA levels of ORM2, GDNF, and C3 following LPS exposure. These proteins are known to play a role in modulating microglial activation, neuronal survival, and memory. CONCLUSION Our results indicate that 4R decreases the levels of pro-inflammatory cytokines; improves memory function; activates STAT3, Akt1, and CREB phosphorylation; and upregulates the mRNA levels of ORM2, GDNF, and C3. These effects are independent of the α7 nicotinic receptor.
Collapse
Affiliation(s)
- Luis A Rojas-Colón
- Department of Biochemistry, Universidad Central del Caribe School of Medicine, Av. Sta. Juanita, Bayamón, 00960, Puerto Rico
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Fabiola A Morales-Vías
- Department of Biochemistry, Universidad Central del Caribe School of Medicine, Av. Sta. Juanita, Bayamón, 00960, Puerto Rico
| | - Madeline Lebrón-Dávila
- Department of Biochemistry, Universidad Central del Caribe School of Medicine, Av. Sta. Juanita, Bayamón, 00960, Puerto Rico
| | - Pedro A Ferchmin
- Department of Biochemistry, Universidad Central del Caribe School of Medicine, Av. Sta. Juanita, Bayamón, 00960, Puerto Rico
| | - John B Redell
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Geronimo Maldonado-Martínez
- University of Puerto Rico Molecular Science Research Center, Av. Juan Ponce de León, San Juan, 00926, Puerto Rico
| | - Wanda I Vélez-Torres
- Department of Biochemistry, Universidad Central del Caribe School of Medicine, Av. Sta. Juanita, Bayamón, 00960, Puerto Rico.
| |
Collapse
|
50
|
Bekhbat M, Mukhara D, Dozmorov MG, Stansfield JC, Benusa SD, Hyer MM, Rowson SA, Kelly SD, Qin Z, Dupree JL, Tharp GK, Tansey MG, Neigh GN. Adolescent stress sensitizes the adult neuroimmune transcriptome and leads to sex-specific microglial and behavioral phenotypes. Neuropsychopharmacology 2021; 46:949-958. [PMID: 33558677 PMCID: PMC8115118 DOI: 10.1038/s41386-021-00970-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 01/30/2023]
Abstract
Adolescent exposure to chronic stress, a risk factor for mood disorders in adulthood, sensitizes the neuroinflammatory response to a subsequent immune challenge. We previously showed that chronic adolescent stress (CAS) in rats led to distinct patterns of neuroimmune priming in adult male and female rats. However, sex differences in the neuroimmune consequences of CAS and their underlying mechanisms are not fully understood. Here we hypothesized that biological sex would dictate differential induction of inflammation-related transcriptomic pathways and immune cell involvement (microglia activation and leukocyte presence) in the hippocampus of male and female rats with a history of CAS. Adolescent rats underwent CAS (six restraint and six social defeat episodes during postnatal days 38-49), and behavioral assessments were conducted in adolescence and adulthood. Neuroimmune measures were obtained following vehicle or a systemic lipopolysaccharide (LPS) challenge in adulthood. CAS led to increased time in the corners of the open field in adolescence. In males, CAS also increased social avoidance. As adults, CAS rats displayed an exaggerated enrichment of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) pathway and chemokine induction following LPS challenge, and increased number of perivascular CD45+ cells in the hippocampus. However, CAS females, but not males, showed exaggerated glucocorticoid receptor (GR) pathway enrichment and increased microglial complexity. These results provide further insight to the mechanisms by which peripheral immune events may influence neuroimmune responses differentially among males and females and further demonstrate the importance of adolescent stress in shaping adult responses.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Deepika Mukhara
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Mikhail G. Dozmorov
- grid.417264.20000 0001 2194 2791Center for Clinical and Translational Research, Virginia Commonwealth University Medical Center, Richmond, VA 23298 USA
| | - John C. Stansfield
- grid.417264.20000 0001 2194 2791Center for Clinical and Translational Research, Virginia Commonwealth University Medical Center, Richmond, VA 23298 USA
| | - Savannah D. Benusa
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Molly M. Hyer
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Sydney A. Rowson
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Sean D. Kelly
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Zhaohui Qin
- grid.189967.80000 0001 0941 6502Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Jeffrey L. Dupree
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Gregory K. Tharp
- grid.189967.80000 0001 0941 6502Division of Developmental & Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30322 USA
| | - Malú G. Tansey
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Gretchen N. Neigh
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA ,grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA ,grid.189967.80000 0001 0941 6502Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322 USA
| |
Collapse
|