1
|
Wang P, Hu J, Chen C, Jiang Z, Zhang Y, Lin K, Liao L, Wang X. The immune regulatory mechanism of ketamine-induced psychiatric disorders: A new perspective on drug-induced psychiatric symptoms. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111194. [PMID: 39542202 DOI: 10.1016/j.pnpbp.2024.111194] [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: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Ketamine, a psychoactive substance strictly regulated by international drug conventions, is classified as a "new type drug" due to its excitatory, hallucinogenic, or inhibitory effects. The etiology of ketamine-induced psychiatric symptoms is multifaceted, with the immune regulatory mechanism being the most prominent among several explanatory theories. In recent years, the interaction between the immune system and nervous system have garnered significant attention in neuropsychiatric disorder research. Notably, the infiltration of peripheral lymphocytes into the central nervous system has emerged as an early hallmark of certain neuropsychiatric disorders. However, a notable gap exists in the current literature, regarding the immune regulatory mechanisms, specifically the peripheral immune alterations, associated with ketamine-induced psychiatric symptoms. To address this void, this article endeavors to provide a comprehensive overview of the pathophysiological processes implicated in psychiatric disorders or symptoms, encompassing those elicited by ketamine. This analysis delves into aspects such as nerve damage, alterations within the central immune system, and the regulation of the peripheral immune system. By emphasizing the intricate crosstalk between the peripheral immune system and the central nervous system, this study sheds light on their collaborative role in the onset and progression of psychiatric diseases or symptoms. This insight offers fresh perspectives on the underlying mechanisms, diagnosis and therapeutic strategies for mental disorders stemming from drug abuse.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Kexin Lin
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Sepúlveda P, Ferreira AFF, Sandoval C, Bergoc G, Moreno ACR, Nunes MT, Torrão ADS. Thyroid Hormone Supplementation Restores Cognitive Deficit, Insulin Signaling, and Neuroinflammation in the Hippocampus of a Sporadic Alzheimer's-like Disease Rat Model. Cells 2024; 13:1793. [PMID: 39513900 PMCID: PMC11545223 DOI: 10.3390/cells13211793] [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: 05/27/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 11/16/2024] Open
Abstract
Thyroid hormones play a crucial role in the development of the central nervous system and are considered pivotal to cognitive functions in the adult brain. Recently, thyroid dysfunction has been associated with Alzheimer's disease. The aim of this study was to assess the neuroprotective effects of triiodothyronine (T3) on insulin signaling, neuroinflammation, apoptosis, and cognitive function in a streptozotocin (STZ)-induced sporadic Alzheimer's disease-like model. Male Wistar rats underwent stereotaxic surgery for intracerebroventricular injections of streptozotocin (STZ; 2 mg/kg) or vehicle in the lateral ventricles to induce an AD-like model. The animals received a daily dose of 1.5 μg of T3/100 g body weight or the same volume of vehicle for 30 days and were subdivided into four experimental groups: (1) animals receiving citrate treated with saline (Control = CTL); (2) animals receiving citrate treated with T3 (T3); (3) animals receiving STZ treated with saline (STZ); and (4) animals receiving STZ treated with T3 (STZ + T3). The novel object recognition test was used to measure cognitive function. Serum analysis, real-time RT-PCR, immunohistochemistry, and immunoblotting analyses were also carried out. Our results demonstrated that T3 treatment reversed cognitive impairment and increased Akt and GSK3 phosphorylation in the treated group, while also reducing microglial activation (Iba-1) and GFAP expression (reactive astrocytes), along with TNF-α, IL-6, and IL-1β levels in the hippocampus. Additionally, T3 treatment increased levels of the anti-apoptotic protein Bcl-2 and reduced the expression of the pro-apoptotic protein BAX in the hippocampus. Our study demonstrated that T3 could potentially protect neurons in an AD model induced by STZ.
Collapse
Affiliation(s)
- Paulina Sepúlveda
- Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Ana Flavia Fernandes Ferreira
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Giovanna Bergoc
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Ana Caroline Rippi Moreno
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Maria Tereza Nunes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| | - Andréa da Silva Torrão
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (A.F.F.F.); (G.B.); (A.C.R.M.); (M.T.N.)
| |
Collapse
|
3
|
Sleiman A, Miller KB, Flores D, Kuan J, Altwasser K, Smith BJ, Kozbenko T, Hocking R, Wood SJ, Huff J, Adam-Guillermin C, Hamada N, Yauk C, Wilkins R, Chauhan V. AOP report: Development of an adverse outcome pathway for deposition of energy leading to learning and memory impairment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:57-84. [PMID: 39228295 DOI: 10.1002/em.22622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024]
Abstract
Understanding radiation-induced non-cancer effects on the central nervous system (CNS) is essential for the risk assessment of medical (e.g., radiotherapy) and occupational (e.g., nuclear workers and astronauts) exposures. Herein, the adverse outcome pathway (AOP) approach was used to consolidate relevant studies in the area of cognitive decline for identification of research gaps, countermeasure development, and for eventual use in risk assessments. AOPs are an analytical construct describing critical events to an adverse outcome (AO) in a simplified form beginning with a molecular initiating event (MIE). An AOP was constructed utilizing mechanistic information to build empirical support for the key event relationships (KERs) between the MIE of deposition of energy to the AO of learning and memory impairment through multiple key events (KEs). The evidence for the AOP was acquired through a documented scoping review of the literature. In this AOP, the MIE is connected to the AO via six KEs: increased oxidative stress, increased deoxyribonucleic acid (DNA) strand breaks, altered stress response signaling, tissue resident cell activation, increased pro-inflammatory mediators, and abnormal neural remodeling that encompasses atypical structural and functional alterations of neural cells and surrounding environment. Deposition of energy directly leads to oxidative stress, increased DNA strand breaks, an increase of pro-inflammatory mediators and tissue resident cell activation. These KEs, which are themselves interconnected, can lead to abnormal neural remodeling impacting learning and memory processes. Identified knowledge gaps include improving quantitative understanding of the AOP across several KERs and additional testing of proposed modulating factors through experimental work. Broadly, it is envisioned that the outcome of these efforts could be extended to other cognitive disorders and complement ongoing work by international radiation governing bodies in their review of the system of radiological protection.
Collapse
Affiliation(s)
- Ahmad Sleiman
- Institut de Radioprotection et de Sûreté Nucléaire, St. Paul Lez Durance, Provence, France
| | - Kathleen B Miller
- Department of Health and Exercise Science, Morrison College Family of Health, University of St. Thomas, Saint Paul, Minnesota, USA
| | - Danicia Flores
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Jaqueline Kuan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Kaitlyn Altwasser
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Robyn Hocking
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | | | - Janice Huff
- NASA Langley Research Center, Hampton, Virginia, USA
| | | | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth Wilkins
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Environmental and Radiation Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Elahi S, Rezaeifar M, Osman M, Shahbaz S. Exploring the role of galectin-9 and artemin as biomarkers in long COVID with chronic fatigue syndrome: links to inflammation and cognitive function. Front Immunol 2024; 15:1443363. [PMID: 39386210 PMCID: PMC11461188 DOI: 10.3389/fimmu.2024.1443363] [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: 06/03/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
This study aimed to assess plasma galectin-9 (Gal-9) and artemin (ARTN) concentrations as potential biomarkers to differentiate individuals with Long COVID (LC) patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) from SARS-CoV-2 recovered (R) and healthy controls (HCs). Receiver operating characteristic (ROC) curve analysis determined a cut-off value of plasma Gal-9 and ARTN to differentiate LC patients from the R group and HCs in two independent cohorts. Positive correlations were observed between elevated plasma Gal-9 levels and inflammatory markers (e.g. SAA and IP-10), as well as sCD14 and I-FABP in LC patients. Gal-9 also exhibited a positive correlation with cognitive failure scores, suggesting its potential role in cognitive impairment in LC patients with ME/CFS. This study highlights plasma Gal-9 and/or ARTN as sensitive screening biomarkers for discriminating LC patients from controls. Notably, the elevation of LPS-binding protein in LC patients, as has been observed in HIV infected individuals, suggests microbial translocation. However, despite elevated Gal-9, we found a significant decline in ARTN levels in the plasma of people living with HIV (PLWH). Our study provides a novel and important role for Gal-9/ARTN in LC pathogenesis.
Collapse
Affiliation(s)
- Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, Edmonton, AB, Canada
- Women and Children Health Research Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, Edmonton, AB, Canada
- Glycomics Institute of Alberta, Edmonton, AB, Canada
- Alberta Transplant Institute, Edmonton, AB, Canada
| | - Maryam Rezaeifar
- School of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
| | - Mohammed Osman
- Li Ka Shing Institute of Virology, Edmonton, AB, Canada
- Women and Children Health Research Institute, Edmonton, AB, Canada
- Department of Medicine, Division of Rheumatology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shima Shahbaz
- School of Dentistry, Division of Foundational Sciences, Edmonton, AB, Canada
| |
Collapse
|
5
|
Xu D, Liu J, Meng S, Sun M, Chen Y, Hong Y. Isoflurane-induced neuroinflammation and NKCC1/KCC2 dysregulation result in long-term cognitive disorder in neonatal mice. BMC Anesthesiol 2024; 24:200. [PMID: 38840092 PMCID: PMC11151488 DOI: 10.1186/s12871-024-02587-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND The inhalational anesthetic isoflurane is commonly utilized in clinical practice, particularly in the field of pediatric anesthesia. Research has demonstrated its capacity to induce neuroinflammation and long-term behavioral disorders; however, the underlying mechanism remains unclear [1]. The cation-chloride cotransporters Na+-K+-2Cl--1 (NKCC1) and K+-2Cl--2 (KCC2) play a pivotal role in regulating neuronal responses to gamma-aminobutyric acid (GABA) [2]. Imbalances in NKCC1/KCC2 can disrupt GABA neurotransmission, potentially leading to neural circuit hyperexcitability and reduced inhibition following neonatal exposure to anesthesia [3]. Therefore, this study postulates that anesthetics have the potential to dysregulate NKCC1 and/or KCC2 during brain development. METHODS We administered 1.5% isoflurane anesthesia to neonatal rats for a duration of 4 h at postnatal day 7 (PND7). Anxiety levels were assessed using the open field test at PND28, while cognitive function was evaluated using the Morris water maze test between PND31 and PND34. Protein levels of NKCC1, KCC2, BDNF, and phosphorylated ERK (P-ERK) in the hippocampus were measured through Western blotting analysis. Pro-inflammatory cytokines IL-1β, IL-6, and TNF-α were quantified using ELISA. RESULTS We observed a decrease in locomotion trajectories within the central region and a significantly shorter total distance in the ISO group compared to CON pups, indicating that isoflurane induces anxiety-like behavior. In the Morris water maze (MWM) test, rats exposed to isoflurane exhibited prolonged escape latency onto the platform. Additionally, isoflurane administration resulted in reduced time spent crossing in the MWM experiment at PND34, suggesting long-term impairment of memory function. Furthermore, we found that isoflurane triggered activation of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α; downregulated KCC2/BDNF/P-ERK expression; and increased the NKCC1/KCC2 ratio in the hippocampus of PND7 rats. Bumetadine (NKCC1 specific inhibitors) reversed cognitive damage and effective disorder induced by isoflurane in neonatal rats by inhibiting TNF-α activation, normalizing IL-6 and IL-1β levels, restoring KCC2 expression levels as well as BDNF and ERK signaling pathways. Based on these findings, it can be speculated that BDNF, P-ERK, IL-1β, IL-6 and TNF - α may act downstream of the NKCC1/KCC2 pathway. CONCLUSIONS Our findings provide evidence that isoflurane administration in neonatal rats leads to persistent cognitive deficits through dysregulation of the Cation-Chloride Cotransporters NKCC1 and KCC2, BDNF, p-ERK proteins, as well as neuroinflammatory processes.
Collapse
Affiliation(s)
- Dongni Xu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Jiayi Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Shiyu Meng
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Meixian Sun
- The Eighth People's Hospital of Qingdao, Qingdao, Shandong Province, China
| | - Yuqing Chen
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
| | - Yu Hong
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 Yanjiang West Road, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Gao T, Wang T, Wu L, Tong Y, Tian J, Zhao K, Wang H. Saikosaponin-d alleviates depression by promoting NLRP3 ubiquitination and inhibiting inflammasome activation. Int Immunopharmacol 2024; 127:111324. [PMID: 38070467 DOI: 10.1016/j.intimp.2023.111324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024]
Abstract
Saikosaponin-d (SSd) is a triterpene saponin from the roots of Bupleurum chinese. Recent studies have revealed its antidepressant activity, but its mechanism involved is unclear. This study's objective was to ascertain how SSd may reduce depression in depressed mice subjected to chronic unpredictable animal stress (CUMS) and to investigate the mechanisms underlying these effects. Models of CUMS depression were established and different groups were treated with SSd and escitalopram. After the last day of administration of the treatment, behavioral tests were performed. ELISA was used to measure the expression of IL-1β, TNF-α, and IL-18, and western blot was used to measure the presence of proteins associated with NLRP3. Hippocampal neuronal damage was observed using Nissl staining, and NLRP3 ubiquitination assay was performed by immunoprecipitation and gene silencing. An inflammatory cell model was constructed by treating BV2 cells with lipopolysaccharides (LPS) and adenosine triphosphate (ATP) to verify the ubiquitination modification of NLRP3 by SSd. Behavioral tests demonstrated that SSd effectively alleviated depression-like symptoms. SSd should substantially limit the degrees of proteins associated with NLRP3, as properly as limit the harm to hippocampal neurons. Gene silencing results showed that SSd regulates NLRP3 through the E3 ubiquitin ligase MARCHF7. In vitro, SSd remarkably increased the protein expression of K48-linked ubiquitin in inflammatory BV2 cells, while decreasing the protein levels of NLRP3. Our findings suggest that SSd has antidepressant effects in CUMS mice by promoting ubiquitination of NLRP3 to inhibit inflammasome activation and improve the inflammatory state.
Collapse
Affiliation(s)
- Tiantian Gao
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Tao Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Lili Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Yue Tong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinlong Tian
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Keke Zhao
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Hanqing Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| |
Collapse
|
7
|
Gao Q, Gao Z, Su M, Huang Y, Zhang C, Li C, Zhan H, Liu B, Zhou X. Umbilical Cord Mesenchymal Stem Cells Overexpressing Heme Oxygenase-1 Promotes Symptoms Recovery in Cystitis Rats by Alleviating Neuroinflammation. Stem Cells Int 2023; 2023:8887091. [PMID: 38020203 PMCID: PMC10663085 DOI: 10.1155/2023/8887091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/20/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) seriously reduces the patient's quality of life, yet current therapies only provide partial relief. In the spinal dorsal horn (SDH), neuroinflammation plays a pivotal role in the development of IC. Injection of human umbilical cord mesenchymal stem cells (hUMSCs) to reduce inflammation is an effective strategy, and heme oxygenase-1 (HO-1) exhibits anti-nociceptive effect in neuroinflammatory pain. This study aimed to test the therapeutic effects of hUMSCs overexpressing HO-1 on cyclophosphamide-induced cystitis rat model. Cystitis rats were transplanted with altered cells and then assessed for 3 weeks. A series of behavioral measurements would be trial including suprapubic mechanical allodynia, depressive-like behaviors, micturition frequency, and short-term memory function. Additionally, western blot, immunofluorescence staining, and ELISA kit test for anti-inflammation effect. HUMSCs were capable of being transduced to overexpress HO-1. Injection of hUMSCs overexpressing HO-1 was more effective than hUMSCs alone in alleviating behavioral symptoms in rats. Furthermore, hUMSCs overexpressing HO-1 inhibited the activation of glial and TLR4/p65/NLRP3 pathway, decreased the levels of pro-inflammatory cytokines in the SDH region. Surprisingly, it markedly increased anti-inflammatory cytokine IL-10, reduced MDA content, and protected GSH concentrations in local environment. Our results suggest that injecting hUMSCs overexpressing HO-1 intrathecally can significantly promote functional outcomes in cystitis rats by reducing neuroinflammation, at least, partly through downregulating TLR4/p65/NLRP3 signaling pathway in the SDH region. This cell therapy affords a new strategy for IC/BPS treatment.
Collapse
Affiliation(s)
- Qiongqiong Gao
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Zhentao Gao
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Minzhi Su
- Department of Rehabilitation, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Yong Huang
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Chi Zhang
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Cuiping Li
- Department of Biotherapy Center, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Hailun Zhan
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Bolong Liu
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| | - Xiangfu Zhou
- Department of Urology, The Third Affiliated Hospital and Lingnan Hospital of the Sun Yat-Sen University, 2693 Kaichuang Road, Guangzhou 510700, China
| |
Collapse
|
8
|
Riddell C, Yonelinas AP, Shields GS. When stress enhances memory encoding: The beneficial effects of changing context. Neurobiol Learn Mem 2023; 205:107836. [PMID: 37820758 PMCID: PMC10909400 DOI: 10.1016/j.nlm.2023.107836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/29/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023]
Abstract
The effects of acute stress on memory encoding are complex, and we do not yet know all of the conditions that can determine whether stress at encoding improves or impairs memory. Recent work has found that changing contexts between encoding and stress can abolish the effects of post-encoding stress on memory, suggesting that context may play an important role in the effects of stress on memory. However, the role of context in the effects of stress on memory encoding is not yet known. We addressed this gap by examining the effects of context on the influence of acute stress on memory encoding. In a 2 × 2 experimental design, participants (N = 103) completed either a stressor (i.e., Socially Evaluated Cold Presser Test) or control task (i.e., warm water control) before completing a memory encoding task, which occurred in either in the same room as or a different room from the stressor or control task. Memory retrieval was tested for each participant within the context that they completed the encoding task. We found that, relative to nonstressed (i.e., control) participants, stressed participants who switched contexts prior to encoding showed better memory for both negative and neutral images. In contrast, when the stressor or control task occurred in the same room as memory encoding, stress had no beneficial effect on memory. These results highlight the importance of the ongoing context as a determinant of the effects of stress on memory encoding and present a challenge to current theoretical accounts of stress and memory.
Collapse
Affiliation(s)
- Cameron Riddell
- Department of Psychology and Center for Neuroscience, University of California, Davis, CA, USA
| | - Andrew P Yonelinas
- Department of Psychology and Center for Neuroscience, University of California, Davis, CA, USA
| | - Grant S Shields
- Department of Psychological Science, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
9
|
Dehdar K, Raoufy MR. Brain structural and functional alterations related to anxiety in allergic asthma. Brain Res Bull 2023; 202:110727. [PMID: 37562517 DOI: 10.1016/j.brainresbull.2023.110727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/29/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Psychiatric disorders are common in patients with allergic asthma, and they can have a significant impact on their quality of life and disease control. Recent studies have suggested that there may be potential immune-brain communication mechanisms in asthma, which can activate inflammatory responses in different brain areas, leading to structural and functional alterations and behavioral changes. However, the precise mechanisms underlying these alterations remain unclear. In this paper, we comprehensively review the relevant research on asthma-induced brain structural and functional alterations that lead to the initiation and promotion of anxiety. We summarize the possible pathways for peripheral inflammation to affect the brain's structure and function. Our review highlights the importance of addressing neuropsychiatric disorders in the clinical guidelines of asthma, to improve the quality of life of these patients. We suggest that a better understanding of the mechanisms underlying psychiatric comorbidities in asthma could lead to the development of more effective treatments for these patients.
Collapse
Affiliation(s)
- Kolsoum Dehdar
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
Weber MA, Kerr G, Thangavel R, Conlon MM, Abdelmotilib HA, Halhouli O, Zhang Q, Geerling JC, Narayanan NS, Aldridge GM. Alpha-synuclein pre-formed fibrils injected into prefrontal cortex primarily spread to cortical and subcortical structures and lead to isolated behavioral symptoms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526365. [PMID: 36778400 PMCID: PMC9915664 DOI: 10.1101/2023.01.31.526365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are characterized by diffuse spread of alpha-synuclein (α-syn) throughout the brain. Patients with PDD and DLB have a neuropsychological pattern of deficits that include executive dysfunction, such as abnormalities in planning, timing, working memory, and behavioral flexibility. The prefrontal cortex (PFC) plays a major role in normal executive function and often develops α-syn aggregates in DLB and PDD. To investigate the consequences of α-syn pathology in the cortex, we injected human α-syn pre-formed fibrils into the PFC of wildtype mice. We report that PFC PFFs: 1) induced α-syn aggregation in multiple cortical and subcortical regions with sparse aggregation in midbrain and brainstem nuclei; 2) did not affect interval timing or spatial learning acquisition but did mildly alter behavioral flexibility as measured by intraday reversal learning; 3) increased open field exploration; and 4) did not affect susceptibility to an inflammatory challenge. This model of cortical-dominant pathology aids in our understanding of how local α-syn aggregation might impact some symptoms in PDD and DLB.
Collapse
Affiliation(s)
- Matthew A. Weber
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Gemma Kerr
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Ramasamy Thangavel
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Mackenzie M. Conlon
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City
| | | | - Oday Halhouli
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Qiang Zhang
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Joel C. Geerling
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | | | - Georgina M. Aldridge
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| |
Collapse
|
11
|
Arjunan A, Song J. Pharmacological and physiological roles of adipokines and myokines in metabolic-related dementia. Biomed Pharmacother 2023; 163:114847. [PMID: 37150030 DOI: 10.1016/j.biopha.2023.114847] [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: 03/21/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023] Open
Abstract
Dementia is a detrimental neuropathologic condition with considerable physical, mental, social, and financial impact on patients and society. Patients with metabolic syndrome (MetS), a group of diseases that occur in tandem and increase the risk of neurologic diseases, have a higher risk of dementia. The ratio between muscle and adipose tissue is crucial in MetS, as these contain many hormones, including myokines and adipokines, which are involved in crosstalk and local paracrine/autocrine interactions. Evidence suggests that abnormal adipokine and myokine synthesis and release may be implicated in various MetS, such as atherosclerosis, diabetic mellitus (DM), and dyslipidemia, but their precise role is unclear. Here we review the literature on adipokine and myokine involvement in MetS-induced dementia via glucose and insulin homeostasis regulation, neuroinflammation, vascular dysfunction, emotional changes, and cognitive function.
Collapse
Affiliation(s)
- Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| |
Collapse
|
12
|
Pu Z, Wen H, Jiang H, Hou Q, Yan H. Berberine improves negative symptoms and cognitive function in patients with chronic schizophrenia via anti-inflammatory effect: a randomized clinical trial. Chin Med 2023; 18:41. [PMID: 37069570 PMCID: PMC10108529 DOI: 10.1186/s13020-023-00746-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 04/04/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Based on the neuroinflammation hypothesis in schizophrenia and known anti-inflammatory effects of berberine, the aim of the present study is to investigate the efficacy of berberine in treating negative symptoms and cognitive deficits in adult patients with chronic schizophrenia. METHODS Enrolled participants were randomized to receive berberine or placebo for 3 months. The Scale for the Assessment of Negative Symptoms (SANS), Trail-making Test A (TMT-A), Trail-making Test B (TMT-B), and Hopkins Verbal Learning Test (HVLT) were used to evaluate the negative symptoms and cognitive function at four-time points (baseline, 1st, 2nd, and 3rd month). Serum levels of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) were used as inflammatory markers. 106 patients with per-protocol were analyzed, 56 in the experimental (berberine) group and 50 in the control (placebo) group. RESULTS From baseline to month 3, patients receiving berberine demonstrated a decrease in total scores on clinical scales SANS, TMT-A and TMT-B and showed a serum level reduction of IL-1β, IL-6 and TNF-α comparing with patients in the control group (P < 0.05). There were positive correlations between the change of serum IL-1β level and the change of SANS (r = 0.210, P = 0.039), TMT-A (r = 0.522, P < 0.001), and TMT-B (r = 0.811, P < 0.001); between the change of serum IL-6 level and the change of TMT-A (r = 0.562, P < 0.001), and TMT-B (r = 0.664, P < 0.001); between the change of serum TNF-α level and the change of TMT-B (r = 0.472, P < 0.001) after berberine treatment. CONCLUSIONS Berberine is an anti-inflammatory agent that can potentially mitigate the negative symptoms and cognitive deficits in patients with schizophrenia.
Collapse
Affiliation(s)
- Zhengping Pu
- Department of Psychiatry, Kangci Hospital of Jiaxing, No. 3118 Huancheng North Road, Tongxiang, 314500, Zhejiang, China
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Minhang, Shanghai, 201108, China
| | - Hui Wen
- Department of Traditional Chinese Medicine, Second People's Hospital of Tongxiang, Tongxiang, 314500, Zhejiang, China
| | - Hongxia Jiang
- Department of Psychiatry, Kangci Hospital of Jiaxing, No. 3118 Huancheng North Road, Tongxiang, 314500, Zhejiang, China.
| | - Qingmei Hou
- Department of Clinical Psychology, The Second Specialized Hospital of Hegang, Hegang, 154102, Heilongjiang, China
| | - Hui Yan
- Department of Psychiatry, Second People's Hospital of Taizhou, Taizhou, 317200, Zhejiang, China
| |
Collapse
|
13
|
Zaichenko MI, Philenko P, Sidorina V, Grigoryan GA. Acute and Chronic Lipopolysaccharide-Induced Stress Changes Expression of Proinflammatory Cytokine Genes in the Rat Brain Region-Specifically and Affects Learning and Memory. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:526-538. [PMID: 37080938 DOI: 10.1134/s0006297923040089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Goal of the current work was to conduct comparative analysis of the effects of acute and chronic lipopolysaccharide-induced stress on the behavior of rats in the Morris water maze test and on expression of mRNA of proinflammatory cytokines and BDNF in different brain structures. Relevance of this study is related to poor understanding of the effects of acute and chronic stress on manifestation of cognitive brain functions, as well as ambiguity of the literature data on the effects of both stresses on hypothalamic pituitary axis and expression of the proinflammatory cytokine genes. In the experiments with rats, acute lipopolysaccharide (LPS)-induced stress improved learning in the Morris water maze. For the period of learning, the rats under acute stress swam on average less distance to reach a hidden platform, spent less time in the peripheral zone of the pool (thigmotaxis), and had low speed compared to the control animals and to the group of rats under chronic LPS-induced stress. In the test without a platform in the pool there were no significant differences between the groups on the time spent in the platform quadrant and distance swum. Acute stress caused substantial increase of the TNF-α and IL-1β mRNA concentrations in the hippocampus and amygdala, but not in the frontal lobe in comparison with the control animals. Although chronic stress increased the levels of the TNF-α and IL-1β mRNA in the amygdala and hippocampus compared to the control groups, significance between the groups was only marginal and BDNF concentration did not differ from the control animals in any of the brain structures mentioned. Expression of the IL-6 mRNA only marginally increased in the amygdala of the animals under the acute LPS-induced stress and marginally decreased in the animals under chronic LPS stress in the hippocampus relative to the control groups. In total, the most pronounced molecular-biochemical changes occurred in the amygdala and hippocampus, where increase of the expression of the TNF-α and IL-1β interleukins mRNAs were observed in the animals under acute and chronic LPS-induced stress and no changes in the BDNF mRNA concentration were observed in the frontal lobe.
Collapse
Affiliation(s)
- Mariya I Zaichenko
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
| | - Pavel Philenko
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Viktoriya Sidorina
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Grigory A Grigoryan
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| |
Collapse
|
14
|
Buka SL, Lee YH, Goldstein JM. Infections During Pregnancy and Risks for Adult Psychosis: Findings from the New England Family Study. Curr Top Behav Neurosci 2023; 61:49-69. [PMID: 36376640 DOI: 10.1007/7854_2022_397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
For the past 40 years, our team has conducted a unique program of research investigating the prenatal risks for schizophrenia and related adult psychiatric disorders. The New England Family Study is a long-term prospective cohort study of over 16,000 individuals followed from the prenatal period for over 50 years. This chapter summarizes several major phases and findings from this work, highlighting recent results on maternal prenatal bacterial infections and brain imaging. Implications regarding the causes and potential prevention of major psychotic disorders are discussed.
Collapse
Affiliation(s)
- Stephen L Buka
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA.
| | - Younga Heather Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA
| | - Jill M Goldstein
- Departments of Psychiatry and Medicine, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
15
|
Zhan Y, Al-Nusaif M, Ding C, Zhao L, Dong C. The potential of the gut microbiome for identifying Alzheimer's disease diagnostic biomarkers and future therapies. Front Neurosci 2023; 17:1130730. [PMID: 37179559 PMCID: PMC10174259 DOI: 10.3389/fnins.2023.1130730] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Being isolated from the peripheral system by the blood-brain barrier, the brain has long been considered a completely impervious tissue. However, recent findings show that the gut microbiome (GM) influences gastrointestinal and brain disorders such as Alzheimer's disease (AD). Despite several hypotheses, such as neuroinflammation, tau hyperphosphorylation, amyloid plaques, neurofibrillary tangles, and oxidative stress, being proposed to explain the origin and progression of AD, the pathogenesis remains incompletely understood. Epigenetic, molecular, and pathological studies suggest that GM influences AD development and have endeavored to find predictive, sensitive, non-invasive, and accurate biomarkers for early disease diagnosis and monitoring of progression. Given the growing interest in the involvement of GM in AD, current research endeavors to identify prospective gut biomarkers for both preclinical and clinical diagnoses, as well as targeted therapy techniques. Here, we discuss the most recent findings on gut changes in AD, microbiome-based biomarkers, prospective clinical diagnostic uses, and targeted therapy approaches. Furthermore, we addressed herbal components, which could provide a new venue for AD diagnostic and therapy research.
Collapse
Affiliation(s)
- Yu Zhan
- Department of Neurology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Murad Al-Nusaif
- Department of Neurology, First Affiliated Hospital, Dalian Medical University, Dalian, China
- Liaoning Provincial Key Laboratories for Research on the Pathogenic Mechanism of Neurological Disease, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Cong Ding
- The Center for Gerontology and Geriatrics, Dalian Friendship Hospital, Dalian, China
| | - Li Zhao
- Department of Neurology, First Affiliated Hospital, Dalian Medical University, Dalian, China
- *Correspondence: Li Zhao,
| | - Chunbo Dong
- Department of Neurology, First Affiliated Hospital, Dalian Medical University, Dalian, China
- Chunbo Dong,
| |
Collapse
|
16
|
Zhou Y, Ma Y, Yu C, Chen Y, Ding J, Yu J, Zhou R, Wang X, Fan T, Shi C. Detection Analysis of Perioperative Plasma and CSF Reveals Risk Biomarkers of Postoperative Delirium of Parkinson's Disease Patients Undergoing Deep Brain Stimulation of the Subthalamic Nuclei. Clin Interv Aging 2022; 17:1739-1749. [PMID: 36474580 PMCID: PMC9719687 DOI: 10.2147/cia.s388690] [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: 09/16/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Objective This study aimed to explore possible biomarkers of postoperative delirium (POD) of Parkinson's disease (PD) patients received deep brain stimulation (DBS) of the subthalamic nuclei. Materials and methods This nested case control study analyzed perioperative plasma and cerebral spinal fluid (CSF) of patients (n = 40) who developed POD undergone DBS surgery (n = 10) and those who did not (n = 30). Blood sample was collected before surgery and on the first day postoperative, CSF sample was collected at the beginning of the operation. POD was assessed by the Confusion Assessment Method (CAM) twice a day between 7:00 am and 7:00 pm after the surgery until discharge. Plasma and CSF sample from the two groups were analyzed to investigate possible biomarkers for POD in PD patients. Results There was no difference between POD and Non-POD groups on the concentration of Interleukin 6 and Tumor Necrosis Factor-α in CSF, preoperative plasma and postoperative plasma. There was no difference between POD and Non-POD groups on the concentration of S100 calcium-binding protein β protein (S100β) and Neurofilament light chain (NFL) in preoperative plasma and postoperative plasma. The concentration of C-reactive protein (CRP), NFL and S100β were significant higher in POD group than non-POD group in CSF. The concentration of CRP was significantly higher in POD group than non-POD group in preoperative plasma and postoperative plasma. CSF concentration of S100β might be a potential biomarker for POD via the receiver operating characteristic curve analysis and the area under the curve value of 0.973. Conclusion For PD patients received DBS surgery, CSF S100β might be a marker for aiding detection of high-risk patients with delirium. This requires further confirmation in clinical trials.
Collapse
Affiliation(s)
- Yongde Zhou
- Department of Anesthesiology, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Yu Ma
- Department of Neurosurgery, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Cuiping Yu
- Department of Anesthesiology, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Yao Chen
- Department of Anesthesiology, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Jian Ding
- Department of Anesthesiology, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Jianfeng Yu
- Department of Anesthesiology, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Rongsong Zhou
- Department of Neurosurgery, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Xiaoxiao Wang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Ting Fan
- Department of Anesthesiology, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Chengmei Shi
- Department of Anesthesiology, Peking University Third Hospital, Beijing, People’s Republic of China,Correspondence: Chengmei Shi; Ting Fan, Email ;
| |
Collapse
|
17
|
Duindam HB, Kessels RP, van den Borst B, Pickkers P, Abdo WF. Long-term cognitive performance and its relation to anti-inflammatory therapy in a cohort of survivors of severe COVID-19. Brain Behav Immun Health 2022; 25:100513. [PMID: 36159208 PMCID: PMC9482799 DOI: 10.1016/j.bbih.2022.100513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/07/2022] [Accepted: 09/11/2022] [Indexed: 12/12/2022] Open
Abstract
Background and objectives Long-term cognitive performance data in former critically ill COVID-19 patients are sparse. Current evidence suggests that cognitive decline is related to neuroinflammation, which might be attenuated by COVID-19 related anti-inflammatory therapies. The objective of this prospective cohort study was to study long term cognitive outcomes following severe COVID-19 and the relation to anti-inflammatory therapies. Methods Prospective observational cohort of patients that survived an intensive care unit (ICU) admission due to severe COVID-19. Six months after hospital discharge, we extensively assessed both objective cognitive functioning and subjective cognitive complaints. Furthermore, patients were stratified in cohorts according to their anti-inflammatory treatment (i.e. no immunomodulatory therapy, dexamethasone, or both dexamethasone and interleukin-6 receptor antagonist tocilizumab). Results 96 patients were included (March 2020–June 2021, median [IQR] age 61 [55–69] years). 91% received invasive mechanical ventilation, and mean ± SD severity-of-disease APACHE–II–score at admission was 15.8 ± 4.1. After 6.5 ± 1.3 months, 27% of patients scored cognitively impaired. Patients that did or did not develop cognitive impairments were similar in ICU-admission parameters, clinical course and delirium incidence. Patients with subjective cognitive complaints (20%) were more likely women (61% vs 26%), and had a shorter ICU stay (median [IQR] 8 [5–15] vs 18 [9–31], p = 0.002). Objective cognitive dysfunction did not correlate with subjective cognitive dysfunction. 27% of the participants received dexamethasone during intensive care admission, 44% received additional tocilizumab and 29% received neither. Overall occurrence and severity of cognitive dysfunction were not affected by anti-inflammatory therapy, although patients treated with both dexamethasone and tocilizumab had worse executive functioning scores (Trail Making Test interference) than patients without anti-inflammatory treatment (T-score 40.3 ± 13.5 vs 49.1 ± 9.3, p = 0.007). Discussion A relevant proportion of critically ill COVID-19 patients shows deficits in long-term cognitive functioning. Apart from more pronounced executive dysfunction, overall, anti-inflammatory therapy appeared not to affect long-term cognitive performance. Our findings provide insight in long-term cognitive outcomes in patients who survived COVID-19, that may facilitate health-care providers counseling patients and their caregivers. Up to a third of survivors of severe COVID-19 develops long-term cognitive impairment. Subjective cognitive complaints six months following severe COVID-19 do not correlate with objective cognitive impairment. COVID-19 related immunomodulatory therapy is not associated with the overall incidence of cognitive dysfunction.
Collapse
|
18
|
Abstract
Systemic inflammation elicited by sepsis can induce an acute cerebral dysfunction known as sepsis-associated encephalopathy (SAE). Recent evidence suggests that SAE is common but shows a dynamic trajectory over time. Half of all patients with sepsis develop SAE in the intensive care unit, and some survivors present with sustained cognitive impairments for several years after initial sepsis onset. It is not clear why some, but not all, patients develop SAE and also the factors that determine the persistence of SAE. Here, we first summarize the chronic pathology and the dynamic changes in cognitive functions seen after the onset of sepsis. We then outline the cerebral effects of sepsis, such as neuroinflammation, alterations in neuronal synapses and neurovascular changes. We discuss the key factors that might contribute to the development and persistence of SAE in older patients, including premorbid neurodegenerative pathology, side effects of sedatives, renal dysfunction and latent virus reactivation. Finally, we postulate that some of the mechanisms that underpin neuropathology in SAE may also be relevant to delirium and persisting cognitive impairments that are seen in patients with severe COVID-19.
Collapse
Affiliation(s)
- Tatsuya Manabe
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
19
|
Meenakshi Sundaram DN, Plianwong S, Kc R, Ostergaard H, Uludağ H. In Vitro Cytotoxicity and Cytokine Production by Lipid-Substituted Low Molecular Weight Branched PEIs Used for Gene Delivery. Acta Biomater 2022; 148:279-297. [PMID: 35738388 DOI: 10.1016/j.actbio.2022.06.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022]
Abstract
Lipid-modified low molecular weight branched polyethyleneimines (PEIs) are promising non-viral gene delivery systems that have been successfully explored for treatment of various diseases. The present study aims to determine in vitro safety of these delivery systems based on assessment of cytotoxicity with peripheral blood mononuclear cells (PBMCs), hemolysis with human red blood cells (RBC) and cytokine secretion from several sources of PBMCs. The viability of cells treated with lipopolymer/pDNA complexes was dependent on the polymer:pDNA ratio used but remained low at therapeutically relevant concentrations for most lipopolymers, except for the propionic acid substituted PEIs. The extent of hemolysis was minimal and below the accepted safety levels with most of the lipopolymers; however, some linoleic acid substituted PEIs yielded significant hemolysis activity. Unlike strong cytokine secretion from PMA/IO stimulated cells, most lipopolymer/pDNA complexes remained non-responsive, showing minimal changes in cytokine secretion (TNF-α, IL-6 and IFN-γ) irrespective of the lipopolymer/pDNA formulations. The 0.6 kDa PEI with lauric acid substituent displayed slight cytokine upregulation, however it remained low relative to the positive controls. This study demonstrated that the lipid modified LMW PEIs are expected to be safe in contact with blood components. However, close attention to lipopolymer concentration and ratio of polymer to pDNA in formulations might be required for individual lipopolymers for optimal safety response in nucleic acid therapies. STATEMENT OF SIGNIFICANCE: : This manuscript investigated the safety aspects of various lipid modified low molecular weight polyethylenimine (LMW-PEI) polymers employed for pDNA delivery through in vitro studies. Using peripheral blood mononuclear cells (PBMCs) from multiple sources, we show that the hemolysis ability was minimal for most polymers, although a particular lipid substituent (linoleic acid) at specific ratios exhibited hemolysis. The levels of pro-inflammatory cytokines (TNF-α, IL-6 and IFN-γ) were slightly upregulated only with a lauric acid substituted 0.6PEI, but remained low relative to positive control treatments. We further report the beneficial effect of polyacrylic acid additives on hemolysis and cytokine secretion to a reasonable extent. This study confirms the feasibility of using LMW-PEI as safe delivery agents for various therapeutic purposes.
Collapse
Affiliation(s)
| | - Samarwadee Plianwong
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada; Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Remant Kc
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | - Hanne Ostergaard
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
20
|
Aschman T, Mothes R, Heppner FL, Radbruch H. What SARS-CoV-2 does to our brains. Immunity 2022; 55:1159-1172. [PMID: 35777361 PMCID: PMC9212726 DOI: 10.1016/j.immuni.2022.06.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/22/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Abstract
Neurological symptoms in SARS-CoV-2-infected patients have been reported, but their cause remains unclear. In theory, the neurological symptoms observed after SARS-CoV-2 infection could be (1) directly caused by the virus infecting brain cells, (2) indirectly by our body’s local or systemic immune response toward the virus, (3) by coincidental phenomena, or (4) a combination of these factors. As indisputable evidence of intact and replicating SARS-CoV-2 particles in the central nervous system (CNS) is currently lacking, we suggest focusing on the host’s immune reaction when trying to understand the neurocognitive symptoms associated with SARS-CoV-2 infection. In this perspective, we discuss the possible immune-mediated mechanisms causing functional or structural CNS alterations during acute infection as well as in the post-infectious context. We also review the available literature on CNS affection in the context of COVID-19 infection, as well as observations from animal studies on the molecular pathways involved in sickness behavior.
Collapse
|
21
|
Zhang Y, Gao Y, Yang F, Wu X, Tang Z, Liu H. Neuroglobin alleviates the neurotoxicity of sevoflurane to fetal rats by inhibiting neuroinflammation and affecting microglial polarization. Brain Res Bull 2022; 183:142-152. [DOI: 10.1016/j.brainresbull.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 01/20/2023]
|
22
|
Grant CV, Russart KLG, Pyter LM. A novel targeted approach to delineate a role for estrogen receptor-β in ameliorating murine mammary tumor-associated neuroinflammation. Endocrine 2022; 75:949-958. [PMID: 34797509 DOI: 10.1007/s12020-021-02931-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/27/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Circulating estrogens in breast cancer patients and survivors are often extremely low due to menopause and estrogen-reducing cancer treatments. Simultaneously, circulating inflammatory markers, and inflammatory proteins in brains of rodent tumor models, can be elevated and correlate with debilitating neurological and psychological comorbidities. Because estrogen has anti-inflammatory properties in the brain, we hypothesized that mammary tumor-induced neuroinflammation is driven, in part, by reduced brain estrogen signaling. METHODS An ovariectomized mouse model of postmenopausal breast cancer utilizing the ERα-positive 67NR mammary tumor cell line was used for these experiments. A novel, orally bioavailable, and brain penetrant ERβ agonist was administered daily via oral gavage. Following treatment, estrogen-responsive genes were measured in brain regions. Central and circulating inflammatory markers were measured via RT-qPCR and a multiplex cytokine array, respectively. RESULTS We present novel findings that peripheral mammary tumors alter estrogen signaling genes including receptors and aromatase in the hypothalamus, hippocampus, and frontal cortex. Mammary tumors induced peripheral and central inflammation, however, pharmacological ERβ activation was not sufficient to reduce this inflammation. CONCLUSIONS Data presented here suggest that compensating for low circulating estrogen with ERβ brain activation is not sufficient to attenuate mammary tumor-induced neuroinflammation, and is therefore not a likely candidate for the treatment of behavioral symptoms in patients. The novel finding that mammary tumors alter estrogen signaling-related genes is a clinically relevant advancement to the understanding of how peripheral tumor biology modulates neurobiology. This is necessary to predict and prevent behavioral comorbidities (e.g., cognitive impairment) prevalent in cancer patients and survivors.
Collapse
Affiliation(s)
- Corena V Grant
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA
| | - Kathryn L G Russart
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Minnesota State University Moorhead, Moorhead, MN, USA
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA.
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
H. Alhowai A, Almogbel Y, A.H. Abdel A, A. Aldubay M, Alfheeaid HA, Felemban SG, Chigurupat S, F. Alharbi I, S. Alharbi H. Metformin Induced Cognitive Impairment and Neuroinflammation in CMF-Treated Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.228.235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
24
|
The Effects of Modified Curcumin Preparations on Glial Morphology in Aging and Neuroinflammation. Neurochem Res 2022; 47:813-824. [PMID: 34988899 DOI: 10.1007/s11064-021-03499-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is characterized by reactive microglia and astrocytes (collectively called gliosis) in the central nervous system and is considered as one of the main pathological hallmarks in different neurodegenerative diseases such as Alzheimer's disease, age-related dementia, and multiple sclerosis. Upon activation, glia undergoes structural and morphological changes such as the microglial cells swell in size and astrocytes become bushy, which play both beneficial and detrimental roles. Hence, they are unable to perform the normal physiological role in brain immunity. Curcumin, a cytokine suppressive anti-inflammatory drug, has a high proven pre-clinical potency and efficacy to reverse chronic neuroinflammation by attenuating the activation and morphological changes that occur in the microglia and astrocytes. This review will highlight the recent findings on the tree structure changes of microglia and astrocytes in neuroinflammation and the effects of curcumin against the activation and morphology of glial cells.
Collapse
|
25
|
Pilarzyk K, Farmer R, Porcher L, Kelly MP. The Role of PDE11A4 in Social Isolation-Induced Changes in Intracellular Signaling and Neuroinflammation. Front Pharmacol 2021; 12:749628. [PMID: 34887755 PMCID: PMC8650591 DOI: 10.3389/fphar.2021.749628] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 11A (PDE11A), an enzyme that degrades cyclic nucleotides (cAMP and cGMP), is the only PDE whose mRNA expression in brain is restricted to the hippocampal formation. Previously, we showed that chronic social isolation changes subsequent social behaviors in adult mice by reducing expression of PDE11A4 in the membrane fraction of the ventral hippocampus (VHIPP). Here we seek extend these findings by determining 1) if isolation-induced decreases in PDE11A4 require chronic social isolation or if they occur acutely and are sustained long-term, 2) if isolation-induced decreases occur uniquely in adults (i.e., not adolescents), and 3) how the loss of PDE11 signaling may increase neuroinflammation. Both acute and chronic social isolation decrease PDE11A4 expression in adult but not adolescent mice. This decrease in PDE11A4 is specific to the membrane compartment of the VHIPP, as it occurs neither in the soluble nor nuclear fractions of the VHIPP nor in any compartment of the dorsal HIPP. The effect of social isolation on membrane PDE11A4 is also selective in that PDE2A and PDE10A expression remain unchanged. Isolation-induced decreases in PDE11A4 expression appear to be functional as social isolation elicited changes in PDE11A-relevant signal transduction cascades (i.e., decreased pCamKIIα and pS6-235/236) and behavior (i.e., increased remote long-term memory for social odor recognition). Interestingly, we found that isolation-induced decreases in membrane PDE11A4 correlated with increased expression of interleukin-6 (IL-6) in the soluble fraction, suggesting pro-inflammatory signaling for this cytokine. This effect on IL-6 is consistent with the fact that PDE11A deletion increased microglia activation, although it left astrocytes unchanged. Together, these data suggest that isolation-induced decreases in PDE11A4 may alter subsequent social behavior via increased neuroinflammatory processes in adult mice.
Collapse
Affiliation(s)
- Katy Pilarzyk
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Reagan Farmer
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Latarsha Porcher
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Michy P Kelly
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.,Center for Aging Research, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
26
|
Sriwattanapong K, Sa-Ard-Iam N, Boonprakong L, Subbalekha K, Trachoo V, Suratannon N, Porntaveetus T, Shotelersuk V. Reduced ELANE and SLPI expression compromises dental pulp cell activity. Cell Prolif 2021; 54:e13132. [PMID: 34580954 PMCID: PMC8560611 DOI: 10.1111/cpr.13132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 02/03/2023] Open
Abstract
Background Patients with ELANE variants and severe congenital neutropenia (SCN) commonly develop oral complications. Whether they are caused only by low neutrophil count or the combination of neutropenia and aberrant dental cells is unknown. Methods Genetic variant was identified with exome sequencing. Dental pulp cells isolated from the SCN patient with an ELANE mutation were investigated for gene expression, enzyme activity, proliferation, colony formation, wound healing, apoptosis, ROS, attachment, spreading and response to lipopolysaccharide. Results ELANE cells had diminished expression of ELANE and SLPI and reduced neutrophil elastase activity. Moreover, ELANE cells exhibited impaired proliferation, colony forming, migration, attachment and spreading; and significantly increased ROS formation and apoptosis, corresponding with increased Cyclin D1 and MMP2 levels. The intrinsic levels of TGF‐β1 and TNF‐α were significantly increased; however, IL‐6, IL‐8 and NF‐kB1 were significantly decreased in ELANE cells compared with those in controls. After exposure to lipopolysaccharide, ELANE cells grew larger, progressed to more advanced cell spreading stages and showed significantly increased SLPI, TNF‐α and NF‐kB1 and tremendously increased IL‐6 and IL‐8 expression, compared with controls. Conclusion This study, for the first time, suggests that in addition to neutropenia, the aberrant levels and functions of ELANE, SLPI and their downstream molecules in pulp cells play an important role in oral complications in SCN patients. In addition, pulp cells with diminished neutrophil elastase and SLPI are highly responsive to inflammation.
Collapse
Affiliation(s)
- Kanokwan Sriwattanapong
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Noppadol Sa-Ard-Iam
- Center of Excellence in Periodontal Disease and Dental Implant, Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Lawan Boonprakong
- Oral Biology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Keskanya Subbalekha
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorapat Trachoo
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Narissara Suratannon
- Pediatric Allergy & Clinical Immunology Research Unit, Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | - Thantrira Porntaveetus
- Genomics and Precision Dentistry Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
27
|
Porcher L, Bruckmeier S, Burbano SD, Finnell JE, Gorny N, Klett J, Wood SK, Kelly MP. Aging triggers an upregulation of a multitude of cytokines in the male and especially the female rodent hippocampus but more discrete changes in other brain regions. J Neuroinflammation 2021; 18:219. [PMID: 34551810 PMCID: PMC8459490 DOI: 10.1186/s12974-021-02252-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/25/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Despite widespread acceptance that neuroinflammation contributes to age-related cognitive decline, studies comparing protein expression of cytokines in the young versus old brains are surprisingly limited in terms of the number of cytokines and brain regions studied. Complicating matters, discrepancies abound-particularly for interleukin 6 (IL-6)-possibly due to differences in sex, species/strain, and/or the brain regions studied. METHODS As such, we clarified how cytokine expression changes with age by using a Bioplex and Western blot to measure multiple cytokines across several brain regions of both sexes, using 2 mouse strains bred in-house as well as rats obtained from NIA. Parametric and nonparametric statistical tests were used as appropriate. RESULTS In the ventral hippocampus of C57BL/6J mice, we found age-related increases in IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-6, IL-9, IL-10, IL-12p40, IL-12p70, IL-13, IL-17, eotaxin, G-CSF, interfeuron δ, KC, MIP-1a, MIP-1b, rantes, and TNFα that are generally more pronounced in females, but no age-related change in IL-5, MCP-1, or GM-CSF. We also find aging is uniquely associated with the emergence of a module (a.k.a. network) of 11 strongly intercorrelated cytokines, as well as an age-related shift from glycosylated to unglycosylated isoforms of IL-10 and IL-1β in the ventral hippocampus. Interestingly, age-related increases in extra-hippocampal cytokine expression are more discreet, with the prefrontal cortex, striatum, and cerebellum of male and female C57BL/6J mice demonstrating robust age-related increase in IL-6 expression but not IL-1β. Importantly, we found this widespread age-related increase in IL-6 also occurs in BALB/cJ mice and Brown Norway rats, demonstrating conservation across species and rearing environments. CONCLUSIONS Thus, age-related increases in cytokines are more pronounced in the hippocampus compared to other brain regions and can be more pronounced in females versus males depending on the brain region, genetic background, and cytokine examined.
Collapse
Affiliation(s)
- Latarsha Porcher
- Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Sophie Bruckmeier
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Rm 216, Baltimore, MD, 21201, USA
| | - Steven D Burbano
- Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Julie E Finnell
- Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Nicole Gorny
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Rm 216, Baltimore, MD, 21201, USA
| | - Jennifer Klett
- Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Susan K Wood
- Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Michy P Kelly
- Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, 29209, USA. .,Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSFII Rm 216, Baltimore, MD, 21201, USA. .,Center for Research on Aging, University of Maryland School of Medicine, 20 Penn St, HSFII Rm 216, Baltimore, MD, 21201, USA.
| |
Collapse
|
28
|
Posillico CK, Garcia-Hernandez RE, Tronson NC. Sex differences and similarities in the neuroimmune response to central administration of poly I:C. J Neuroinflammation 2021; 18:193. [PMID: 34488804 PMCID: PMC8418962 DOI: 10.1186/s12974-021-02235-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The neuroimmune system is required for normal neural processes, including modulation of cognition, emotion, and adaptive behaviors. Aberrant neuroimmune activation is associated with dysregulation of memory and emotion, though the precise mechanisms at play are complex and highly context dependent. Sex differences in neuroimmune activation and function further complicate our understanding of its roles in cognitive and affective regulation. METHODS Here, we characterized the physiological sickness and inflammatory response of the hippocampus following intracerebroventricular (ICV) administration of a synthetic viral mimic, polyinosinic:polycytidylic acid (poly I:C), in both male and female C57Bl/6N mice. RESULTS We observed that poly I:C induced weight loss, fever, and elevations of cytokine and chemokines in the hippocampus of both sexes. Specifically, we found transient increases in gene expression and protein levels of IL-1α, IL-1β, IL-4, IL-6, TNFα, CCL2, and CXCL10, where males showed a greater magnitude of response compared with females. Only males showed increased IFNα and IFNγ in response to poly I:C, whereas both males and females exhibited elevations of IFNβ, demonstrating a specific sex difference in the anti-viral response in the hippocampus. CONCLUSION Our data suggest that type I interferons are one potential node mediating sex-specific cytokine responses and neuroimmune effects on cognition. Together, these findings highlight the importance of using both males and females and analyzing a broad set of inflammatory markers in order to identify the precise, sex-specific roles for neuroimmune dysregulation in neurological diseases and disorders.
Collapse
Affiliation(s)
- Caitlin K Posillico
- Psychology Department, University of Michigan, 530 Church St., Ann Arbor, MI 48109, USA
| | | | - Natalie C Tronson
- Psychology Department, University of Michigan, 530 Church St., Ann Arbor, MI 48109, USA.
| |
Collapse
|
29
|
Miller BJ, Herzig KH, Jokelainen J, Karhu T, Keinänen-Kiukaanniemi S, Järvelin MR, Veijola J, Viinamäki H, Päivikki Tanskanen, Jääskeläinen E, Isohanni M, Timonen M. Inflammation, hippocampal volume, and cognition in schizophrenia: results from the Northern Finland Birth Cohort 1966. Eur Arch Psychiatry Clin Neurosci 2021; 271:609-622. [PMID: 32382794 DOI: 10.1007/s00406-020-01134-x] [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: 01/23/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023]
Abstract
Increased blood interleukin-6 (IL-6) levels are a replicated abnormality in schizophrenia, and may be associated with smaller hippocampal volumes and greater cognitive impairment. These findings have not been investigated in a population-based birth cohort. The general population Northern Finland Birth Cohort 1966 was followed until age 43. Subjects with schizophrenia were identified through the national Finnish Care Register. Blood IL-6 levels were measured in n = 82 subjects with schizophrenia and n = 5373 controls at age 31. Additionally, 31 patients with schizophrenia and 63 healthy controls underwent brain structural MRI at age 34, and cognitive testing at ages 34 and 43. Patients with schizophrenia had significantly higher median (interquartile range) blood IL-6 levels than controls (5.31, 0.85-17.20, versus 2.42, 0.54-9.36, p = 0.02) after controlling for potential confounding factors. In both schizophrenia and controls, higher blood IL-6 levels were predictors of smaller hippocampal volumes, but not cognitive performance at age 34. We found evidence for increased IL-6 levels in patients with midlife schizophrenia from a population-based birth cohort, and replicated associations between IL-6 levels and hippocampal volumes. Our results complement and extend the previous findings, providing additional evidence that IL-6 may play a role in the pathophysiology of schizophrenia and associated brain alterations.
Collapse
Affiliation(s)
- Brian J Miller
- Department of Psychiatry and Health Behavior, Augusta University, 997 Saint Sebastian Way, Augusta, GA, 30912, USA.
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,Medical Research Center (MRC) and Oulu University Hospital, Oulu, Finland.,Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
| | - Jari Jokelainen
- Medical Research Center (MRC) and Oulu University Hospital, Oulu, Finland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Toni Karhu
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Sirkka Keinänen-Kiukaanniemi
- Medical Research Center (MRC) and Oulu University Hospital, Oulu, Finland.,Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Marjo-Riitta Järvelin
- Medical Research Center (MRC) and Oulu University Hospital, Oulu, Finland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland.,Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.,MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Juha Veijola
- Medical Research Center (MRC) and Oulu University Hospital, Oulu, Finland.,Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland
| | - Heimo Viinamäki
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.,Psychiatry, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | | | - Erika Jääskeläinen
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland
| | - Matti Isohanni
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Markku Timonen
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
| |
Collapse
|
30
|
IL-6 and IL-1β upregulation and tau protein phosphorylation in response to chronic alcohol exposure in the mouse hippocampus. Neuroreport 2021; 32:851-857. [PMID: 34029285 DOI: 10.1097/wnr.0000000000001661] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alcoholism and alcohol abuse can lead to memory loss and cognitive dysfunction. The neuroinflammatory response plays an important role in the neurotoxic mechanism of chronic alcohol exposure. Additionally, the phosphorylation status of the tau protein is closely related to neurotoxicity and synaptic function. As inflammatory cytokines have been shown to regulate tau phosphorylation, in the present study, the aim was to determine whether cognitive impairment caused by chronic alcohol exposure is associated with neuroinflammation and tau hyperphosphorylation in the hippocampus. We established a chronic alcohol exposure model of C57BL/6J mice. The Y maze was used to assess the spatial recognition ability of mice, and ELISA was used to detect the levels of inflammatory cytokines IL-1β and IL-6 in the serum. Immunohistochemical and western blot assays were used to assess the expression levels of IL-1β and IL-6, as well as tau protein and its phosphorylation status in the hippocampus. We also analyzed the mRNA and protein expression of the synapse-associated proteins PSD95 and synaptophysin in the hippocampus. Our results showed that chronic alcohol exposure impaired the spatial recognition ability of mice upregulated the expression of IL-1β and IL-6 in the serum and hippocampus and increased the phosphorylation of tau protein in the hippocampus. In addition, chronic alcohol exposure downregulated PSD95 and synaptophysin protein levels. The present results indicate that hippocampal IL-1β, IL-6, and phosphorylated tau proteins may be involved in the neurotoxic mechanism of chronic alcohol exposure by mediating synaptic dysfunction.
Collapse
|
31
|
Growth differentiation factor 15 neutralization does not impact anorexia or survival in lipopolysaccharide-induced inflammation. iScience 2021; 24:102554. [PMID: 34189431 PMCID: PMC8215224 DOI: 10.1016/j.isci.2021.102554] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/24/2021] [Accepted: 05/14/2021] [Indexed: 02/02/2023] Open
Abstract
Growth differentiation factor 15 (GDF15) causes anorexia and weight loss in animal models, and higher circulating levels are associated with cachexia and reduced survival in cancer and other chronic diseases such as sepsis. To investigate the role of sepsis-induced GDF15, we examined whether GDF15 neutralization via a validated and highly potent monoclonal antibody, mAB2, modulates lipopolysaccharide (LPS)-induced anorexia, weight loss, and mortality in rodents. LPS injection transiently increased circulating GDF15 in wild-type mice, decreased food intake and body weight, and increased illness behavior and mortality at a high dose. GDF15 neutralization with mAB2 did not prevent or exacerbate any of the effects of LPS. Similarly, in GDF15 knockout mice, the LPS effect on appetite and survival was comparable with that observed in wild-type controls. Therefore, effective inhibition of circulating active GDF15 via an antibody or via gene knockout demonstrated that survival in the LPS acute inflammation model was independent of GDF15. A novel highly potent anti-GDF15 antibody, mAB2, was characterized LPS transiently increased GDF15 production in mice mAB2 did not prevent or exacerbate the effects of LPS on anorexia or survival GDF15 knockout mice showed comparable effects of LPS with wild-type controls
Collapse
|
32
|
Bondy E, Norton SA, Voss M, Marks RB, Boudreaux MJ, Treadway MT, Oltmanns TF, Bogdan R. Inflammation is associated with future depressive symptoms among older adults. Brain Behav Immun Health 2021; 13:100226. [PMID: 34589741 PMCID: PMC8474183 DOI: 10.1016/j.bbih.2021.100226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 11/07/2022] Open
Abstract
Inflammation has been reliably associated with depression. However, the directionality of this association is poorly understood, with evidence that elevated inflammation may promote and precede the development of depression, as well as arise following its expression. Using data from older adults (N = 1,072, ages 60-73) who participated in the ongoing longitudinal St. Louis Personality and Aging Network (SPAN) study, we examined whether inflammatory markers (interleukin-6: IL-6, C-reactive protein: CRP, and tumor necrosis factor α: TNFα) and depression were prospectively predictive of one another. Fasting serum samples and self-reports of depressive symptoms (Beck Depression Inventory-II) were obtained from participants at 2 sessions approximately 2 years apart. Structural equation models as well as regressions that accounted for a host of potentially confounding covariates and depression at baseline revealed that baseline IL-6 and CRP, but not baseline TNFα were associated with elevated depressive symptoms at the follow-up session (IL-6: β = 0.080, p = 0.036; CRP: β = 0.083, p = 0.03; TNFα: β = 0.039, p = 0.314). However, there was no association between baseline depressive symptoms and follow-up inflammatory markers (βs = -0.12 to -0.006, all ps > 0.05). Collectively, these data suggest that inflammation prospectively predicts depression, but depression does not predict inflammation in older age. These data add to a growing literature suggesting that inflammatory signaling may plausibly promote the development of depression.
Collapse
Affiliation(s)
- Erin Bondy
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| | - Sara A Norton
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| | - Michaela Voss
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| | - Rebecca B Marks
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| | - Michael J Boudreaux
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| | | | - Thomas F Oltmanns
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, USA
| |
Collapse
|
33
|
Basile MS, Ciurleo R, Bramanti A, Petralia MC, Fagone P, Nicoletti F, Cavalli E. Cognitive Decline in Rheumatoid Arthritis: Insight into the Molecular Pathogenetic Mechanisms. Int J Mol Sci 2021; 22:ijms22031185. [PMID: 33530359 PMCID: PMC7865873 DOI: 10.3390/ijms22031185] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/21/2022] Open
Abstract
Cognitive decline refers to a deterioration of intellectual and learning abilities and related memory problems, and is often associated with behavioral alterations, which prevents sufferers from carrying out the most common daily activities, such as maintaining normal productive interpersonal relationships, communicating, and leading an autonomous life. Numerous studies have highlighted the association between cognitive decline and autoimmune disorders, including rheumatoid arthritis (RA). RA is a chronic, inflammatory, autoimmune disease that involves systems and organs other than the bones and joints, with varying severity among patients. Here, we review the studies investigating the link between cognitive decline and RA, focusing on the main molecular pathogenetic mechanisms involved. The emerging body of data suggests that clinical, psychological, and biological factors may contribute to the pathogenesis of cognitive decline in RA, including cardiovascular complications, chronic pain, depression, inflammatory factors, changes in hormone levels, drug side effects, and genetics. Further studies are warranted in order to fully clarify the basis underlying the association between cognitive decline and RA and to find new possible diagnostic strategies and therapeutic targets for RA patients.
Collapse
Affiliation(s)
- Maria Sofia Basile
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.S.B.); (R.C.); (A.B.); (M.C.P.)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.S.B.); (R.C.); (A.B.); (M.C.P.)
| | - Alessia Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.S.B.); (R.C.); (A.B.); (M.C.P.)
| | - Maria Cristina Petralia
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (M.S.B.); (R.C.); (A.B.); (M.C.P.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (P.F.); (E.C.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (P.F.); (E.C.)
- Correspondence:
| | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy; (P.F.); (E.C.)
| |
Collapse
|
34
|
Wu L, Han Y, Zheng Z, Peng G, Liu P, Yue S, Zhu S, Chen J, Lv H, Shao L, Sheng Y, Wang Y, Li L, Li L, Wang B. Altered Gut Microbial Metabolites in Amnestic Mild Cognitive Impairment and Alzheimer's Disease: Signals in Host-Microbe Interplay. Nutrients 2021; 13:228. [PMID: 33466861 PMCID: PMC7829997 DOI: 10.3390/nu13010228] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/18/2022] Open
Abstract
Intimate metabolic host-microbiome crosstalk regulates immune, metabolic, and neuronal response in health and disease, yet remains untapped for biomarkers or intervention for disease. Our recent study identified an altered microbiome in patients with pre-onset amnestic mild cognitive impairment (aMCI) and dementia Alzheimer's disease (AD). Thus, we aimed to characterize the gut microbial metabolites among AD, aMCI, and healthy controls (HC). Here, a cohort of 77 individuals (22 aMCI, 27 AD, and 28 HC) was recruited. With the use of liquid-chromatography/gas chromatography mass spectrometry metabolomics profiling, we identified significant differences between AD and HC for tryptophan metabolites, short-chain fatty acids (SCFAs), and lithocholic acid, the majority of which correlated with altered microbiota and cognitive impairment. Notably, tryptophan disorders presented in aMCI and SCFAs decreased progressively from aMCI to AD. Importantly, indole-3-pyruvic acid, a metabolite from tryptophan, was identified as a signature for discrimination and prediction of AD, and five SCFAs for pre-onset and progression of AD. This study showed fecal-based gut microbial signatures were associated with the presence and progression of AD, providing a potential target for microbiota or dietary intervention in AD prevention and support for the host-microbe crosstalk signals in AD pathophysiology.
Collapse
Affiliation(s)
- Li Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Yuqiu Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Zhipeng Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (G.P.); (P.L.)
| | - Ping Liu
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (G.P.); (P.L.)
| | - Siqing Yue
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Research Center of Environmental Science, Zhejiang University of Technology, Hangzhou 310032, China;
| | - Shuai Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Jun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Hanying Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Lifang Shao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
| | - Yan Sheng
- Department of Ophthalmology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Liang Li
- Department of Chemistry, Alberta University, Edmondon, AB T6G 2G2, Canada;
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
- Research Units of Infectious disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou 310003, China
| | - Baohong Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.W.); (Y.H.); (Z.Z.); (S.Z.); (J.C.); (H.L.); (L.S.); (L.L.)
- Research Units of Infectious disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou 310003, China
| |
Collapse
|
35
|
Shields GS, Deer LK, Hastings PD, Hostinar CE. Adiposity, Inflammation, and Working Memory: Evidence for a Vicious Cycle. Brain Behav Immun Health 2021; 13. [PMID: 33899030 PMCID: PMC8061900 DOI: 10.1016/j.bbih.2021.100202] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Overweight and obesity constitute the fifth leading cause of preventable deaths worldwide. One pathway through which excess weight contributes to poor health outcomes is via inflammatory activity and changes in cognitive processes. Prior theory has proposed a vicious cycle whereby obesity potentiates inflammatory activity, which alters cognitive processes such as working memory, which in turn leads to a reduced ability to self-regulate and therefore manage weight. However, to date no longitudinal studies have examined this potential dynamic. In the current study, we addressed this gap by assessing the relations among fat mass, C-reactive protein (CRP), and working memory across time in a large sample of 8536 children followed through adolescence in the Avon Longitudinal Study of Parents and Children in the United Kingdom. Adiposity was quantified via dual emission x-ray absorptiometry (DEXA) at ages 9 and 15.5 years old, and inflammatory activity was indexed via circulating serum C-reactive protein (CRP) levels assessed with a high-sensitivity assay at those same ages. Working memory was assessed between these two time points, at age 10, permitting examination of the temporal relations between working memory, adiposity, and inflammatory activity. As hypothesized, we found that fat mass predicted later poor working memory, and this association was statistically mediated by CRP. Further, we found that poor working memory predicted greater subsequent fat mass and CRP, and the link between working memory and subsequent CRP was partially mediated by fat mass. These results thus could be taken to suggest the existence of a vicious cycle of mutually amplifying adiposity, inflammatory activity, and poor working memory over time. Adiposity and inflammatory activity (CRP) was measured at ages 9 and 15.5 yrs. Working memory was assessed at age 10 yrs. Adiposity predicted working memory, and this association was mediated by CRP. Working memory predicted later adiposity and CRP, this was mediated by adiposity.
Collapse
Affiliation(s)
- Grant S. Shields
- Center for Mind and Brain, University of California, Davis, CA, United States
| | - LillyBelle K. Deer
- Center for Mind and Brain, University of California, Davis, CA, United States
- Department of Psychology, University of California, Davis, CA, United States
| | - Paul D. Hastings
- Center for Mind and Brain, University of California, Davis, CA, United States
- Department of Psychology, University of California, Davis, CA, United States
| | - Camelia E. Hostinar
- Center for Mind and Brain, University of California, Davis, CA, United States
- Department of Psychology, University of California, Davis, CA, United States
- Corresponding author. Center for Mind and Brain, University of California, Davis, CA, United States.
| |
Collapse
|
36
|
Resveratrol ameliorates sevoflurane-induced cognitive impairment by activating the SIRT1/NF-κB pathway in neonatal mice. J Nutr Biochem 2021; 90:108579. [PMID: 33388350 DOI: 10.1016/j.jnutbio.2020.108579] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/19/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022]
Abstract
Sevoflurane, the most commonly used inhaled anesthetic in pediatric anesthesia, has been reported to induce cognitive impairment in developing brain in preclinical and clinical settings. However, the mechanism and therapeutic measures of this developmental neurotoxicity need to be further investigated. Resveratrol, a natural polyphenolic agent, has been reported to improve cognitive function in neurological disorders and aging models through anti-inflammatory activity. However, its effect on sevoflurane-induced cognitive impairment in developing mice remains unknown. The present study was designed to investigate the therapeutic potential of resveratrol on sevoflurane-induced cognitive impairment. Six-day-old mice received anesthesia with 3% sevoflurane 2 h daily on postnatal days (P) 6, P7 and P8. About 100 mg/kg resveratrol were intraperitoneally administered for 6 consecutive days to neonatal mice before anesthesia. Sevoflurane exposure significantly suppressed the expression of Sirtuin 1 (SIRT1) and activated microglia in hippocampi. Furthermore, the levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were markedly increased after sevoflurane exposure. Strikingly, resveratrol pretreatment ameliorated sevoflurane-induced SIRT1 inhibition and microglial activation. Of note, resveratrol reversed sevoflurane-induced imbalance of M1/M2 microglia ratio revealed by increasing mRNA level of clusters of differentiation 206 (CD206) and decreasing mRNA levels of clusters of differentiation 86 (CD86) and suppressor of cytokine signaling 3 (SOCS3). Consequently, sevoflurane-induced cognitive impairment in developing mice was ameliorated by resveratrol pretreatment. Taken together, repeated sevoflurane exposure to the developing brain resulted in SIRT1 inhibition, NF-κB acetylation, and microglial activation. Resveratrol pretreatment ameliorated cognitive impairment in developing mice received sevoflurane exposure by modulating SIRT1-NF-κB pathway in microglia. In this regard, our findings open novel directions to explore promising therapeutic targets for preventing the developmental neurotoxicity of sevoflurane.
Collapse
|
37
|
Simon LS, Taylor PC, Choy EH, Sebba A, Quebe A, Knopp KL, Porreca F. The Jak/STAT pathway: A focus on pain in rheumatoid arthritis. Semin Arthritis Rheum 2020; 51:278-284. [PMID: 33412435 DOI: 10.1016/j.semarthrit.2020.10.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/02/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
Pain is a manifestation of rheumatoid arthritis (RA) that is mediated by inflammatory and non-inflammatory mechanisms and negatively affects quality of life. Recent findings from a Phase 3 clinical trial showed that patients with RA who were treated with a Janus kinase 1 (Jak1) and Janus kinase 2 (Jak2) inhibitor achieved significantly greater improvements in pain than those treated with a tumor necrosis factor blocker; both treatments resulted in similar changes in standard clinical measures and markers of inflammation. These findings suggest that Jak1 and Jak2 inhibition may relieve pain in RA caused by inflammatory and non-inflammatory mechanisms and are consistent with the overarching involvement of the Jak-signal transducer and activator of transcription (Jak/STAT) pathway in mediating the action, expression, and regulation of a multitude of pro- and anti-inflammatory cytokines. In this review, we provide an overview of pain in RA, the underlying importance of cytokines regulated directly or indirectly by the Jak/STAT pathway, and therapeutic targeting of the Jak/STAT pathway in RA. As highlighted herein, multiple cytokines directly or indirectly regulated by the Jak/STAT pathway play important roles in mediating various mechanisms underlying pain in RA. Having a better understanding of these mechanisms may help clinicians make treatment decisions that optimize the control of inflammation and pain.
Collapse
Affiliation(s)
| | - Peter C Taylor
- Botnar Research Centre, University of Oxford, Oxford, UK
| | - Ernest H Choy
- CREATE Centre, Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | | | | | | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85718, USA.
| |
Collapse
|
38
|
Savi FF, de Oliveira A, de Medeiros GF, Bozza FA, Michels M, Sharshar T, Dal-Pizzol F, Ritter C. What animal models can tell us about long-term cognitive dysfunction following sepsis: A systematic review. Neurosci Biobehav Rev 2020; 124:386-404. [PMID: 33309906 DOI: 10.1016/j.neubiorev.2020.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 01/28/2023]
Abstract
Survivors of sepsis often develop long-term cognitive impairments. This review aimed at exploring the results of the behavioral tools and tests which have been used to evaluate cognitive dysfunction in different animal models of sepsis. Two independent investigators searched for sepsis- and cognition-related keywords. 6323 publications were found, of which 355 were selected based on their title, and 226 of these were chosen based on manuscript review. LPS was used to induce sepsis in 171 studies, while CLP was used in 55 studies. Inhibitory avoidance was the most widely used method for assessing aversive memory, followed by fear conditioning and continuous multi-trial inhibitory avoidance. With regard to non-aversive memory, most studies used the water maze, open-field, object recognition, Y-maze, plus maze, and radial maze tests. Both CLP and LPS models of sepsis were effective in inducing short- and long-term behavioral impairment. Our findings help elucidate the mechanisms involved in the pathophysiology of sepsis-induced cognitive changes, as well as the available methods and tests used to study this in animal models.
Collapse
Affiliation(s)
- Felipe Figueredo Savi
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil
| | - Alexandre de Oliveira
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil
| | | | - Fernando Augusto Bozza
- Laboratório de Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas (INI), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Monique Michels
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil
| | - Tarek Sharshar
- Laboratoire de Neuropathologie Expérimentale, Institut Pasteur, Paris, France; Department of Neuro-Intensive Care Medicine, Sainte-Anne Hospital, Paris-Descartes University, Paris, France
| | - Felipe Dal-Pizzol
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil; Laboratoire de Neuropathologie Expérimentale, Institut Pasteur, Paris, France
| | - Cristiane Ritter
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Brazil.
| |
Collapse
|
39
|
Effects of the excitation or inhibition of basal forebrain cholinergic neurons on cognitive ability in mice exposed to chronic intermittent hypoxia. Brain Res Bull 2020; 164:235-248. [DOI: 10.1016/j.brainresbull.2020.08.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/20/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
|
40
|
Tanshinone IIA suppresses lipopolysaccharide-induced neuroinflammatory responses through NF-κB/MAPKs signaling pathways in human U87 astrocytoma cells. Brain Res Bull 2020; 164:136-145. [DOI: 10.1016/j.brainresbull.2020.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023]
|
41
|
Cornelius ADA, Hosseini S, Schreier S, Fritzsch D, Weichert L, Michaelsen-Preusse K, Fendt M, Kröger A. Langat virus infection affects hippocampal neuron morphology and function in mice without disease signs. J Neuroinflammation 2020; 17:278. [PMID: 32951602 PMCID: PMC7504599 DOI: 10.1186/s12974-020-01951-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 09/10/2020] [Indexed: 12/22/2022] Open
Abstract
Background Tick-borne encephalitis virus (TBEV) is an important human pathogen that can cause the serious illness tick-borne encephalitis (TBE). Patients with clinical symptoms can suffer from severe meningoencephalitis with sequelae that include cognitive disorders and paralysis. While less than 30% of patients with clinical symptoms develop meningoencephalitis, the number of seropositive individuals in some regions indicates a much higher prevalence of TBEV infections, either with no or subclinical symptoms. The functional relevance of these subclinical TBEV infections and their influence on brain functions, such as learning and memory, has not been investigated so far. Methods To compare the effect of low and high viral replication in the brain, wildtype and Irf-7−/− mice were infected with Langat virus (LGTV), which belongs to the TBEV-serogroup. The viral burden was analyzed in the olfactory bulb and the hippocampus. Open field, elevated plus maze, and Morris water maze experiments were performed to determine the impact on anxiety-like behavior, learning, and memory formation. Spine density of hippocampal neurons and activation of microglia and astrocytes were analyzed. Results In contrast to susceptible Irf-7−/− mice, wildtype mice showed no disease signs upon LGTV infection. Detection of viral RNA in the olfactory bulb revealed CNS infections in wildtype and Irf-7−/− mice. Very low levels of viral replication were detectable in the hippocampus of wildtype mice. Although wildtype mice develop no disease signs, they showed reduced anxiety-like behavior and impaired memory formation, whereas Irf-7−/− mice were not affected. This impairment was associated with a significant decrease in spine density of neurons in the hippocampal CA1 region of wildtype mice. Microglia activation and astrogliosis were detected in the hippocampus. Conclusion In this study, we demonstrate that subclinical infections by viruses from the TBEV-serogroup affected anxiety-like behavior. Virus replication in the olfactory bulb induced far-reaching effects on hippocampal neuron morphology and impaired hippocampus-dependent learning and memory formation.
Collapse
Affiliation(s)
- Angela D A Cornelius
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany.,Present Address: Institute of Virology, Hannover Medical School, 30625, Hannover, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106, Braunschweig, Germany.,Neuroinflammation and Neurodegeneration Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Sarah Schreier
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, Leipziger Strasse 44, D-39120, Magdeburg, Germany
| | - David Fritzsch
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, Leipziger Strasse 44, D-39120, Magdeburg, Germany
| | - Loreen Weichert
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany.,Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, Leipziger Strasse 44, D-39120, Magdeburg, Germany
| | - Kristin Michaelsen-Preusse
- Department of Cellular Neurobiology, Zoological Institute, TU Braunschweig, 38106, Braunschweig, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, 39120, Magdeburg, Germany.,Center of Behavioral Brain Sciences, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Andrea Kröger
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany. .,Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, Leipziger Strasse 44, D-39120, Magdeburg, Germany. .,Center of Behavioral Brain Sciences, Otto-von-Guericke University, 39120, Magdeburg, Germany. .,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| |
Collapse
|
42
|
Interleukin-6: A neuro-active cytokine contributing to cognitive impairment in Duchenne muscular dystrophy? Cytokine 2020; 133:155134. [DOI: 10.1016/j.cyto.2020.155134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
|
43
|
Antifungal drug miconazole ameliorated memory deficits in a mouse model of LPS-induced memory loss through targeting iNOS. Cell Death Dis 2020; 11:623. [PMID: 32796824 PMCID: PMC7429861 DOI: 10.1038/s41419-020-2619-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease (AD) is closely related to neuroinflammation, and the increase in inflammatory cytokine generation and inducible nitric oxide synthase (iNOS) expression in the brain of a patient with AD is well known. Excessive cytokines can stimulate iNOS in microglia and astroglia and overproduce nitric oxide, which can be toxic to neurons. The disease–gene–drug network analysis based on the GWAS/OMIM/DEG records showed that miconazole (MCZ) affected AD through interactions with NOS. Inhibiting iNOS can reduce neuroinflammation, thus preventing AD progression. To investigate the prophylactic role of antifungal agent in the AD development, a lipopolysaccharide-induced memory disorder mouse model was used, and cognitive function was assessed by Morris water maze test and passive avoidance test. MCZ treatment significantly attenuated cognitive impairment, suppressed iNOS and cyclooxygenase-2 expression, and activation of astrocyte and microglial BV2 cells, as well as reduced cytokine levels in the brains and lipopolysaccharide-treated astrocytes and microglia BV2 cells. In further mechanism studies, Pull-down assay and iNOS luciferase activity data showed that MCZ binds to iNOS and inhibited transcriptional activity. Our results suggest that MCZ is useful for ameliorating the neuroinflammation-mediated AD progression by blocking iNOS expression.
Collapse
|
44
|
He Y, Ruganzu JB, Jin H, Peng X, Ji S, Ma Y, Zheng L, Yang W. LRP1 knockdown aggravates Aβ 1-42-stimulated microglial and astrocytic neuroinflammatory responses by modulating TLR4/NF-κB/MAPKs signaling pathways. Exp Cell Res 2020; 394:112166. [PMID: 32645395 DOI: 10.1016/j.yexcr.2020.112166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 12/22/2022]
Abstract
Neuroinflammation is an important pathological feature and an early event in the pathogenesis of Alzheimer's disease (AD), which is characterized by activation of microglia and astrocytes. Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic receptor that is abundantly expressed in neurons, microglia, and astrocytes, and plays a critical role in AD pathogenesis. There is increasing evidence to show that LRP1 regulates inflammatory responses by modulating the release of pro-inflammatory cytokines and phagocytosis. However, the effects of LRP1 on β-amyloid protein (Aβ)-induced microglial and astrocytic neuroinflammatory responses and its underlying mechanisms have not been studied in detail. In the present study, knockdown of LRP1 significantly enhanced Aβ1-42-stimulated neuroinflammation by increasing the production of pro-inflammatory cytokines in both BV2 microglial cells and mouse primary astrocytes. Furthermore, it is revealed that LRP1 knockdown further led to the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways. The phosphorylation of IκBα, p38, and JNK was significantly up-regulated in LRP1 knockdown BV2 microglial cells and primary astrocytes. Meanwhile, LRP1 knockdown increased expression of the NF-κB p65 subunit in the nucleus while decreased its expression in the cytoplasm. Besides, the upstream signaling adaptor molecules such as toll-like receptor 4 (TLR4), myeloid differentiation primary response protein 88 (MyD88), and tumor necrosis factor receptor-associated factor 6 (TRAF6) were also further increased. Moreover, blockade of NF-κB, p38, and JNK inhibited the production of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) induced by the knockdown of LRP1. Taken together, these findings indicated that LRP1 as an effective therapeutic target against AD and other neuroinflammation related diseases.
Collapse
Affiliation(s)
- Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Yanbing Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Liming Zheng
- Basic Medical Experimental Teaching Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
45
|
Mendez Luque LF, Blackmon AL, Ramanathan G, Fleischman AG. Key Role of Inflammation in Myeloproliferative Neoplasms: Instigator of Disease Initiation, Progression. and Symptoms. Curr Hematol Malig Rep 2020; 14:145-153. [PMID: 31119475 DOI: 10.1007/s11899-019-00508-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Chronic inflammation is a characteristic feature of myeloproliferative neoplasm (MPN) and impacts many aspects of the disease including initiation, progression, and symptomatology. RECENT FINDINGS The chronic inflammatory state of MPN results from disruption of immune signaling pathways leading to overproduction of inflammatory cytokines by both the neoplastic clones and bystander immune cells. This chronic inflammation may allow for the neoplastic clone to gain a selective advantage. The symptomatic burden felt by MPN patients may be a result of the chronic inflammation associated with MPN, as several cytokines have been linked with different symptoms. Pharmacologic as well as nonpharmacologic treatments of the inflammatory component of this disease may lead to decreased symptomatic burden, prevention of disease progression, and improvement in overall disease trajectory. Inflammation plays a key role in the pathogenesis of MPN and represents an important therapeutic target.
Collapse
Affiliation(s)
- Laura F Mendez Luque
- Department of Biological Chemistry, University of California Irvine, 839 Medical Sciences Rd, Sprague Hall 126, Irvine, CA, 92617, USA
| | - Amanda L Blackmon
- Division of Hematology/Oncology, Department of Medicine, University of California Irvine, 839 Medical Sciences Rd, Sprague Hall 126, Irvine, CA, 92617, USA
| | - Gajalakshmi Ramanathan
- Division of Hematology/Oncology, Department of Medicine, University of California Irvine, 839 Medical Sciences Rd, Sprague Hall 126, Irvine, CA, 92617, USA
| | - Angela G Fleischman
- Department of Biological Chemistry, University of California Irvine, 839 Medical Sciences Rd, Sprague Hall 126, Irvine, CA, 92617, USA. .,Division of Hematology/Oncology, Department of Medicine, University of California Irvine, 839 Medical Sciences Rd, Sprague Hall 126, Irvine, CA, 92617, USA. .,Chao Family Comprehensive Cancer Center, University of California Irvine, 839 Medical Sciences Rd, Sprague Hall 126, Irvine, CA, 92617, USA.
| |
Collapse
|
46
|
Gruol DL, Hernandez RV, Roberts A. Alcohol Enhances Responses to High Frequency Stimulation in Hippocampus from Transgenic Mice with Increased Astrocyte Expression of IL-6. Cell Mol Neurobiol 2020; 41:1299-1310. [PMID: 32562098 DOI: 10.1007/s10571-020-00902-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Recent studies show that alcohol exposure can induce glial production of neuroimmune factors in the CNS. Of these, IL-6 has gained attention because it is involved in a number of important physiological and pathophysiological processes that could be affected by alcohol-induced CNS production of IL-6, particularly under conditions of excessive alcohol use. For example, IL-6 has been shown to play a role in hippocampal behaviors and synaptic plasticity (long-term potentiation; LTP) associated with memory and learning. Surprisingly, in our in vitro studies of LTP at the Schaffer collateral to CA1 pyramidal neuron synapse in hippocampus from transgenic mice that express elevated levels of astrocyte produced IL-6 (TG), LTP was not altered by the increased levels of IL-6. However, exposure to acute alcohol revealed neuroadaptive changes that served to protect LTP against the alcohol-induced reduction of LTP observed in hippocampus from non-transgenic control mice (WT). Here we examined the induction phase of LTP to assess if presynaptic neuroadaptive changes occurred in the hippocampus of TG mice that contributed to the resistance of LTP to alcohol. Results are consistent with a role for IL-6-induced neuroadaptive effects on presynaptic mechanisms involved in transmitter release in the resistance of LTP to alcohol in hippocampus from the TG mice. These actions are important with respect to a role for IL-6 in physiological and pathophysiological processes in the CNS and in CNS actions of alcohol, especially when excessive alcohol used is comorbid with conditions associated with elevated levels of IL-6 in the CNS.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Ruben V Hernandez
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
47
|
Shu H, Wang M, Song M, Sun Y, Shen X, Zhang J, Jin X. Acute Nicotine Treatment Alleviates LPS-Induced Impairment of Fear Memory Reconsolidation Through AMPK Activation and CRTC1 Upregulation in Hippocampus. Int J Neuropsychopharmacol 2020; 23:687-699. [PMID: 32516360 PMCID: PMC7727489 DOI: 10.1093/ijnp/pyaa043] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Fear memory is a fundamental capability for animals and humans to survive. Its impairment results in the disability to avoid danger. When memory is reactivated, a reconsolidation process, which can be disrupted by various stimuli, including inflammation, is required to become permanent. Nicotine has been shown to improve cognitive deficits induced by inflammation and other stimuli. Therefore, in the present study, we investigated the effect of nicotine on lipopolysaccharide (LPS)-induced impairment of fear memory reconsolidation and the underlying mechanism. METHODS Step-through inhibitory avoidance task was recruited to study fear memory of rat, i.p. LPS (0.5 mg/kg) treatment was used to induce inflammation, and western blot and immunostaining were applied to detect protein expression and distribution in medial prefrontal cortex and hippocampus. RESULTS Our data showed that LPS induced fear memory reconsolidation impairment without affecting retrieval. In addition, LPS significantly increased inflammation factors tumor necrosis factor-α and interleukin-1 beta and decreased CREB-regulated transcription coactivator 1 (CRTC1) expression and adenosine monophosphate-activated protein kinase (AMPK) activation in hippocampus. More importantly, LPS significantly decreased CRTC1 expression and AMPK activation in neurons by activating microglia cells. Of note, either nicotine treatment or activation of AMPK by intracerebroventricular infusion of metformin reduced LPS-induced impairment of fear memory reconsolidation and ameliorated inflammation factor tumor necrosis factor-α and interleukin-1 beta as well as the expression of CRTC1. CONCLUSIONS In conclusion, our results showed that acute nicotine treatment alleviates LPS-induced impairment of fear memory reconsolidation through activation of AMPK and upregulation of CRTC1 in hippocampus.
Collapse
Affiliation(s)
- Hui Shu
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China,Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, China
| | - Mengwei Wang
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Song
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanyun Sun
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xianzhi Shen
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfang Zhang
- School of Medicine, Ningbo University, Ningbo, China,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China,Correspondence: Xinchun Jin, PhD, School of Basic Medical Sciences, Capital Medical University, Beijing, 100054, China () or Junfang Zhang, PhD, Ningbo University, Ningbo, 315211, China ()
| | - Xinchun Jin
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China,Correspondence: Xinchun Jin, PhD, School of Basic Medical Sciences, Capital Medical University, Beijing, 100054, China () or Junfang Zhang, PhD, Ningbo University, Ningbo, 315211, China ()
| |
Collapse
|
48
|
Zhang W, Xiong BR, Zhang LQ, Huang X, Zhou WC, Zou Q, Manyande A, Wang J, Tian XB, Tian YK. Disruption of the GABAergic system contributes to the development of perioperative neurocognitive disorders after anesthesia and surgery in aged mice. CNS Neurosci Ther 2020; 26:913-924. [PMID: 32488976 PMCID: PMC7415208 DOI: 10.1111/cns.13388] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Aims Perioperative neurocognitive disorders (PND) are associated with cognitive impairment in the preoperative or postoperative period, and neuroinflammation is thought to be the most important mechanisms especially during the postoperative period. The GABAergic system is easily disrupted by neuroinflammation. This study investigated the impact of the GABAergic system on PND after anesthesia and surgery. Methods An animal model of laparotomy with inhalation anesthesia in 16‐month‐old mice was addressed. Effects of the GABAergic system were assessed using biochemical analysis. Pharmacological blocking of α5GABAARs or P38 mitogen‐activated protein kinase (MAPK) were applied to investigate the effects of the GABAergic system. Results After laparotomy, the hippocampus‐dependent memory and long‐term potentiation were impaired, the levels of IL‐6, IL‐1β and TNF‐α up‐regulated in the hippocampus, the concentration of GABA decreased, and the protein levels of the surface α5GABAARs up‐regulated. Pharmacological blocking of α5GABAARs with L655,708 alleviated laparotomy induced cognitive deficits. Further studies found that the P38 MAPK signaling pathway was involved and pharmacological blocking with SB203,580 alleviated memory dysfunctions. Conclusions Anesthesia and surgery caused neuroinflammation in the hippocampus, which consequently disrupted the GABAergic system, increased the expressions of surface α5GABAARs especially through the P38 MAPK signaling pathway, and eventually led to hippocampus‐dependent memory dysfunctions.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing-Rui Xiong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Long-Qing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Huang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Chang Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Zou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, United Kingdom
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Xue-Bi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ke Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
Abstract
This scientific commentary refers to ‘The cognitive consequences of the COVID-19 epidemic: collateral damage?’, by Ritchie et al. (https://doi.org/10.1093/braincomms/fcaa069).
Collapse
Affiliation(s)
- Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| |
Collapse
|
50
|
Mac Giollabhui N, Swistun D, Murray S, Moriarity DP, Kautz MM, Ellman LM, Olino TM, Coe CL, Abramson LY, Alloy LB. Executive dysfunction in depression in adolescence: the role of inflammation and higher body mass. Psychol Med 2020; 50:683-691. [PMID: 30919789 PMCID: PMC6765453 DOI: 10.1017/s0033291719000564] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND There is substantial evidence that many depressed individuals experience impaired executive functioning. Understanding the causes of executive dysfunction in depression is clinically important because cognitive impairment is a substantial contributor to functional impairment. This study investigated whether elevated levels of an inflammatory cytokine [interleukin-6 (IL-6)] and/or higher body mass index (BMI) concurrently and/or prospectively accounted for the relationship between depressive symptoms and impaired executive functioning in adolescents. METHODS A diverse, community sample of adolescents (N = 288; mean age = 16.33; 51.4% female; 59.0% African-American) completed assessments of height and weight, IL-6, depressive symptoms, and self-report/behavioral measures of executive functioning (selective attention, switching attention) and future orientation annually over 3 years. Adolescents experiencing acute illness or medical conditions that affect inflammation were excluded from analyses. Path analysis within a structural equation modeling framework simultaneously examined the concurrent and prospective relationships between BMI, IL-6, depressive symptoms, and the measures of cognitive functioning across three timepoints. RESULTS Across all timepoints, higher BMI was prospectively associated with higher levels of IL-6 and depressive symptoms, while higher levels of IL-6 were associated with worse performance on three behavioral and self-report measures of cognitive functioning. Higher depressive symptoms also were prospectively associated with elevated IL-6 and both higher depressive symptoms and a higher BMI predicted worse future executive functioning via increased IL-6. CONCLUSIONS More severe depressive symptoms and increased BMI may disrupt executive functioning via elevated IL-6.
Collapse
Affiliation(s)
| | - Dominika Swistun
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Susan Murray
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | | | - Marin M Kautz
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Lauren M Ellman
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Thomas M Olino
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Christopher L Coe
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Lyn Y Abramson
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Lauren B Alloy
- Department of Psychology, Temple University, Philadelphia, PA, USA
| |
Collapse
|