151
|
Gereau GB, Torruella-Suárez ML, Sizer SE, Xia M, Zhou D, Wykoff LA, Teklezghi AT, Alvarez-Pamir A, Boyt KM, Kash TL, McElligott ZA. GABA release from central amygdala neurotensin neurons differentially modulates ethanol consumption in male and female mice. Neuropsychopharmacology 2024; 49:1151-1161. [PMID: 38418568 PMCID: PMC11109172 DOI: 10.1038/s41386-024-01830-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024]
Abstract
The central nucleus of the amygdala is known to play key roles in alcohol use and affect. Neurotensin neurons in the central nucleus of the amygdala have been shown to regulate alcohol drinking in male mice. However, little is known about which neurotransmitters released by these cells drive alcohol consumption or whether these cells drive alcohol consumption in female mice. Here we show that knockdown of GABA release from central amygdala neurotensin neurons using a Nts-cre-dependent vGAT-shRNA-based AAV strategy reduces alcohol drinking in male, but not female, mice. This manipulation did not impact avoidance behavior, except in a fasted novelty-suppressed feeding test, in which vGAT shRNA mice demonstrated increased latency to feed on a familiar high-value food reward, an effect driven by male mice. In contrast, vGAT shRNA female mice showed heightened sensitivity to thermal stimulation. These data show a role for GABA release from central amygdala neurotensin neurons in modulating consumption of rewarding substances in different motivational states.
Collapse
Affiliation(s)
- Graydon B Gereau
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - María L Torruella-Suárez
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Graduate Program in Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah E Sizer
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mengfan Xia
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Diana Zhou
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luke A Wykoff
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adonay T Teklezghi
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ali Alvarez-Pamir
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristen M Boyt
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zoé A McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
152
|
Qiu O, Zhao J, Shi Z, Li H, Wang S, Liao K, Tang M, Xie J, Huang X, Zhang W, Zhou L, Yang X, Zhou Z, Xu L, Huang R, Miao Y, Qiu Y, Lin Y. Asparagine endopeptidase deficiency mitigates radiation-induced brain injury by suppressing microglia-mediated neuronal senescence. iScience 2024; 27:109698. [PMID: 38655198 PMCID: PMC11035374 DOI: 10.1016/j.isci.2024.109698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Mounting evidence supports the role of neuroinflammation in radiation-induced brain injury (RIBI), a chronic disease characterized by delayed and progressive neurological impairment. Asparagine endopeptidase (AEP), also known as legumain (LGMN), participates in multiple malignancies and neurodegenerative diseases and may potentially be involved in RIBI. Here, we found AEP expression was substantially elevated in the cortex and hippocampus of wild-type (Lgmn+/+) mice following whole-brain irradiation. Lgmn knockout (Lgmn-/-) alleviated neurological impairment caused by whole-brain irradiation by suppressing neuronal senescence. Bulk RNA and metabolomic sequencing revealed AEP's involvement in the antigen processing and presentation pathway and neuroinflammation. This was further confirmed by co-culturing Lgmn+/+ primary neurons with the conditioned media derived from irradiated Lgmn+/+ or Lgmn-/- primary microglia. Furthermore, esomeprazole inhibited the enzymatic activity of AEP and RIBI. These findings identified AEP as a critical factor of neuroinflammation in RIBI, highlighting the prospect of targeting AEP as a therapeutic approach.
Collapse
Affiliation(s)
- Ouwen Qiu
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Jianyi Zhao
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Zhonggang Shi
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Huan Li
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Siyuan Wang
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Keman Liao
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Minchao Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Guangxi 530021, P.R. China
| | - Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi 530007, P.R. China
| | - Xi Huang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Guangxi 530021, P.R. China
| | - Wenrui Zhang
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Li Zhou
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Xi Yang
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Zhiyi Zhou
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Lei Xu
- Department of Radiation, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Renhua Huang
- Department of Radiation, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yifeng Miao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yongming Qiu
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| | - Yingying Lin
- Brain Injury Center, Shanghai Institute of Head Trauma, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, P.R. China
| |
Collapse
|
153
|
Schwartzer JJ, Church JS, Russo JN, Ragoonaden S. Offspring behavioral outcomes following maternal allergic asthma in the IL-4-deficient mouse. J Neuroimmunol 2024; 390:578341. [PMID: 38613873 PMCID: PMC11088503 DOI: 10.1016/j.jneuroim.2024.578341] [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: 01/14/2024] [Revised: 03/16/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
Maternal allergic asthma (MAA) during pregnancy has been associated with increased risk of neurodevelopmental disorders in humans, and rodent studies have demonstrated that inducing a T helper-2-mediated allergic response during pregnancy leads to an offspring behavioral phenotype characterized by decreased social interaction and increased stereotypies. The interleukin (IL)-4 cytokine is hypothesized to mediate the neurobehavioral impact of MAA on offspring. Utilizing IL-4 knockout mice, this study assessed whether MAA without IL-4 signaling would still impart behavioral deficits. C57 and IL-4 knockout female mice were sensitized to ovalbumin, exposed to repeated MAA inductions, and their offspring performed social, cognitive, and motor tasks. Only C57 offspring of MAA dams displayed social and cognitive deficits, while IL-4 knockout mice showed altered motor activity compared with C57 mice. These findings highlight a key role for IL-4 signaling in MAA-induced behavioral deficits and more broadly in normal brain development.
Collapse
Affiliation(s)
- Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA.
| | - Jamie S Church
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Jenna N Russo
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Shanthini Ragoonaden
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| |
Collapse
|
154
|
Hou K, Pan H, Shahpasand-Kroner H, Hu C, Abskharon R, Seidler P, Mekkittikul M, Balbirnie M, Lantz C, Sawaya MR, Dolinsky JL, Jones M, Zuo X, Loo JA, Frautschy S, Cole G, Eisenberg DS. D-peptide-magnetic nanoparticles fragment tau fibrils and rescue behavioral deficits in a mouse model of Alzheimer's disease. SCIENCE ADVANCES 2024; 10:eadl2991. [PMID: 38691615 PMCID: PMC11062580 DOI: 10.1126/sciadv.adl2991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/29/2024] [Indexed: 05/03/2024]
Abstract
Amyloid fibrils of tau are increasingly accepted as a cause of neuronal death and brain atrophy in Alzheimer's disease (AD). Diminishing tau aggregation is a promising strategy in the search for efficacious AD therapeutics. Previously, our laboratory designed a six-residue, nonnatural amino acid inhibitor D-TLKIVW peptide (6-DP), which can prevent tau aggregation in vitro. However, it cannot block cell-to-cell transmission of tau aggregation. Here, we find D-TLKIVWC (7-DP), a d-cysteine extension of 6-DP, not only prevents tau aggregation but also fragments tau fibrils extracted from AD brains to neutralize their seeding ability and protect neuronal cells from tau-induced toxicity. To facilitate the transport of 7-DP across the blood-brain barrier, we conjugated it to magnetic nanoparticles (MNPs). The MNPs-DP complex retains the inhibition and fragmentation properties of 7-DP alone. Ten weeks of MNPs-DP treatment appear to reverse neurological deficits in the PS19 mouse model of AD. This work offers a direction for development of therapies to target tau fibrils.
Collapse
Affiliation(s)
- Ke Hou
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| | - Hope Pan
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| | - Hedieh Shahpasand-Kroner
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research and Clinical Core, Los Angeles, CA, USA
- Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Carolyn Hu
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| | - Romany Abskharon
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| | - Paul Seidler
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - Marisa Mekkittikul
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research and Clinical Core, Los Angeles, CA, USA
- Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Melinda Balbirnie
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| | - Carter Lantz
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Michael R. Sawaya
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Joshua L. Dolinsky
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| | - Mychica Jones
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research and Clinical Core, Los Angeles, CA, USA
- Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Xiaohong Zuo
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research and Clinical Core, Los Angeles, CA, USA
- Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Sally Frautschy
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research and Clinical Core, Los Angeles, CA, USA
- Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Greg Cole
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research and Clinical Core, Los Angeles, CA, USA
- Department of Medicine, UCLA, Los Angeles, CA, USA
| | - David S. Eisenberg
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Howard Hughes Medical Institute, Los Angeles, CA, USA
| |
Collapse
|
155
|
Zhang Y, Wang Y, Wei R, Li X, Luo B, Zhang J, Zhang K, Fang S, Liu X, Chen G. Mitochondrial antioxidant elamipretide improves learning and memory impairment induced by chronic sleep deprivation in mice. Brain Behav 2024; 14:e3508. [PMID: 38688894 PMCID: PMC11061203 DOI: 10.1002/brb3.3508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/10/2024] [Accepted: 03/16/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The inflammation and synaptic dysfunction induced by mitochondrial dysfunction play essential roles in the learning and memory impairment associated with sleep dysfunction. Elamipretide (SS-31), a novel mitochondrion-targeted antioxidant, was proven to improve mitochondrial dysfunction, the inflammatory response, synaptic dysfunction, and cognitive impairment in models of cerebral ischemia, sepsis, and type 2 diabetes. However, the potential for SS-31 to improve the cognitive impairment induced by chronic sleep deprivation (CSD) and its underlying mechanisms is unknown. METHODS Adult c57BL/6J mice were subjected to CSD for 21 days using an activity wheel accompanied by daily intraperitoneal injection of SS-31 (5 mg/kg). The novel object recognition and Morris water maze test were used to evaluate hippocampus-dependent cognitive function. Western blotting and reverse transcription-quantitative polymerase chain reaction assays were used to determine the effects of CSD and SS-31 on markers of mitochondria, inflammation response, and synaptic function. Enzyme-linked immunosorbent assays were used to examine the levels of proinflammatory cytokines. RESULTS SS-31 could improve the cognitive impairment induced by CSD. In particular, SS-31 treatment restored the CSD-induced decrease in sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor γ coactivator alpha levels and the increase in levels nuclear factor kappa-B and inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha. Furthermore, SS-31 significantly increased the levels of brain-derived neurotrophic factor, postsynaptic density protein-95, and synaptophysin in CSD mice. CONCLUSION Taken together, these results suggest that SS-31 could improve CSD-induced mitochondrial biogenesis dysfunction, inflammatory response, synaptic dysfunction, and cognitive impairment by increasing SIRT1 expression levels.
Collapse
Affiliation(s)
- Yue‐Ming Zhang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Ya‐Tao Wang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Ru‐Meng Wei
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Xue‐Yan Li
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Bao‐Ling Luo
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Jing‐Ya Zhang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Kai‐Xuan Zhang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Shi‐Kun Fang
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Xue‐Chun Liu
- Department of NeurologyThe Second People's Hospital of Hefei and Affiliated Hefei Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Gui‐Hai Chen
- Department of Neurology (Sleep Disorders)The Affiliated Chaohu Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| |
Collapse
|
156
|
Pei J, Zhang C, Zhang X, Zhao Z, Zhang X, Yuan Y. Low-intensity transcranial ultrasound stimulation improves memory in vascular dementia by enhancing neuronal activity and promoting spine formation. Neuroimage 2024; 291:120584. [PMID: 38522806 DOI: 10.1016/j.neuroimage.2024.120584] [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: 11/23/2023] [Revised: 03/01/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024] Open
Abstract
Memory is closely associated with neuronal activity and dendritic spine formation. Low-intensity transcranial ultrasound stimulation (TUS) improves the memory of individuals with vascular dementia (VD). However, it is unclear whether neuronal activity and dendritic spine formation under ultrasound stimulation are involved in memory improvement in VD. In this study, we found that seven days of TUS improved memory in VD model while simultaneously increasing pyramidal neuron activity, promoting dendritic spine formation, and reducing dendritic spine elimination. These effects lasted for 7 days but disappeared on 14 d after TUS. Neuronal activity and dendritic spine formation strongly corresponded to improvements in memory behavior over time. In addition, we also found that the memory, neuronal activity and dendritic spine of VD mice cannot be restored again by TUS of 7 days after 28 d. Collectively, these findings suggest that TUS increases neuronal activity and promotes dendritic spine formation and is thus important for improving memory in patients with VD.
Collapse
Affiliation(s)
- Jiamin Pei
- School of Electrical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China
| | - Cong Zhang
- Department of Neurology, Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, The Second Hospital of Hebei Medical University, No.215 Heping Road, Shijiazhuang 050000, China
| | - Xiao Zhang
- Department of Neurology, Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, The Second Hospital of Hebei Medical University, No.215 Heping Road, Shijiazhuang 050000, China
| | - Zhe Zhao
- School of Electrical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China
| | - Xiangjian Zhang
- Department of Neurology, Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, The Second Hospital of Hebei Medical University, No.215 Heping Road, Shijiazhuang 050000, China.
| | - Yi Yuan
- School of Electrical Engineering, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China; Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, No.438 Hebei Street, Qinhuangdao 066004, China.
| |
Collapse
|
157
|
Zhang T, Chu Y, Wang Y, Wang Y, Wang J, Ji X, Zhang G, Shi G, Cui R, Kang Y. Testosterone deficiency worsens mitochondrial dysfunction in APP/PS1 mice. Front Aging Neurosci 2024; 16:1390915. [PMID: 38752208 PMCID: PMC11094339 DOI: 10.3389/fnagi.2024.1390915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
Background Recent studies show testosterone (T) deficiency worsens cognitive impairment in Alzheimer's disease (AD) patients. Mitochondrial dysfunction, as an early event of AD, is becoming critical hallmark of AD pathogenesis. However, currently, whether T deficiency exacerbates mitochondrial dysfunction of men with AD remains unclear. Objective The purpose of this study is to explore the effects of T deficiency on mitochondrial dysfunction of male AD mouse models and its potential mechanisms. Methods Alzheimer's disease animal model with T deficiency was performed by castration to 3-month-old male APP/PS1 mice. Hippocampal mitochondrial function of mice was analyzed by spectrophotometry and flow cytometry. The gene expression levels related to mitochondrial biogenesis and mitochondrial dynamics were determined through quantitative real-time PCR (qPCR) and western blot analysis. SH-SY5Y cells treated with flutamide, T and/or H2O2 were processed for analyzing the potential mechanisms of T on mitochondrial dysfunction. Results Testosterone deficiency significantly aggravated the cognitive deficits and hippocampal pathologic damage of male APP/PS1 mice. These effects were consistent with exacerbated mitochondrial dysfunction by gonadectomy to male APP/PS1 mice, reflected by further increase in oxidative damage and decrease in mitochondrial membrane potential, complex IV activity and ATP levels. More importantly, T deficiency induced the exacerbation of compromised mitochondrial homeostasis in male APP/PS1 mice by exerting detrimental effects on mitochondrial biogenesis and mitochondrial dynamics at mRNA and protein level, leading to more defective mitochondria accumulated in the hippocampus. In vitro studies using SH-SY5Y cells validated T's protective effects on the H2O2-induced mitochondrial dysfunction, mitochondrial biogenesis impairment, and mitochondrial dynamics imbalance. Administering androgen receptor (AR) antagonist flutamide weakened the beneficial effects of T pretreatment on H2O2-treated SH-SY5Y cells, demonstrating a critical role of classical AR pathway in maintaining mitochondrial function. Conclusion Testosterone deficiency exacerbates hippocampal mitochondrial dysfunction of male APP/PS1 mice by accumulating more defective mitochondria. Thus, appropriate T levels in the early stage of AD might be beneficial in delaying AD pathology by improving mitochondrial biogenesis and mitochondrial dynamics.
Collapse
Affiliation(s)
- Tianyun Zhang
- Postdoctoral Research Station of Biology, Hebei Medical University, Shijiazhuang, China
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yun Chu
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yue Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Yu Wang
- Postdoctoral Research Station of Biology, Hebei Medical University, Shijiazhuang, China
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
| | - Jinyang Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Department of Neurology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoming Ji
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Guoliang Zhang
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Geming Shi
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Rui Cui
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Yunxiao Kang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
158
|
Chen Z, Yu J, Wang H, Xu P, Fan L, Sun F, Huang S, Zhang P, Huang H, Gu S, Zhang B, Zhou Y, Wan X, Pei G, Xu HE, Cheng J, Wang S. Flexible scaffold-based cheminformatics approach for polypharmacological drug design. Cell 2024; 187:2194-2208.e22. [PMID: 38552625 DOI: 10.1016/j.cell.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 04/28/2024]
Abstract
Effective treatments for complex central nervous system (CNS) disorders require drugs with polypharmacology and multifunctionality, yet designing such drugs remains a challenge. Here, we present a flexible scaffold-based cheminformatics approach (FSCA) for the rational design of polypharmacological drugs. FSCA involves fitting a flexible scaffold to different receptors using different binding poses, as exemplified by IHCH-7179, which adopted a "bending-down" binding pose at 5-HT2AR to act as an antagonist and a "stretching-up" binding pose at 5-HT1AR to function as an agonist. IHCH-7179 demonstrated promising results in alleviating cognitive deficits and psychoactive symptoms in mice by blocking 5-HT2AR for psychoactive symptoms and activating 5-HT1AR to alleviate cognitive deficits. By analyzing aminergic receptor structures, we identified two featured motifs, the "agonist filter" and "conformation shaper," which determine ligand binding pose and predict activity at aminergic receptors. With these motifs, FSCA can be applied to the design of polypharmacological ligands at other receptors.
Collapse
Affiliation(s)
- Zhangcheng Chen
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Yu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Huan Wang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Peiyu Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Luyu Fan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Fengxiu Sun
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Sijie Huang
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pei Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Shuo Gu
- ComMedX, Beijing 100094, China
| | | | - Yue Zhou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Gang Pei
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Sheng Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
159
|
Dutton A, Patel CD, Taylor SA, Garland CR, Turnbaugh EM, Alers-Velazquez R, Mehrbach J, Nautiyal KM, Leib DA. Asymptomatic neonatal herpes simplex virus infection in mice leads to long-term cognitive impairment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590596. [PMID: 38712140 PMCID: PMC11071430 DOI: 10.1101/2024.04.22.590596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Neonatal herpes simplex virus (nHSV) is a devastating infection impacting approximately 14,000 newborns globally each year. Infection is associated with high neurologic morbidity and mortality, making early intervention and treatment critical. Clinical outcomes of symptomatic nHSV infections are well-studied, but little is known about the frequency of, or outcomes following, sub-clinical or asymptomatic nHSV. Given the ubiquitous nature of HSV infection and frequency of asymptomatic shedding in adults, subclinical infections are underreported, yet could contribute to long-term neurological damage. To assess potential neurological morbidity associated with subclinical nHSV infection, we developed a low-dose (100 PFU) HSV infection protocol in neonatal C57BL/6 mice. At this dose, HSV DNA was detected in the brain by PCR but was not associated with acute clinical symptoms. However, months after initial inoculation with 100 PFU of HSV, we observed impaired mouse performance on a range of cognitive and memory performance tasks. Memory impairment was induced by infection with either HSV-1 or HSV-2 wild-type viruses, but not by a viral mutant lacking the autophagy-modulating Beclin-binding domain of the neurovirulence gene γ34.5. Retroviral expression of wild type γ34.5 gene led to behavioral pathology in mice, suggesting that γ34.5 expression may be sufficient to cause cognitive impairment. Maternal immunization and HSV-specific antibody treatment prevented offspring from developing neurological sequelae following nHSV-1 infection. Altogether, these results support the idea that subclinical neonatal infections may lead to cognitive decline in adulthood, with possible profound implications for research on human neurodegenerative disorders such as Alzheimer's Disease.
Collapse
|
160
|
Qu Y, Wei C, Du P, Che W, Zhang C, Ouyang W, Bian Y, Xu F, Hu B, Du K, Wu H, Liu J, Liu Q. Integration of cognitive tasks into artificial general intelligence test for large models. iScience 2024; 27:109550. [PMID: 38595796 PMCID: PMC11001637 DOI: 10.1016/j.isci.2024.109550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
Abstract
During the evolution of large models, performance evaluation is necessary for assessing their capabilities. However, current model evaluations mainly rely on specific tasks and datasets, lacking a united framework for assessing the multidimensional intelligence of large models. In this perspective, we advocate for a comprehensive framework of cognitive science-inspired artificial general intelligence (AGI) tests, including crystallized, fluid, social, and embodied intelligence. The AGI tests consist of well-designed cognitive tests adopted from human intelligence tests, and then naturally encapsulates into an immersive virtual community. We propose increasing the complexity of AGI testing tasks commensurate with advancements in large models and emphasizing the necessity for the interpretation of test results to avoid false negatives and false positives. We believe that cognitive science-inspired AGI tests will effectively guide the targeted improvement of large models in specific dimensions of intelligence and accelerate the integration of large models into human society.
Collapse
Affiliation(s)
- Youzhi Qu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chen Wei
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Penghui Du
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wenxin Che
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chi Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | | | | | - Feiyang Xu
- iFLYTEK AI Research, Hefei 230088, China
| | - Bin Hu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Kai Du
- Institute for Artificial Intelligence, Peking University, Beijing 100871, China
| | - Haiyan Wu
- Centre for Cognitive and Brain Sciences and Department of Psychology, University of Macau, Macau 999078, China
| | - Jia Liu
- Department of Psychology, Tsinghua University, Beijing 100084, China
| | - Quanying Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
161
|
Sultanakhmetov G, Limlingan SJM, Fukuchi A, Tsuda K, Suzuki H, Kato I, Saito T, Weitemier AZ, Ando K. Mark4 ablation attenuates pathological phenotypes in a mouse model of tauopathy. Brain Commun 2024; 6:fcae136. [PMID: 38712317 PMCID: PMC11073748 DOI: 10.1093/braincomms/fcae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/20/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Accumulation of abnormally phosphorylated tau proteins is linked to various neurodegenerative diseases, including Alzheimer's disease and frontotemporal dementia. Microtubule affinity-regulating kinase 4 (MARK4) has been genetically and pathologically associated with Alzheimer's disease and reported to enhance tau phosphorylation and toxicity in Drosophila and mouse traumatic brain-injury models but not in mammalian tauopathy models. To investigate the role of MARK4 in tau-mediated neuropathology, we crossed P301S tauopathy model (PS19) and Mark4 knockout mice. We performed behaviour, biochemical and histology analyses to evaluate changes in PS19 pathological phenotype with and without Mark4. Here, we demonstrated that Mark4 deletion ameliorated the tau pathology in a mouse model of tauopathy. In particular, we found that PS19 with Mark4 knockout showed improved mortality and memory compared with those bearing an intact Mark4 gene. These phenotypes were accompanied by reduced neurodegeneration and astrogliosis in response to the reduction of pathological forms of tau, such as those phosphorylated at Ser356, AT8-positive tau and thioflavin S-positive tau. Our data indicate that MARK4 critically contributes to tau-mediated neuropathology, suggesting that MARK4 inhibition may serve as a therapeutic avenue for tauopathies.
Collapse
Affiliation(s)
- Grigorii Sultanakhmetov
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Sophia Jobien M Limlingan
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Aoi Fukuchi
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Keisuke Tsuda
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Hirokazu Suzuki
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Iori Kato
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Taro Saito
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Adam Z Weitemier
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
162
|
Arozal W, Safutra MS, Barinda AJ, Hardi H, Dwita NC, Lee HJ. Comparative Neuroprotective Effects of Moringa oleifera Seed Oil and Aqueous Extract on Cognitive Functions on a High-Fat, High-Fructose Diet Mice: Focus on Senescence Markers. ScientificWorldJournal 2024; 2024:8034401. [PMID: 38633104 PMCID: PMC11022517 DOI: 10.1155/2024/8034401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/22/2024] [Accepted: 03/30/2024] [Indexed: 04/19/2024] Open
Abstract
Several studies have demonstrated that Moringa oleifera (MO) has different pharmacological properties, including neuroprotective effects. However, the role of MO in preventing brain impairment in high-fat, high-fructose diet (HFFD) remains unknown. This study aimed to investigate the neuroprotective effects of MO leaves aqueous extract (MOE) and moringa seed oil (MOO) against brain impairment in mice with HFFD. Twenty-eight male mice were randomly divided into four groups: normal diet, HFFD, HFFD + MOE 500 mg/kgBW, and HFFD + MOO 2 mL/kgBW. Cognitive function was assessed using the Y-maze and novel object recognition (NOR) tests. The p16, p21, and BDNF expressions were analyzed using the RT-PCR method. Senescence-associated beta-galactosidase (SA-β-gal) staining in the brain was also performed. The results showed that administration of MOE or MOO could increase the percentage of alternation and recognition of new objects, prevent the increase of p16 and p21 expression, and ameliorate SA-β-Gal activity in the brain. MOO, but not MOE, increased BDNF expression in senescence brains isolated from HFFD mice. The findings indicate that MOO and MOE possess neuroprotective properties, with MOO demonstrating a greater ability to inhibit the brain senescence process compared to MOE.
Collapse
Affiliation(s)
- Wawaimuli Arozal
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Agian Jeffilano Barinda
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Metabolic Cardiovascular and Aging Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Harri Hardi
- Clinical Pharmacology Specialist Study Program, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Nounik Cheri Dwita
- Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Hee J. Lee
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
163
|
Ren YR, Cui WT, Jiang KL, He KQ, Lu YM, Chen Y, Pan WJ. Protective Mechanism of Polysaccharide ORP-1 Isolated from Oudemansiella raphanipes against Age-Related Cognitive Decline through the Microbiota-Gut-Brain Axis. Mol Nutr Food Res 2024; 68:e2300739. [PMID: 38528314 DOI: 10.1002/mnfr.202300739] [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: 10/18/2023] [Revised: 01/31/2024] [Indexed: 03/27/2024]
Abstract
Age-related cognitive decline is primarily attributed to the progressive weakening of synaptic function and loss of synapses, while age-related gut microbial dysbiosis is known to impair synaptic plasticity and cognitive behavior by metabolic alterations. To improve the health of the elderly, the protective mechanisms of Oudemansiella raphanipes polysaccharide (ORP-1) against age-related cognitive decline are investigated. The results demonstrate that ORP-1 and its gut microbiota-derived metabolites SCFAs restore a healthy gut microbial population to handle age-related gut microbiota dysbiosis mainly by increasing the abundance of beneficial bacteria Dubosiella, Clostridiales, and Prevotellaceae and reducing the abundance of harmful bacteria Desulfovibrio, strengthen intestinal barrier integrity by abolishing age-related alterations of tight junction (TJ) and mucin 2 (MUC2) proteins expression, diminish age-dependent increase in circulating inflammatory factors, ameliorate cognitive decline by reversing memory- and synaptic plasticity-related proteins levels, and restrain hyperactivation of microglia-mediated synapse engulfment and neuroinflammation. These findings expand the understanding of prebiotic-microbiota-host interactions.
Collapse
Affiliation(s)
- Yu-Ru Ren
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
| | - Wen-Ting Cui
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
| | - Kai-Li Jiang
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
| | - Kai-Qi He
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
| | - Yong-Ming Lu
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
| | - Yan Chen
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
- Key Laboratory for Ecological Engineering and Biotechnology of Anhui Province, Hefei, 230601, P. R. China
- Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601, P. R. China
| | - Wen-Juan Pan
- School of life Sciences, Anhui University, Hefei, 230601, P. R. China
| |
Collapse
|
164
|
Zhao D, Huang ZK, Liang Y, Li ZJ, Zhang XW, Li KH, Wu H, Zhang XD, Li CS, An D, Sun X, An MX, Shi JX, Bao YJ, Tian L, Wang DF, Wu AH, Chen YH, Zhao WD. Monocytes Release Pro-Cathepsin D to Drive Blood-to-Brain Transcytosis in Diabetes. Circ Res 2024; 134:e17-e33. [PMID: 38420756 DOI: 10.1161/circresaha.123.323622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Microvascular complications are the major outcome of type 2 diabetes progression, and the underlying mechanism remains to be determined. METHODS High-throughput RNA sequencing was performed using human monocyte samples from controls and diabetes. The transgenic mice expressing human CTSD (cathepsin D) in the monocytes was constructed using CD68 promoter. In vivo 2-photon imaging, behavioral tests, immunofluorescence, transmission electron microscopy, Western blot analysis, vascular leakage assay, and single-cell RNA sequencing were performed to clarify the phenotype and elucidate the molecular mechanism. RESULTS Monocytes expressed high-level CTSD in patients with type 2 diabetes. The transgenic mice expressing human CTSD in the monocytes showed increased brain microvascular permeability resembling the diabetic microvascular phenotype, accompanied by cognitive deficit. Mechanistically, the monocytes release nonenzymatic pro-CTSD to upregulate caveolin expression in brain endothelium triggering caveolae-mediated transcytosis, without affecting the paracellular route of brain microvasculature. The circulating pro-CTSD activated the caveolae-mediated transcytosis in brain endothelial cells via its binding with low-density LRP1 (lipoprotein receptor-related protein 1). Importantly, genetic ablation of CTSD in the monocytes exhibited a protective effect against the diabetes-enhanced brain microvascular transcytosis and the diabetes-induced cognitive impairment. CONCLUSIONS These findings uncover the novel role of circulatory pro-CTSD from monocytes in the pathogenesis of cerebral microvascular lesions in diabetes. The circulatory pro-CTSD is a potential target for the intervention of microvascular complications in diabetes.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Zeng-Kang Huang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yu Liang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Zhi-Jun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Kun-Hang Li
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Hao Wu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Xu-Dong Zhang
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Chen-Sheng Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Dong An
- School of Mechanical Engineering, Shenyang Jianzhu University, China (D.A.)
| | - Xue Sun
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Ming-Xin An
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Jun-Xiu Shi
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Yi-Jun Bao
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang, China (D.Z., K.-H.L., X.-D.Z., Y.-J.B.)
| | - Li Tian
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Di-Fei Wang
- Department of Gerontology (L.T., D.-F.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - An-Hua Wu
- Department of Neurosurgery (A.-H.W.), Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China (D.Z., Z.-K.H., Y.L., Z.-J.L., X.-W.Z., H.W., C.-S.L., X.S., M.-X.A., J.-X.S., Y.-H.C., W.-D.Z.)
| |
Collapse
|
165
|
Eyraud N, Bloch S, Brizard B, Pena L, Tharsis A, Surget A, El-Hage W, Belzung C. Influence of Stress Severity on Contextual Fear Extinction and Avoidance in a Posttraumatic-like Mouse Model. Brain Sci 2024; 14:311. [PMID: 38671963 PMCID: PMC11048507 DOI: 10.3390/brainsci14040311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Posttraumatic stress disorder (PTSD) is a widespread fear-related psychiatric affection associated with fear extinction impairments and important avoidance behaviors. Trauma-related exposure therapy is the current first-hand treatment for PTSD, yet it needs to be improved to shorten the time necessary to reach remission and increase responsiveness. Additional studies to decipher the neurobiological bases of extinction and effects on PTSD-like symptoms could therefore be of use. However, a PTSD-like animal model exhibiting pronounced PTSD-related phenotypes even after an extinction training directly linked to the fearful event is necessary. Thus, using a contextual fear conditioning model of PTSD, we increased the severity of stress during conditioning to search for effects on extinction acquisition and on pre- and post-extinction behaviors. During conditioning, mice received either two or four electrical shocks while a control group was constituted of mice only exposed to the context. Stressed mice exhibited important fear generalization, high fear reaction to the context and selective avoidance of a contextual reminder even after the extinction protocol. Increasing the number of footshocks did not induce major changes on these behaviors.
Collapse
Affiliation(s)
- Noémie Eyraud
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| | - Solal Bloch
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| | - Bruno Brizard
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| | - Laurane Pena
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| | - Antoine Tharsis
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| | - Alexandre Surget
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| | - Wissam El-Hage
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
- Pôle de Psychiatrie et d’Addictologie, Centre Hospitalier Régional Universitaire de Tours, 37000 Tours, France
| | - Catherine Belzung
- Institut National de la Santé et de la Recherche Médicale (INSERM), Imaging Brain & Neuropsychiatry iBraiN U1253, Université de Tours, 37032 Tours, France
| |
Collapse
|
166
|
Li X, Wu X, Lu T, Kuang C, Si Y, Zheng W, Li Z, Xue Y. Perineuronal Nets in the CNS: Architects of Memory and Potential Therapeutic Target in Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3412. [PMID: 38542386 PMCID: PMC10970535 DOI: 10.3390/ijms25063412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
The extracellular matrix (ECM) within the brain possesses a distinctive composition and functionality, influencing a spectrum of physiological and pathological states. Among its constituents, perineuronal nets (PNNs) are unique ECM structures that wrap around the cell body of many neurons and extend along their dendrites within the central nervous system (CNS). PNNs are pivotal regulators of plasticity in CNS, both during development and adulthood stages. Characterized by their condensed glycosaminoglycan-rich structures and heterogeneous molecular composition, PNNs not only offer neuroprotection but also participate in signal transduction, orchestrating neuronal activity and plasticity. Interfering with the PNNs in adult animals induces the reactivation of critical period plasticity, permitting modifications in neuronal connections and promoting the recovery of neuroplasticity following spinal cord damage. Interestingly, in the adult brain, PNN expression is dynamic, potentially modulating plasticity-associated states. Given their multifaceted roles, PNNs have emerged as regulators in the domains of learning, memory, addiction behaviors, and other neuropsychiatric disorders. In this review, we aimed to address how PNNs contribute to the memory processes in physiological and pathological conditions.
Collapse
Affiliation(s)
- Xue Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xianwen Wu
- Department of Laboratory Animal Sciences, Peking University Health Sciences Center, Beijing 100191, China;
| | - Tangsheng Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chenyan Kuang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China;
| | - Yue Si
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zheng
- Peking-Tsinghua Centre for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
| | - Zhonghao Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yanxue Xue
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
| |
Collapse
|
167
|
Li Y, Zhang S, Tang C, Yang B, Atrooz F, Ren Z, Mohan C, Salim S, Wu T. Autoimmune and neuropsychiatric phenotypes in a Mecp2 transgenic mouse model on C57BL/6 background. Front Immunol 2024; 15:1370254. [PMID: 38524134 PMCID: PMC10960363 DOI: 10.3389/fimmu.2024.1370254] [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: 01/14/2024] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Systemic Lupus Erythematosus (SLE) impacts the central nervous system (CNS), leading to severe neurological and psychiatric manifestations known as neuropsychiatric lupus (NPSLE). The complexity and heterogeneity of clinical presentations of NPSLE impede direct investigation of disease etiology in patients. The limitations of existing mouse models developed for NPSLE obstruct a comprehensive understanding of this disease. Hence, the identification of a robust mouse model of NPSLE is desirable. Methods C57BL/6 mice transgenic for human MeCP2 (B6.Mecp2Tg1) were phenotyped, including autoantibody profiling through antigen array, analysis of cellularity and activation of splenic immune cells through flow cytometry, and measurement of proteinuria. Behavioral tests were conducted to explore their neuropsychiatric functions. Immunofluorescence analyses were used to reveal altered neurogenesis and brain inflammation. Various signaling molecules implicated in lupus pathogenesis were examined using western blotting. Results B6.Mecp2Tg1 exhibits elevated proteinuria and an overall increase in autoantibodies, particularly in female B6.Mecp2Tg1 mice. An increase in CD3+CD4+ T cells in the transgenic mice was observed, along with activated germinal center cells and activated CD11b+F4/80+ macrophages. Moreover, the transgenic mice displayed reduced locomotor activity, heightened anxiety and depression, and impaired short-term memory. Immunofluorescence analysis revealed IgG deposition and immune cell infiltration in the kidneys and brains of transgenic mice, as well as altered neurogenesis, activated microglia, and compromised blood-brain barrier (BBB). Additionally, protein levels of various key signaling molecules were found to be differentially modulated upon MeCP2 overexpression, including GFAP, BDNF, Albumin, NCoR1, mTOR, and NLRP3. Discussion Collectively, this work demonstrates that B6.Mecp2Tg1 mice exhibit lupus-like phenotypes as well as robust CNS dysfunctions, suggesting its utility as a new animal model for NPSLE.
Collapse
Affiliation(s)
- Yaxi Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Shu Zhang
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Bowen Yang
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Zhifeng Ren
- Department of Physics, University of Houston, Houston, TX, United States
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
168
|
Bakoyiannis I, Ducourneau EG, N'diaye M, Fermigier A, Ducroix-Crepy C, Bosch-Bouju C, Coutureau E, Trifilieff P, Ferreira G. Obesogenic diet induces circuit-specific memory deficits in mice. eLife 2024; 13:e80388. [PMID: 38436653 PMCID: PMC10911750 DOI: 10.7554/elife.80388] [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: 05/18/2022] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Obesity is associated with neurocognitive dysfunction, including memory deficits. This is particularly worrisome when obesity occurs during adolescence, a maturational period for brain structures critical for cognition. In rodent models, we recently reported that memory impairments induced by obesogenic high-fat diet (HFD) intake during the periadolescent period can be reversed by chemogenetic manipulation of the ventral hippocampus (vHPC). Here, we used an intersectional viral approach in HFD-fed male mice to chemogenetically inactivate specific vHPC efferent pathways to nucleus accumbens (NAc) or medial prefrontal cortex (mPFC) during memory tasks. We first demonstrated that HFD enhanced activation of both pathways after training and that our chemogenetic approach was effective in normalizing this activation. Inactivation of the vHPC-NAc pathway rescued HFD-induced deficits in recognition but not location memory. Conversely, inactivation of the vHPC-mPFC pathway restored location but not recognition memory impairments produced by HFD. Either pathway manipulation did not affect exploration or anxiety-like behaviour. These findings suggest that HFD intake throughout adolescence impairs different types of memory through overactivation of specific hippocampal efferent pathways and that targeting these overactive pathways has therapeutic potential.
Collapse
Affiliation(s)
- Ioannis Bakoyiannis
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Eva Gunnel Ducourneau
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Mateo N'diaye
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Alice Fermigier
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Celine Ducroix-Crepy
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Clementine Bosch-Bouju
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | | | - Pierre Trifilieff
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Guillaume Ferreira
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| |
Collapse
|
169
|
Davila-Valencia I, Saad M, Olthoff G, Faulkner M, Charara M, Farnum A, Dysko RC, Zhang Z. Sex specific effects of buprenorphine on adult hippocampal neurogenesis and behavioral outcomes during the acute phase after pediatric traumatic brain injury in mice. Neuropharmacology 2024; 245:109829. [PMID: 38159797 DOI: 10.1016/j.neuropharm.2023.109829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Traumatic brain injury (TBI) in children often causes cognitive and mental dysfunctions, as well as acute and chronic pain. Adult hippocampal neurogenesis plays a key role in cognition, depression, and pain. Adult hippocampal neurogenesis can be modulated by genetic and environmental factors, such as TBI and opioids. Buprenorphine (BPN), a semisynthetic opioid, is commonly used for pain management in children, however, the effects of BPN on adult hippocampal neurogenesis after pediatric TBI are still unclear. This study investigated the sex-specific effects of BPN on adult hippocampal neurogenesis during acute phase after pediatric TBI. Male and female littermates were randomized on postnatal day 20-21(P20-21) into Sham, TBI+saline and TBI+BPN groups. BPN was administered intraperitoneally to the TBI+BPN mice at 30 min after injury, and then every 6-12 h (h) for 2 days (d). Bromodeoxyuridine (BrdU) was administered intraperitoneally to all groups at 2, 4, 6, and 8-h post-injury. All outcomes were evaluated at 3-d post-BrdU administration. We found that TBI induced significant cognitive impairment, depression, and reduced adult hippocampal neurogenesis in both male and female mice, with more prominent effects in females. BPN significantly improved adult hippocampal neurogenesis and depression in males, but not in females. We further demonstrated that differential expressions of opioid receptors, transcription factors and neuroinflammatory markers at the neurogenic niche might be responsible for the differential effects of BPN in males and females. In conclusion, this study elucidates the effects of BPN on adult hippocampal neurogenesis and behavioral outcomes at the acute phase after pediatric TBI.
Collapse
Affiliation(s)
- Ivan Davila-Valencia
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Mark Saad
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Grace Olthoff
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Megan Faulkner
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Maysoun Charara
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Abigail Farnum
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Robert C Dysko
- Unit for Laboratory Animal Medicine, University of Michigan-Ann Arbor, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| |
Collapse
|
170
|
Aljuraysi S, Platt M, Pulix M, Poptani H, Plagge A. Microcephaly with a disproportionate hippocampal reduction, stem cell loss and neuronal lipid droplet symptoms in Trappc9 KO mice. Neurobiol Dis 2024; 192:106431. [PMID: 38331351 DOI: 10.1016/j.nbd.2024.106431] [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: 12/04/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024] Open
Abstract
Mutations of the human TRAFFICKING PROTEIN PARTICLE COMPLEX SUBUNIT 9 (TRAPPC9) cause a neurodevelopmental disorder characterised by microcephaly and intellectual disability. Trappc9 constitutes a subunit specific to the intracellular membrane-associated TrappII complex. The TrappII complex interacts with Rab11 and Rab18, the latter being specifically associated with lipid droplets (LDs). Here we used non-invasive imaging to characterise Trappc9 knock-out (KO) mice as a model of the human hereditary disorder. KOs developed postnatal microcephaly with many grey and white matter regions being affected. In vivo magnetic resonance imaging (MRI) identified a disproportionately stronger volume reduction in the hippocampus, which was associated with a significant loss of Sox2-positive neural stem and progenitor cells. Diffusion tensor imaging indicated a reduced organisation or integrity of white matter areas. Trappc9 KOs displayed behavioural abnormalities in several tests related to exploration, learning and memory. Trappc9-deficient primary hippocampal neurons accumulated a larger LD volume per cell following Oleic Acid stimulation, and the coating of LDs by Perilipin-2 was much reduced. Additionally, Trappc9 KOs developed obesity, which was significantly more severe in females than in males. Our findings indicate that, beyond previously reported Rab11-related vesicle transport defects, dysfunctions in LD homeostasis might contribute to the neurobiological symptoms of Trappc9 deficiency.
Collapse
Affiliation(s)
- Sultan Aljuraysi
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mark Platt
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Michela Pulix
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK.
| | - Antonius Plagge
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK.
| |
Collapse
|
171
|
Valvaikar S, Vaidya B, Sharma S, Bishnoi M, Kondepudi KK, Sharma SS. Supplementation of probiotic Bifidobacterium breve Bif11 reverses neurobehavioural deficits, inflammatory changes and oxidative stress in Parkinson's disease model. Neurochem Int 2024; 174:105691. [PMID: 38311217 DOI: 10.1016/j.neuint.2024.105691] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Human gut microbiota are thought to affect different physiological processes in the body, including brain functions. Gut dysbiosis has been linked to the progression of Parkinson's disease (PD) and thus, restoring the healthy gut microbiota with supplementation of putative probiotic strains can confer some benefits in PD. In the current study, we explored the neuroprotective potential of Bifidobacterium breve Bif11 supplementation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) treated female Sprague Dawley rats. This study investigated the behavioural, molecular and biochemical parameters in the MPTP rat model. A pharmacological intervention of Bif11 at doses of 1 × 1010 CFU and 2 × 1010 CFU for 21 days was found to attenuate the cognitive and motor changes in the MPTP rat model. Furthermore, it also increased the tyrosine hydroxylase levels, reduced pro-inflammatory markers and decreased oxidative and nitrosative stress in the mid brain of MPTP-lesioned rats. Bif11 supplementation even restored the levels of short-chain fatty acids and decreased intestinal epithelial permeability in MPTP-induced PD model rats. In summary, these findings demonstrate that B. breve Bif11 has the potential to ameliorate symptoms of PD. However, this therapy needs to be further investigated with in-depth mechanistic insights in the future for the treatment of PD.
Collapse
Affiliation(s)
- Sonali Valvaikar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| | - Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| | - Shikha Sharma
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India.
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
172
|
Zhang YN, Chen XL, Guo LY, Jiang PR, Lu H, Pan K, Guo L, Hu YT, Bao AM. Downregulation of peripheral luteinizing hormone rescues ovariectomy-associated cognitive deficits in APP/PS1 mice. Neurobiol Aging 2024; 135:60-69. [PMID: 38185053 DOI: 10.1016/j.neurobiolaging.2023.12.007] [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: 05/04/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 01/09/2024]
Abstract
Alzheimer's disease (AD) is more prevalent in women than men, supposing due to the decline of estrogens in menopause, accompanied by increased gonadotropins such as luteinizing hormone (LH). We and others found that the transcription factor early growth response-1 (EGR1) regulates cholinergic function including the expression of acetylcholinesterase (AChE) and plays a significant role in cognitive decline of AD. Here we investigated in APP/PS1 mice by ovariectomy (OVX) and estradiol (E2) supplementation or inhibition of LH the effect on hippocampus-related cognition and related molecular changes. We found that OVX-associated cognitive impairment was accompanied by increased dorsal hippocampal EGR1 expression, which was rescued by downregulating peripheral LH rather than by supplementing E2. We also found in postmortem AD brains a higher expression of pituitary LH-mRNA and higher EGR1 expression in the posterior hippocampus. Both, in human and mice, there was a significant positive correlation between respectively posterior/dorsal hippocampal EGR1 and peripheral LH expression. We conclude that peripheral increased LH and increased posterior hippocampal EGR1 plays a significant role in AD pathology.
Collapse
Affiliation(s)
- Ya-Nan Zhang
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xin-Lu Chen
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China; Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, East Qingchun Road 3#, Hangzhou, Zhejiang 310016, China
| | - Ling-Yu Guo
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Pei-Ran Jiang
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Brain Bank for Health and Disease, Hangzhou, China
| | - Hui Lu
- National Brain Bank for Health and Disease, Hangzhou, China
| | - Kai Pan
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Lei Guo
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yu-Ting Hu
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China; Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, East Qingchun Road 3#, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
173
|
Liu W, Jia M, Zhang K, Chen J, Zhu X, Li R, Xu Z, Zang Y, Wang Y, Pan J, Ma D, Yang J, Wang D. Increased A1 astrocyte activation-driven hippocampal neural network abnormality mediates delirium-like behavior in aged mice undergoing cardiac surgery. Aging Cell 2024; 23:e14074. [PMID: 38155547 PMCID: PMC10928578 DOI: 10.1111/acel.14074] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023] Open
Abstract
Delirium is the most common neurological complication after cardiac surgery with adverse impacts on surgical outcomes. Advanced age is an independent risk factor for delirium occurrence but its underlying mechanisms are not fully understood. Although increased A1 astrocytes and abnormal hippocampal networks are involved in neurodegenerative diseases, whether A1 astrocytes and hippocampal network changes are involved in the delirium-like behavior of aged mice remains unknown. In the present study, a mice model of myocardial ischemia-reperfusion mimicking cardiac surgery and various assessments were used to investigate the different susceptibility of the occurrence of delirium-like behavior between young and aged mice and the underlying mechanisms. The results showed that surgery significantly increased hippocampal A1 astrocyte activation in aged compared to young mice. The high neuroinflammatory state induced by surgery resulted in glutamate accumulation in the extrasynaptic space, which subsequently decreased the excitability of pyramidal neurons and increased the PV interneurons inhibition through enhancing N-methyl-D-aspartate receptors' tonic currents in the hippocampus. These further induced the abnormal activities of the hippocampal neural networks and consequently contributed to delirium-like behavior in aged mice. Notably, the intraperitoneal administration of exendin-4, a glucagon-like peptide-1 receptor agonist, downregulated A1 astrocyte activation and alleviated delirium-like behavior in aged mice, while IL-1α, TNF-α, and C1q in combination administered intracerebroventricularly upregulated A1 astrocyte activation and induced delirium-like behavior in young mice. Therefore, our study suggested that cardiac surgery increased A1 astrocyte activation which subsequently impaired the hippocampal neural networks and triggered delirium development.
Collapse
Affiliation(s)
- Wenxue Liu
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Min Jia
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Keyin Zhang
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jiang Chen
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical SchoolNanjing UniversityNanjingChina
| | - Xiyu Zhu
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Ruisha Li
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Zhenjun Xu
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yanyu Zang
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research CenterNanjing UniversityNanjingChina
| | - Yapeng Wang
- Department of Cardio‐Thoracic Surgery, Nanjing Drum Tower HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeNanjingChina
| | - Jun Pan
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of MedicineImperial College London, Chelsea and Westminster HospitalLondonUK
- Perioperative and Systems Medicine Laboratory, Children’s Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dongjin Wang
- Department of Cardio‐Thoracic Surgery, Institute of Cardiothoracic Vascular Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
174
|
Benítez-Castañeda A, Anaya-Martínez V, Espadas-Alvarez ADJ, Gutierrez-Váldez AL, Razgado-Hernández LF, Reyna-Velazquez PE, Quintero-Macias L, Martínez-Fong D, Florán-Garduño B, Aceves J. Transfection of the BDNF Gene in the Surviving Dopamine Neurons in Conjunction with Continuous Administration of Pramipexole Restores Normal Motor Behavior in a Bilateral Rat Model of Parkinson's Disease. PARKINSON'S DISEASE 2024; 2024:3885451. [PMID: 38419644 PMCID: PMC10901579 DOI: 10.1155/2024/3885451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
In Parkinson's disease (PD), progressive degeneration of nigrostriatal innervation leads to atrophy and loss of dendritic spines of striatal medium spiny neurons (MSNs). The loss disrupts corticostriatal transmission, impairs motor behavior, and produces nonmotor symptoms. Nigral neurons express brain-derived neurotropic factor (BDNF) and dopamine D3 receptors, both protecting the dopamine neurons and the spines of MSNs. To restore motor and nonmotor symptoms to normality, we assessed a combined therapy in a bilateral rat Parkinson's model, with only 30% of surviving neurons. The preferential D3 agonist pramipexole (PPX) was infused for four ½ months via mini-osmotic pumps and one month after PPX initiation; the BDNF-gene was transfected into the surviving nigral cells using the nonviral transfection NTS-polyplex vector. Overexpression of the BDNF-gene associated with continuous PPX infusion restored motor coordination, balance, normal gait, and working memory. Recovery was also related to the restoration of the average number of dendritic spines of the striatal projection neurons and the number of TH-positive neurons of the substantia nigra and ventral tegmental area. These positive results could pave the way for further clinical research into this promising therapy.
Collapse
Affiliation(s)
- Alina Benítez-Castañeda
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | | | | | | | | | | | - Liz Quintero-Macias
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Daniel Martínez-Fong
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Benjamín Florán-Garduño
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Jorge Aceves
- Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| |
Collapse
|
175
|
Gouveia F, Fonseca C, Silva A, Camins A, Teresa Cruz M, Ettcheto M, Fortuna A. Intranasal irbesartan reverts cognitive decline and activates the PI3K/AKT pathway in an LPS-induced neuroinflammation mice model. Int Immunopharmacol 2024; 128:111471. [PMID: 38199198 DOI: 10.1016/j.intimp.2023.111471] [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: 09/26/2023] [Revised: 12/13/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND New strategies are urgently needed to manage and delay the development of Alzheimer's disease (AD). Neuroinflammation is a significant contributor to cognitive decline in neurodegenerative diseases, including AD. Angiotensin receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs) protect hypertensive patients against AD, but the cellular and molecular mechanisms underlying these effects remain unknown. In light of this, the protective effects of three ARBs and three ACEIs against neuroinflammation and cognitive decline were investigated through comprehensive pharmacologicalin vitro/in vivoscreening. METHODS BV-2 microglia cells were exposed tolipopolysaccharide (LPS) and treated with ARBs and ACEIs to provide initial insights into the anti-inflammatory properties of the drugs. Subsequently, irbesartan was selected, and its efficacy was evaluated inC57/BL6 male miceintranasally administered with irbesartan and injected with LPS. Long-term memory and depressive-like behavior were evaluated; dendritic spines were measured as well as neuroinflammation, neurodegeneration and cognitive decline biomarkers. RESULTS Irbesartan mitigated memory loss and depressive-like behavior in mice treated with LPS, probably because itincreased spine density, ameliorated synapsis dysfunction and activated the PI3K/AKT pathway. Irbesartan elevated the levels of hippocampalsuperoxide dismutase2 andglutathione peroxidaseandsuppressed LPS-induced astrogliosis. CONCLUSIONS Overall, this study provides compelling evidence that multiple intranasal administrations of irbesartan can effectively prevent LPS-induced cognitive decline by activating pathways involved in neuroprotection and anti-inflammatory events. These findings underscore the potential of irbesartan as a preventive strategy against the development of AD and other neurodegenerative conditions associated with neuroinflammation.
Collapse
Affiliation(s)
- Filipa Gouveia
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Carla Fonseca
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal; Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Ana Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - M Teresa Cruz
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
176
|
Althobaiti YS. Oral self-administration of pregabalin in a mouse model and the resulting drug addiction features. Saudi Pharm J 2024; 32:101935. [PMID: 38292403 PMCID: PMC10825552 DOI: 10.1016/j.jsps.2023.101935] [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: 11/16/2023] [Accepted: 12/21/2023] [Indexed: 02/01/2024] Open
Abstract
Prescription drug abuse is an issue that is rapidly growing globally. Pregabalin, an anticonvulsant, analgesic, and anxiolytic medication, is effective in the management of multiple neurological disorders; however, there is increasing concern regarding its widespread illicit use. It has been previously reported in mice that pregabalin can induce conditioned place preference. In this current investigation, the potential of pregabalin to elicit free-choice drinking in a mouse model of drug addiction, and its effect on recognition and withdrawal behaviors after forced abstinence, were studied. Twenty-two male BALB/c mice were randomly divided into three groups (n = 7-8/group); control, pregabalin-30, and pregabalin-60. The study had three phases: habituation (days 1-5) with free water access, free-choice drinking (days 6-13) with pregabalin groups receiving one water and one pregabalin bottle, and forced abstinence (days 14-21) with free water access. On day 13, the first open field test was conducted, followed by the Novel Object Recognition Test. On day 21, the second open field test was performed, followed by the Tail Suspension Test and Forced Swimming Test. Pregabalin elicited voluntary drinking in the higher-dose group, concurrently causing a decline in recognition memory performance in the novel object recognition test. Moreover, pregabalin induced withdrawal behavior after a period of forced abstinence in the forced swimming and tail suspension tests. This is the first report to establish an animal model of free-choice pregabalin drinking that may be used for further molecular studies and targeted therapy for pregabalin addiction.
Collapse
Affiliation(s)
- Yusuf S. Althobaiti
- Address: Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| |
Collapse
|
177
|
Mucellini AB, Laureano DP, Alves MB, Dalle Molle R, Borges MB, Salvador APDA, Pokhvisneva I, Manfro GG, Silveira PP. The impact of poor fetal growth and chronic hyperpalatable diet exposure in adulthood on hippocampal function and feeding patterns in male rats. Dev Psychobiol 2024; 66:e22459. [PMID: 38372503 DOI: 10.1002/dev.22459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 02/20/2024]
Abstract
Poor fetal growth affects eating behavior and the mesocorticolimbic system; however, its influence on the hippocampus has been less explored. Brain insulin sensitivity has been linked to developmental plasticity in response to fetal adversity and to cognitive performance following high-fat diet intake. We investigated whether poor fetal growth and exposure to chronic hyperpalatable food in adulthood could influence the recognition of environmental and food cues, eating behavior patterns, and hippocampal insulin signaling. At 60 days of life, we assigned male offspring from a prenatal animal model of 50% food restriction (FR) to receive either a high-fat and -sugar (HFS) diet or standard chow (CON) diet. Behavioral tests were conducted at 140 days, then tissues were collected. HFS groups showed a diminished hippocampal pAkt/Akt ratio. FR-CON and FR-HFS groups had higher levels of suppressor of cytokine signaling 3, compared to control groups. FR groups showed increased exploration of a novel hyperpalatable food, independent of their diet, and HFS groups exhibited overall lower entropy (less random, more predictable eating behavior) when the environment changed. Poor fetal growth and chronic HFS diet in adulthood altered hippocampal insulin signaling and eating patterns, diminishing the flexibility associated with eating behavior in response to extrinsic changes in food availability in the environment.
Collapse
Affiliation(s)
- Amanda Brondani Mucellini
- Graduate Program in Psychiatry and Behavioral Sciences, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daniela Pereira Laureano
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Child and Adolescent Health, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcio Bonesso Alves
- Graduate Program in Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Roberta Dalle Molle
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Mariana Balbinot Borges
- Faculty of Biomedicine, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Irina Pokhvisneva
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
| | - Gisele Gus Manfro
- Graduate Program in Psychiatry and Behavioral Sciences, Faculty of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patrícia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
178
|
Gubert C, Kong G, Costello C, Adams CD, Masson BA, Qin W, Choo J, Narayana VK, Rogers G, Renoir T, Furness JB, Hannan AJ. Dietary fibre confers therapeutic effects in a preclinical model of Huntington's disease. Brain Behav Immun 2024; 116:404-418. [PMID: 38142919 DOI: 10.1016/j.bbi.2023.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder involving psychiatric, cognitive and motor deficits, as well as peripheral symptoms, including gastrointestinal dysfunction. The R6/1 HD mouse model expresses a mutant human huntingtin transgene and has been shown to provide an accurate disease model. Recent evidence of gut microbiome disruption was shown in preclinical and clinical HD. Therefore, we aimed to assess the potential role of gut microbial modulation in the treatment of HD. The R6/1 HD mice and wild-type littermate controls were randomised to receive diets containing different amounts of fibre: high-fibre (10 % fibre), control (5 % fibre), or zero-fibre (0 % fibre), from 6 to 20 weeks of age. We characterized the onset and progression of motor, cognitive and affective deficits, as well as gastrointestinal function and gut morphological changes. Faeces were collected for gut microbiome profiling using 16S rRNA sequencing, at 14 and 20 weeks of age. When compared to the control diet, high-fibre diet improved the performance of HD mice in behavioral tests of cognitive and affective function, as well as the gastrointestinal function of both HD and wild-type mice. While the diets changed the beta diversity of wild-type mice, no statistical significance was observed at 14 or 20 weeks of age within the HD mice. Analysis of Composition of Microbiomes with Bias Correction (ANCOM-BC) models were performed to evaluate microbiota composition, which identified differences, including a decreased relative abundance of the phyla Actinobacteriota, Campylobacterota and Proteobacteria and an increased relative abundance of the families Bacteroidaceae, Oscillospiraceae and Ruminococcaceae in HD mice when compared to wild-type mice after receiving high-fibre diet. PICRUSt2 revealed that high-fibre diet also decreased potentially pathogenic functional pathways in HD. In conclusion, high-fibre intake was effective in enhancing gastrointestinal function, cognition and affective behaviors in HD mice. These findings indicate that dietary fibre interventions may have therapeutic potential in Huntington's disease to delay clinical onset, and have implications for related disorders exhibiting dysfunction of the gut-brain axis.
Collapse
Affiliation(s)
- Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Geraldine Kong
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Peter Doherty Institute of Infection and Immunity, University of Melbourne, Parkville, Victoria 3000, Australia
| | - Callum Costello
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Cameron D Adams
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jocelyn Choo
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Vinod K Narayana
- Metabolomics Australia Bio21 Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Geraint Rogers
- Microbiome and Host Health, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Infection and Immunity, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville 3010, Australia
| | - John B Furness
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville 3010, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
179
|
Wu W, Luo Z, Shen D, Lan T, Xiao Z, Liu M, Hu L, Sun T, Wang Y, Zhang JN, Zhang C, Wang P, Lu Y, Yang F, Li Q. IL-10 protects against OPC ferroptosis by regulating lipid reactive oxygen species levels post stroke. Redox Biol 2024; 69:102982. [PMID: 38070317 PMCID: PMC10755589 DOI: 10.1016/j.redox.2023.102982] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024] Open
Abstract
Accumulation of reactive oxygen species (ROS), especially on lipids, induces massive cell death in neurons and oligodendrocyte progenitor cells (OPCs) and causes severe neurologic deficits post stroke. While small compounds, such as deferoxamine, lipostatin-1, and ferrostatin-1, have been shown to be effective in reducing lipid ROS, the mechanisms by which endogenously protective molecules act against lipid ROS accumulation and subsequent cell death are still unclear, especially in OPCs, which are critical for maintaining white matter integrity and improving long-term outcomes after stroke. Here, using mouse primary OPC cultures, we demonstrate that interleukin-10 (IL-10), a cytokine playing roles in reducing neuroinflammation and promoting hematoma clearance, significantly reduced hemorrhage-induced lipid ROS accumulation and subsequent ferroptosis in OPCs. Mechanistically, IL-10 activated the IL-10R/STAT3 signaling pathway and upregulated the DLK1/AMPK/ACC axis. Subsequently, IL-10 reprogrammed lipid metabolism and reduced lipid ROS accumulation. In addition, in an autologous blood injection intracerebral hemorrhagic stroke (ICH) mouse model, deficiency of the endogenous Il-10, specific knocking out Il10r or Dlk1 in OPCs, or administration of ACC inhibitor was associated with increased OPC cell death, demyelination, axonal sprouting, and the cognitive deficits during the chronic phase of ICH and vice versa. These data suggest that IL-10 protects against OPC loss and white matter injury by reducing lipid ROS, supporting further development of potential clinical applications to benefit patients with stroke and related disorders.
Collapse
Affiliation(s)
- Weihua Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zhaoli Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Danmin Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ting Lan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zhongnan Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Meng Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Liye Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Tingting Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yamei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Chenguang Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Peipei Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yabin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
180
|
Philibert CE, Disdier C, Lafon PA, Bouyssou A, Oosterlaken M, Galant S, Pizzoccaro A, Tuduri P, Ster J, Liu J, Kniazeff J, Pin JP, Rondard P, Marin P, Vandermoere F. TrkB receptor interacts with mGlu 2 receptor and mediates antipsychotic-like effects of mGlu 2 receptor activation in the mouse. SCIENCE ADVANCES 2024; 10:eadg1679. [PMID: 38277461 PMCID: PMC10816717 DOI: 10.1126/sciadv.adg1679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 12/28/2023] [Indexed: 01/28/2024]
Abstract
Metabotropic glutamate receptor 2 (mGlu2) attracts particular attention as a possible target for a new class of antipsychotics. However, the signaling pathways transducing the effects of mGlu2 in the brain remain poorly characterized. Here, we addressed this issue by identifying native mGlu2 interactome in mouse prefrontal cortex. Nanobody-based affinity purification and mass spectrometry identified 149 candidate mGlu2 partners, including the neurotrophin receptor TrkB. The later interaction was confirmed both in cultured cells and prefrontal cortex. mGlu2 activation triggers phosphorylation of TrkB on Tyr816 in primary cortical neurons and prefrontal cortex. Reciprocally, TrkB stimulation enhances mGlu2-operated Gi/o protein activation. Furthermore, TrkB inhibition prevents the rescue of behavioral deficits by glutamatergic antipsychotics in phencyclidine-treated mice. Collectively, these results reveal a cross-talk between TrkB and mGlu2, which is key to the behavioral response to glutamatergic antipsychotics.
Collapse
Affiliation(s)
- Clémentine Eva Philibert
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Candice Disdier
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pierre-André Lafon
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of MOE, International Research Centre for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Alexandre Bouyssou
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Mathieu Oosterlaken
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Sonya Galant
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Pizzoccaro
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pola Tuduri
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Jeanne Ster
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Jianfeng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of MOE, International Research Centre for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Julie Kniazeff
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Franck Vandermoere
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
181
|
Kadyan S, Park G, Hochuli N, Miller K, Wang B, Nagpal R. Resistant starches from dietary pulses improve neurocognitive health via gut-microbiome-brain axis in aged mice. Front Nutr 2024; 11:1322201. [PMID: 38352704 PMCID: PMC10864001 DOI: 10.3389/fnut.2024.1322201] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Cognitive decline is a common consequence of aging. Dietary patterns that lack fibers and are high in saturated fats worsen cognitive impairment by triggering pro-inflammatory pathways and metabolic dysfunctions. Emerging evidence highlights the neurocognitive benefits of fiber-rich diets and the crucial role of gut-microbiome-brain signaling. However, the mechanisms of this diet-microbiome-brain regulation remain largely unclear. Methods Accordingly, we herein investigated the unexplored neuroprotective mechanisms of dietary pulses-derived resistant starch (RS) in improving aging-associated neurocognitive function in an aged (60-weeks old) murine model carrying a human microbiome. Results and discussion Following 20-weeks dietary regimen which included a western-style diet without (control; CTL) or with 5% w/w fortification with RS from pinto beans (PTB), black-eyed-peas (BEP), lentils (LEN), chickpeas (CKP), or inulin fiber (INU), we find that RS, particularly from LEN, ameliorate the cognitive impairments induced by western diet. Mechanistically, RS-mediated improvements in neurocognitive assessments are attributed to positive remodeling of the gut microbiome-metabolome arrays, which include increased short-chain fatty acids and reduced branched-chain amino acids levels. This microbiome-metabolite-brain signaling cascade represses neuroinflammation, cellular senescence, and serum leptin/insulin levels, while enhancing lipid metabolism through improved hepatic function. Altogether, the data demonstrate the prebiotic effects of RS in improving neurocognitive function via modulating the gut-brain axis.
Collapse
Affiliation(s)
- Saurabh Kadyan
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Gwoncheol Park
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Nathaniel Hochuli
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Katelyn Miller
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Bo Wang
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| | - Ravinder Nagpal
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
182
|
Iwai T, Mishima R, Hirayama S, Nakajima H, Oyama M, Watanabe S, Fujii H, Tanabe M. SYK-623, a δ Opioid Receptor Inverse Agonist, Mitigates Chronic Stress-Induced Behavioral Abnormalities and Disrupted Neurogenesis. J Clin Med 2024; 13:608. [PMID: 38276114 PMCID: PMC10817044 DOI: 10.3390/jcm13020608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The δ opioid receptor (DOR) inverse agonist has been demonstrated to improve learning and memory impairment in mice subjected to restraint stress. Here, we investigated the effects of SYK-623, a new DOR inverse agonist, on behavioral, immunohistochemical, and biochemical abnormalities in a mouse model of imipramine treatment-resistant depression. Male ddY mice received daily treatment of adrenocorticotropic hormone (ACTH) combined with chronic mild stress exposure (ACMS). SYK-623, imipramine, or the vehicle was administered once daily before ACMS. After three weeks, ACMS mice showed impaired learning and memory in the Y-maze test and increased immobility time in the forced swim test. SYK-623, but not imipramine, significantly suppressed behavioral abnormalities caused by ACMS. Based on the fluorescent immunohistochemical analysis of the hippocampus, ACMS induced a reduction in astrocytes and newborn neurons, similar to the reported findings observed in the postmortem brains of depressed patients. In addition, the number of parvalbumin-positive GABA neurons, which play a crucial role in neurogenesis, was reduced in the hippocampus, and western blot analysis showed decreased glutamic acid decarboxylase protein levels. These changes, except for the decrease in astrocytes, were suppressed by SYK-623. Thus, SYK-623 mitigates behavioral abnormalities and disturbed neurogenesis caused by chronic stress.
Collapse
Affiliation(s)
- Takashi Iwai
- Laboratory of Pharmacology, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (T.I.); (R.M.); (H.N.); (M.O.); (S.W.)
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
| | - Rei Mishima
- Laboratory of Pharmacology, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (T.I.); (R.M.); (H.N.); (M.O.); (S.W.)
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
| | - Shigeto Hirayama
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Honoka Nakajima
- Laboratory of Pharmacology, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (T.I.); (R.M.); (H.N.); (M.O.); (S.W.)
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
| | - Misa Oyama
- Laboratory of Pharmacology, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (T.I.); (R.M.); (H.N.); (M.O.); (S.W.)
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
| | - Shun Watanabe
- Laboratory of Pharmacology, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (T.I.); (R.M.); (H.N.); (M.O.); (S.W.)
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
| | - Hideaki Fujii
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
- Laboratory of Medicinal Chemistry, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Mitsuo Tanabe
- Laboratory of Pharmacology, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (T.I.); (R.M.); (H.N.); (M.O.); (S.W.)
- Medicinal Research Laboratories, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan; (S.H.); (H.F.)
| |
Collapse
|
183
|
Seo JY, Jo HR, Lee SH, Kim DG, Lee H, Kim YL, Choi YI, Jung SJ, Son H. TRPC4 deletion elicits behavioral defects in sociability by dysregulating expression of microRNA-138-2. iScience 2024; 27:108617. [PMID: 38188509 PMCID: PMC10770719 DOI: 10.1016/j.isci.2023.108617] [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: 03/07/2023] [Revised: 10/08/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
To investigate whether the defects in transient receptor potential canonical 4 (TRPC4), which is strongly expressed in the hippocampus, are implicated in ASD, we examined the social behaviors of mice in which Trpc4 was deleted (Trpc4-/-). Trpc4-/- mice displayed the core symptoms of ASD, namely, social disability and repetitive behaviors. In microarray analysis of the hippocampus, microRNA (miR)-138-2, the precursor of miR-138, was upregulated in Trpc4-/- mice. We also found that binding of Matrin3 (MATR3), a selective miR-138-2 binding nuclear protein, to miR-138-2 was prominently enhanced, resulting in the downregulation of miR-138 in Trpc4-/- mice. Some parameters of the social defects and repetitive behaviors in the Trpc4-/- mice were rescued by increased miR-138 levels following miR-138-2 infusion in the hippocampus. Together, these results suggest that Trpc4 regulates some signaling components that oppose the development of social behavioral deficits through miR-138 and provide a potential therapeutic strategy for ASD.
Collapse
Affiliation(s)
- Jee Young Seo
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Hye-Ryeong Jo
- Hanyang University Hospital for Rheumatic Diseases, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Seung Hoon Lee
- Hanyang University Hospital for Rheumatic Diseases, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Huiju Lee
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Ye Lim Kim
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Young In Choi
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Sung Jun Jung
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| |
Collapse
|
184
|
Rusin D, Vahl Becirovic L, Lyszczarz G, Krueger M, Benmamar-Badel A, Vad Mathiesen C, Sigurðardóttir Schiöth E, Lykke Lambertsen K, Wlodarczyk A. Microglia-Derived Insulin-like Growth Factor 1 Is Critical for Neurodevelopment. Cells 2024; 13:184. [PMID: 38247874 PMCID: PMC10813844 DOI: 10.3390/cells13020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a peptide hormone essential for the proper development and growth of the organism, as a complete knockout of Igf1 in mice is lethal, causing microcephaly, growth retardation and the defective development of organs. In the central nervous system, neurons and glia have been reported to express Igf1, but their relative importance for postnatal development has not yet been fully defined. In order to address this, here, we obtained mice with a microglia-specific inducible conditional knockout of Igf1. We show that the deficiency in microglial Igf1, starting in the first postnatal week, leads to body and brain growth retardation, severely impaired myelination, changes in microglia numbers, and behavioral abnormalities. These results emphasize the importance of microglial-derived Igf1 for brain development and function and open new perspectives for the investigation of the role of microglial-Igf1 in neurological diseases.
Collapse
Affiliation(s)
- Dominika Rusin
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Lejla Vahl Becirovic
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Gabriela Lyszczarz
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Martin Krueger
- Institute for Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Cecilie Vad Mathiesen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Neuroscience Academy Denmark, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Eydís Sigurðardóttir Schiöth
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Clinical Research, BRIDGE—Brain Research Interdisciplinary Guided Excellence, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Clinical Research, BRIDGE—Brain Research Interdisciplinary Guided Excellence, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
185
|
Reddy DS, Singh T, Ramakrishnan S, Huber M, Wu X. Neuroprotectant Activity of Novel Water-Soluble Synthetic Neurosteroids on Organophosphate Intoxication and Status Epilepticus-Induced Long-Term Neurological Dysfunction, Neurodegeneration, and Neuroinflammation. J Pharmacol Exp Ther 2024; 388:399-415. [PMID: 38071567 PMCID: PMC10801736 DOI: 10.1124/jpet.123.001819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/18/2023] [Indexed: 01/19/2024] Open
Abstract
Organophosphates (OPs) and nerve agents are potent neurotoxic compounds that cause seizures, status epilepticus (SE), brain injury, or death. There are persistent long-term neurologic and neurodegenerative effects that manifest months to years after the initial exposure. Current antidotes are ineffective in preventing these long-term neurobehavioral and neuropathological changes. Additionally, there are few effective neuroprotectants for mitigating the long-term effects of acute OP intoxication. We have pioneered neurosteroids as novel anticonvulsants and neuroprotectants for OP intoxication and seizures. In this study, we evaluated the efficacy of two novel synthetic, water-soluble neurosteroids, valaxanolone (VX) and lysaxanolone (LX), in combating the long-term behavioral and neuropathological impairments caused by acute OP intoxication and SE. Animals were exposed to the OP nerve agent surrogate diisopropylfluorophosphate (DFP) and were treated with VX or LX in addition to midazolam at 40 minutes postexposure. The extent of neurodegeneration, along with various behavioral and memory deficits, were assessed at 3 months postexposure. VX significantly reduced deficits of aggressive behavior, anxiety, memory, and depressive-like traits in control (DFP-exposed, midazolam-treated) animals; VX also significantly prevented the DFP-induced chronic loss of NeuN(+) principal neurons and PV(+) inhibitory neurons in the hippocampus and other regions. Additionally, VX-treated animals exhibited a reduced inflammatory response with decreased GFAP(+) astrogliosis and IBA1(+) microgliosis in the hippocampus, amygdala, and other regions. Similarly, LX showed significant improvement in behavioral and memory deficits, and reduced neurodegeneration and cellular neuroinflammation. Together, these results demonstrate the neuroprotectant effects of the novel synthetic neurosteroids in mitigating the long-term neurologic dysfunction and neurodegeneration associated with OP exposure. SIGNIFICANCE STATEMENT: Survivors of nerve agents and organophosphate (OP) exposures suffer from long-term neurological deficits. Currently, there is no specific drug therapy for mitigating the impact of OP exposure. However, novel synthetic neurosteroids that activate tonic inhibition provide a viable option for treating OP intoxication. The data from this study indicates the neuroprotective effects of synthetic, water-soluble neurosteroids for attenuation of long-term neurological deficits after OP intoxication. These findings establish valaxanolone and lysaxanolone as potent and efficacious neuroprotectants suitable for injectable dosing.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Madeline Huber
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (D.S.R., T.S., S.R., M.H., X.W.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (D.S.R., S.R., X.W.)
| |
Collapse
|
186
|
López-Aranda MF, Bach K, Bui R, Phan M, Lu O, Thadani C, Luchetti A, Mandanas R, Herrera I, López-Ávalos MD, Silva AJ. Early Post-Natal Immune Activation Leads to Object Memory Deficits in Female Tsc2+/- Mice: The Importance of Including Both Sexes in Neuroscience Research. Biomedicines 2024; 12:203. [PMID: 38255309 PMCID: PMC10813674 DOI: 10.3390/biomedicines12010203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
There is evidence that viral infections during pre-natal development constitute a risk factor for neuropsychiatric disorders and lead to learning and memory deficits. However, little is known about why viral infections during early post-natal development have a different impact on learning and memory depending on the sex of the subject. We previously showed that early post-natal immune activation induces hippocampal-dependent social memory deficits in a male, but not in a female, mouse model of tuberous sclerosis complex (TSC; Tsc2+/- mice). Here, we explored the impact of a viral-like immune challenge in object memory. We demonstrate that early post-natal immune activation (during the first 2 weeks of life) leads to object memory deficits in female, but not male, mice that are heterozygous for a gene responsible for tuberous sclerosis complex (Tsc2+/- mice), while no effect was observed in wild type (WT) mice. Moreover, we found that the same immune activation in Tsc2+/- adult mice was not able to cause object memory deficits in females, which suggests that the early post-natal development stage constitutes a critical window for the effects of immune challenge on adult memory. Also, our results suggest that mTOR plays a critical role in the observed deficit in object memory in female Tsc2+/- mice. These results, together with previous results published by our laboratory, showing sex-specific memory deficits due to early post-natal immune activation, reinforce the necessity of using both males and females for research studies. This is especially true for studies related to immune activation, since the higher levels of estrogens in females are known to affect inflammation and to provide neuroprotection.
Collapse
Affiliation(s)
- Manuel F. López-Aranda
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, 29590 Málaga, Spain
| | - Karen Bach
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Raymond Bui
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Miranda Phan
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Odilia Lu
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Chirag Thadani
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Alessandro Luchetti
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Rochelle Mandanas
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Isaiah Herrera
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - María Dolores López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, 29590 Málaga, Spain
| | - Alcino J. Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| |
Collapse
|
187
|
Lai TT, Tsai YH, Liou CW, Fan CH, Hou YT, Yao TH, Chuang HL, Wu WL. The gut microbiota modulate locomotion via vagus-dependent glucagon-like peptide-1 signaling. NPJ Biofilms Microbiomes 2024; 10:2. [PMID: 38228675 DOI: 10.1038/s41522-024-00477-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
Locomotor activity is an innate behavior that can be triggered by gut-motivated conditions, such as appetite and metabolic condition. Various nutrient-sensing receptors distributed in the vagal terminal in the gut are crucial for signal transduction from the gut to the brain. The levels of gut hormones are closely associated with the colonization status of the gut microbiota, suggesting a complicated interaction among gut bacteria, gut hormones, and the brain. However, the detailed mechanism underlying gut microbiota-mediated endocrine signaling in the modulation of locomotion is still unclear. Herein, we show that broad-spectrum antibiotic cocktail (ABX)-treated mice displayed hypolocomotion and elevated levels of the gut hormone glucagon-like peptide-1 (GLP-1). Blockade of the GLP-1 receptor and subdiaphragmatic vagal transmission rescued the deficient locomotor phenotype in ABX-treated mice. Activation of the GLP-1 receptor and vagal projecting brain regions led to hypolocomotion. Finally, selective antibiotic treatment dramatically increased serum GLP-1 levels and decreased locomotion. Colonizing Lactobacillus reuteri and Bacteroides thetaiotaomicron in microbiota-deficient mice suppressed GLP-1 levels and restored the hypolocomotor phenotype. Our findings identify a mechanism by which specific gut microbes mediate host motor behavior via the enteroendocrine and vagal-dependent neural pathways.
Collapse
Affiliation(s)
- Tzu-Ting Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Hsuan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Chia-Wei Liou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Tian Hou
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Tzu-Hsuan Yao
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115202, Taiwan
| | - Wei-Li Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
188
|
Ferreira FM, Gomes SV, Carvalho LCF, de Alcantara AC, da Cruz Castro ML, Perucci LO, Pio S, Talvani A, de Abreu Vieira PM, Calsavara AJC, Costa DC. Potential of piperine for neuroprotection in sepsis-associated encephalopathy. Life Sci 2024; 337:122353. [PMID: 38104862 DOI: 10.1016/j.lfs.2023.122353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
AIMS Sepsis-associated encephalopathy (SAE) is a common complication that increases mortality and leads to long-term cognitive impairment in sepsis survivors. However, no specific or effective therapy has been identified for this complication. Piperine is an alkaloid known for its anti-inflammatory, antioxidant, and neuroprotective properties, which are important characteristics for treatment of SAE. The objective of this study was to evaluate the neuroprotective effect of piperine on SAE in C57BL/6 mice that underwent cecum ligation and perforation surgery (CLP). MAIN METHODS C57BL/6 male mice were randomly assigned to groups that underwent SHAM surgery or CLP. Mice in the CLP group were treated with piperine at doses of 20 or 40 mg/kg for short- (5 days) or long-term (10 days) periods after CLP. KEY FINDINGS Our results revealed that untreated septic animals exhibited increased concentrations of IL-6, TNF, VEGF, MMP-9, TBARS, and NLRP3, and decreased levels of BDNF, sulfhydryl groups, and catalase in the short term. Additionally, the levels of carbonylated proteins and degenerated neuronal cells were increased at both time points. Furthermore, short-term and visuospatial memories were impaired. Piperine treatment reduced MMP-9 activity in the short term and decreased the levels of carbonylated proteins and degenerated neuronal cells in the long term. It also lowered IL-6 and TBARS levels at both time points evaluated. Moreover, piperine increased short-term catalase and long-term BDNF factor levels and improved memory at both time points. SIGNIFICANCE In conclusion, our data demonstrate that piperine exerts a neuroprotective effect on SAE in animals that have undergone CLP.
Collapse
Affiliation(s)
- Flavia Monteiro Ferreira
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Sttefany Viana Gomes
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Luana Cristina Faria Carvalho
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Ana Carolina de Alcantara
- Laboratory of Cognition and Health (LACOS), School of Medicine, Department of Pediatric and Adult Clinics (DECPA), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Maria Laura da Cruz Castro
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Luiza Oliveira Perucci
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Sirlaine Pio
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences (DECBI), Graduate Program in Health and Nutrition, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Paula Melo de Abreu Vieira
- Morphopathology Laboratory, Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Allan Jefferson Cruz Calsavara
- Laboratory of Cognition and Health (LACOS), School of Medicine, Department of Pediatric and Adult Clinics (DECPA), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry (LBM), Department of Biological Sciences (DECBI), Graduate Program in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
189
|
Zhan S, Qi Z, Cai F, Gao Z, Xie J, Hu J. Oxytocin neurons mediate stress-induced social memory impairment. Curr Biol 2024; 34:36-45.e4. [PMID: 38103551 DOI: 10.1016/j.cub.2023.11.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023]
Abstract
Oxytocin has long been thought to play a substantial role in social behaviors, such as social attachment and parenting behavior. However, how oxytocin neurons respond to social and non-social stimuli is largely unknown, especially in high temporal resolution. Here, we recorded the in vivo real-time responses of oxytocin neurons in the paraventricular nucleus of the hypothalamus (PVN) in freely behaving mice. Our results revealed that oxytocin neurons were activated more significantly by stressors than social stimuli. The activation of oxytocin neurons was precisely correlated with struggling behavior during stress. Furthermore, we found that oxytocin mediated stress-induced social memory impairment. Our results reveal an important role of PVN oxytocin neurons in stress-induced social amnesia.
Collapse
Affiliation(s)
- Shulu Zhan
- School of Life Science and Technology, ShanghaiTech University, 393 Huaxia Middle Road, Shanghai 201210, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Institute of Neuroscience, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenhua Qi
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Fang Cai
- School of Life Science and Technology, ShanghaiTech University, 393 Huaxia Middle Road, Shanghai 201210, China
| | - Zilong Gao
- Chinese Institute for Brain Research, Beijing (CIBR), Bldg. 3, No. 9, YIKE Rd, Zhongguancun Life Science Park, Changping District, Beijing 102206, China.
| | - Jingdun Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, 393 Huaxia Middle Road, Shanghai 201210, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
190
|
Zhang M, Zhang Z, Li H, Xia Y, Xing M, Xiao C, Cai W, Bu L, Li Y, Park TE, Tang Y, Ye X, Lin WJ. Blockage of VEGF function by bevacizumab alleviates early-stage cerebrovascular dysfunction and improves cognitive function in a mouse model of Alzheimer's disease. Transl Neurodegener 2024; 13:1. [PMID: 38173017 PMCID: PMC10763201 DOI: 10.1186/s40035-023-00388-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder and the predominant type of dementia worldwide. It is characterized by the progressive and irreversible decline of cognitive functions. In addition to the pathological beta-amyloid (Aβ) deposition, glial activation, and neuronal injury in the postmortem brains of AD patients, increasing evidence suggests that the often overlooked vascular dysfunction is an important early event in AD pathophysiology. Vascular endothelial growth factor (VEGF) plays a critical role in regulating physiological functions and pathological changes in blood vessels, but whether VEGF is involved in the early stage of vascular pathology in AD remains unclear. METHODS We used an antiangiogenic agent for clinical cancer treatment, the humanized monoclonal anti-VEGF antibody bevacizumab, to block VEGF binding to its receptors in the 5×FAD mouse model at an early age. After treatment, memory performance was evaluated by a novel object recognition test, and cerebral vascular permeability and perfusion were examined by an Evans blue assay and blood flow scanning imaging analysis. Immunofluorescence staining was used to measure glial activation and Aβ deposits. VEGF and its receptors were analyzed by enzyme-linked immunosorbent assay and immunoblotting. RNA sequencing was performed to elucidate bevacizumab-associated transcriptional signatures in the hippocampus of 5×FAD mice. RESULTS Bevacizumab treatment administered from 4 months of age dramatically improved cerebrovascular functions, reduced glial activation, and restored long-term memory in both sexes of 5×FAD mice. Notably, a sex-specific change in different VEGF receptors was identified in the cortex and hippocampus of 5×FAD mice. Soluble VEGFR1 was decreased in female mice, while full-length VEGFR2 was increased in male mice. Bevacizumab treatment reversed the altered expression of receptors to be comparable to the level in the wild-type mice. Gene Set Enrichment Analysis of transcriptomic changes revealed that bevacizumab effectively reversed the changes in the gene sets associated with blood-brain barrier integrity and vascular smooth muscle contraction in 5×FAD mice. CONCLUSIONS Our study demonstrated the mechanistic roles of VEGF at the early stage of amyloidopathy and the protective effects of bevacizumab on cerebrovascular function and memory performance in 5×FAD mice. These findings also suggest the therapeutic potential of bevacizumab for the early intervention of AD.
Collapse
Affiliation(s)
- Min Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhan Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Honghong Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuting Xia
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Mengdan Xing
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Chuan Xiao
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Wenbao Cai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Lulu Bu
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yi Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yamei Tang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
191
|
Utyro O, Włoczkowska-Łapińska O, Jakubowski H. Association of GLOD4 with Alzheimer's Disease in Humans and Mice. J Alzheimers Dis 2024; 101:823-834. [PMID: 39302370 PMCID: PMC11492116 DOI: 10.3233/jad-240512] [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] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
Background Glyoxalase domain containing protein 4 (GLOD4), a protein of an unknown function, is associated with Alzheimer's disease (AD). Three GLOD4 isoforms are known. The mechanism underlying GLOD4's association with AD was unknown. Objective To assess GLOD4's role in the central nervous system by studying GLOD4 isoforms expression in human frontal cerebral cortical tissues from AD patients and in brains of Blmh-/-5xFAD mouse AD model of AD. Methods GLOD4 protein and mRNA were quantified in human and mouse brains by western blotting and RT-qPCR, respectively. Mouse brain amyloid-β (Aβ) was quantified by western blotting. Behavioral assessments of mice were performed by cognitive/neuromotor testing. Glod4 gene in mouse neuroblastoma N2a-APPswe cells was silenced by RNA interference and Glod4, Aβ precursor protein (Aβpp), Atg5, p62, and Lc3 proteins and mRNAs were quantified. Results GLOD4 mRNA and protein isoforms were downregulated in cortical tissues from AD patients compared to non-AD controls. Glod4 mRNA was downregulated in brains of Blmh-/-5xFAD mice compared to Blmh+/+5xFAD sibling controls, but not in Blmh-/- mice without the 5xFAD transgene compared to Blmh+/+ sibling controls. The 5xFAD transgene downregulated Glod4 mRNA in Blmh-/- mice of both sexes and in Blmh+/+ males but not females. Attenuated Glod4 was associated with elevated Aβ and worsened memory/sensorimotor performance in Blmh-/-5xFAD mice. Glod4 depletion in N2a-APPswe cells upregulated AβPP, and downregulated autophagy-related Atg5, p62, and Lc3 genes. Conclusions These findings suggest that GLOD4 interacts with AβPP and the autophagy pathway, and that disruption of these interactions leads to Aβ accumulation and cognitive/neurosensory deficits.
Collapse
Affiliation(s)
- Olga Utyro
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | | | - Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
- Department of Microbiology, Biochemistry &Molecular Genetics, Rutgers-New Jersey Medical School, International Center for Public Health, Newark, NJ, USA
| |
Collapse
|
192
|
Jiang LX, Huang GD, Wang HL, Zhang C, Yu X. The olfactory working memory capacity paradigm: A more sensitive and robust method of assessing cognitive function in male 5XFAD mice. J Neurosci Res 2024; 102:e25265. [PMID: 38284863 DOI: 10.1002/jnr.25265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 06/29/2023] [Accepted: 10/07/2023] [Indexed: 01/30/2024]
Abstract
The olfactory working memory capacity (OWMC) paradigm is able to detect cognitive deficits in 5XFAD mice (an animal model of Alzheimer's disease [TG]) as early as 3 months of age, while other behavioral paradigms detect cognitive deficits only at 4-5 months of age. Therefore, we aimed to demonstrate that the OWMC paradigm is more sensitive and consistent in the early detection of declines in cognitive function than other commonly used behavioral paradigms. The prefrontal cortex (PFC), retrosplenial cortex (RSC), subiculum (SUB), and amygdala (AMY) of 5XFAD mice were harvested and subjected to immunostaining to detect the expression of β-amyloid (Aβ). Additionally, we compared the performance of 3-month-old male 5XFAD mice on common behavioral paradigms for assessing cognitive function (i.e., the open field [OF] test, novel object recognition [NOR] test, novel object location [NOL] test, Y-maze, and Morris water maze [MWM]) with that on the OWMC task. In the testing phase of the OWMC task, we varied the delay periods to evaluate the working memory capacity (WMC) of wild-type (WT) mice. Significant amyloid plaque deposition was observed in the PFC, RSC, SUB, and AMY of 3-month-old male 5XFAD mice. However, aside from the OWMC task, the other behavioral tests failed to detect cognitive deficits in 5XFAD mice. Additionally, to demonstrate the efficacy of the OWMC task in assessing WMC, we varied the retention delay periods; we found that the WMC of WT mice decreased with longer delay periods. The OWMC task is a sensitive and robust behavioral assay for detecting changes in cognitive function.
Collapse
Affiliation(s)
- Li-Xin Jiang
- Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, China
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, China
| | - Geng-Di Huang
- Department of Addiction Medicine, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Mental Health Center, Shenzhen Kangning Hospital, Shenzhen, China
- Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, China
| | - Hua-Li Wang
- Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, China
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Xin Yu
- Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University), Beijing, China
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, China
| |
Collapse
|
193
|
Pan S, Zhang Y, Ye T, Kong Y, Cui X, Yuan S, Liu J, Zhang Y. A High-Tryptophan Diet Alleviated Cognitive Impairment and Neuroinflammation in APP/PS1 Mice through Activating Aryl Hydrocarbon Receptor via the Regulation of Gut Microbiota. Mol Nutr Food Res 2024; 68:e2300601. [PMID: 38031265 DOI: 10.1002/mnfr.202300601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/13/2023] [Indexed: 12/01/2023]
Abstract
SCOPE Recent studies have highlighted the vital role of gut microbiota in the pathogenesis of Alzheimer's disease (AD). However, the effect of the regulation of gut microbiota by dietary components on AD remains unknown. Thus, the study explored that a high-tryptophan (Trp) diet alleviates cognitive impairment by regulating microbiota. METHODS AND RESULTS Male APP/PS1 mice are fed 0.5% Trp diet for 4 weeks, and then cognitive function, amyloid-β (Aβ) deposition, microglial activation, proinflammatory cytokines production, and gut microbiota are detected. Moreover, the level of aryl hydrocarbon receptor (AhR) and NF-κB pathway related protein are determined. The results show that high-Trp diet significantly alleviates cognitive impairment and Aβ deposits. Moreover, high-Trp diet significantly inhibits activation of microglia, decreases the level of cluster of differentiation 11b (CD11b), and restrains the activation markers of microglia, such as cyclooxygenase-2 (Cox-2), interleukin (IL)-1β, and IL-6. Notably, high-Trp diet significantly activates AhR, inhibits the phosphorylation of p65, and improves microbiota dysbiosis. CONCLUSIONS These findings demonstrated that high-Trp diet exerts anti-inflammatory effects via upregulating AhR and suppressing NF-κB pathway, and its mechanisms may be mediated by regulating gut microbiota, suggesting that Trp diet may be a potential strategy for AD intervention.
Collapse
Affiliation(s)
- Sipei Pan
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yuhe Zhang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tao Ye
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yu Kong
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaorui Cui
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shushu Yuan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yang Zhang
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| |
Collapse
|
194
|
Fuchigami T, Itokazu Y, Yu RK. Ganglioside GD3 regulates neural stem cell quiescence and controls postnatal neurogenesis. Glia 2024; 72:167-183. [PMID: 37667994 PMCID: PMC10840680 DOI: 10.1002/glia.24468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/14/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
The postnatal neural stem cell (NSC) pool hosts quiescent and activated radial glia-like NSCs contributing to neurogenesis throughout adulthood. However, the underlying regulatory mechanism during the transition from quiescent NSCs to activated NSCs in the postnatal NSC niche is not fully understood. Lipid metabolism and lipid composition play important roles in regulating NSC fate determination. Biological lipid membranes define the individual cellular shape and help maintain cellular organization and are highly heterogeneous in structure and there exist diverse microdomains (also known as lipid rafts), which are enriched with sugar molecules, such as glycosphingolipids. An often overlooked but key aspect is that the functional activities of proteins and genes are highly dependent on their molecular environments. We previously reported that ganglioside GD3 is the predominant species in NSCs and that the reduced postnatal NSC pools are observed in global GD3-synthase knockout (GD3S-KO) mouse brains. The specific roles of GD3 in determining the stage and cell-lineage determination of NSCs remain unclear, since global GD3S-KO mice cannot distinguish if GD3 regulates postnatal neurogenesis or developmental impacts. Here, we show that inducible GD3 deletion in postnatal radial glia-like NSCs promotes NSC activation, resulting in the loss of the long-term maintenance of the adult NSC pools. The reduced neurogenesis in the subventricular zone (SVZ) and the dentate gyrus (DG) of GD3S-conditional-knockout mice led to the impaired olfactory and memory functions. Thus, our results provide convincing evidence that postnatal GD3 maintains the quiescent state of radial glia-like NSCs in the adult NSC niche.
Collapse
Affiliation(s)
- Takahiro Fuchigami
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yutaka Itokazu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Robert K. Yu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
195
|
Pollock NM, Fernandes JP, Woodfield J, Moussa E, Hlavay B, Branton WG, Wuest M, Mohammadzadeh N, Schmitt L, Plemel JR, Julien O, Wuest F, Power C. Gasdermin D activation in oligodendrocytes and microglia drives inflammatory demyelination in progressive multiple sclerosis. Brain Behav Immun 2024; 115:374-393. [PMID: 37914099 DOI: 10.1016/j.bbi.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023] Open
Abstract
Neuroinflammation coupled with demyelination and neuro-axonal damage in the central nervous system (CNS) contribute to disease advancement in progressive multiple sclerosis (P-MS). Inflammasome activation accompanied by proteolytic cleavage of gasdermin D (GSDMD) results in cellular hyperactivation and lytic death. Using multiple experimental platforms, we investigated the actions of GSDMD within the CNS and its contributions to P-MS. Brain tissues from persons with P-MS showed significantly increased expression of GSDMD, NINJ1, IL-1β, and -18 within chronic active demyelinating lesions compared to MS normal appearing white matter and nonMS (control) white matter. Conditioned media (CM) from stimulated GSDMD+/+ human macrophages caused significantly greater cytotoxicity of oligodendroglial and neuronal cells, compared to CM from GSDMD-/- macrophages. Oligodendrocytes and CNS macrophages displayed increased Gsdmd immunoreactivity in the central corpus callosum (CCC) of cuprizone (CPZ)-exposed Gsdmd+/+ mice, associated with greater demyelination and reduced oligodendrocyte precursor cell proliferation, compared to CPZ-exposed Gsdmd-/- animals. CPZ-exposed Gsdmd+/+ mice exhibited significantly increased G-ratios and reduced axonal densities in the CCC compared to CPZ-exposed Gsdmd-/- mice. Proteomic analyses revealed increased brain complement C1q proteins and hexokinases in CPZ-exposed Gsdmd-/- animals. [18F]FDG PET imaging showed increased glucose metabolism in the hippocampus and whole brain with intact neurobehavioral performance in Gsdmd-/- animals after CPZ exposure. GSDMD activation in CNS macrophages and oligodendrocytes contributes to inflammatory demyelination and neuroaxonal injury, offering mechanistic and potential therapeutic insights into P-MS pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Laura Schmitt
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton AB, Canada.
| | | | | | | | - Christopher Power
- Department of Medicine (Neurology), Canada; Department of Medical Microbiology & Immunology, Canada.
| |
Collapse
|
196
|
Urati A, Angati A, Singh Gautam A, Dey M, Pandey SK, Singh RK. Neuroprotective responses of quercetin in regulation of biochemical, structural, and neurobehavioral effects in 28-day oral exposure of iron in rats. Toxicol Mech Methods 2024; 34:57-71. [PMID: 37680063 DOI: 10.1080/15376516.2023.2256840] [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: 07/19/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Iron is one of the essential metals that functions as a cofactor in various biological cascades in the brain. However, excessive iron accumulation in the brain may lead to neurodegeneration and may show toxic effects. Quercetin, a pigment flavonoid compound, has been proven to be a potent antioxidant and anti-inflammatory that can inhibit lipid peroxidation during metal-induced neurotoxicity. Although iron-induced neuroinflammation and neurodegeneration have been reported in many studies, but the proof for its exact mechanisms needs to be explored. PURPOSE The key target of the study was to explore the neuroprotective effect of quercetin after oral exposure of iron in rats and explore its underlying molecular mechanisms. RESULTS The outcomes of the study have shown that oral exposure to ferrous sulfate may modulate behavioral paradigms such as locomotor activity, neuromuscular coordination, and increased anxiety level. The pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), apoptotic protein (caspase 3), beta-amyloid and phosphorylated tau were found to be increased on iron exposure. Also, the expressions of ferritin heavy and light chain, BACE-1 and GFAP expressions were altered. These behavioral, structural, and biochemical alterations in the brain were significantly and dose-dependently reversed by treatment with quercetin. CONCLUSION The current study provides a fundamental understanding of molecular signaling pathways, and structural proteins implicated in iron-induced neurotoxicity along with the ameliorative effects of quercetin.
Collapse
Affiliation(s)
- Anuradha Urati
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Anok Angati
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Mangaldeep Dey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Shivam Kumar Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| |
Collapse
|
197
|
Gallegos CE, Gumilar F, Bartos M, Baier CJ. Long-term behavioral and neurochemical paradoxical alterations elicited following intranasal application of a chlorpyrifos formulation in mice. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 198:105717. [PMID: 38225064 DOI: 10.1016/j.pestbp.2023.105717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/17/2024]
Abstract
The intranasal (IN) administration route represents a pathway for xenobiotics to reach the brain. The present study aimed to address the long-term consequences of IN administration of a chlorpyrifos (CPF) commercial formulation (fCPF) in mice. For this purpose, adult male CF-1 mice were intranasally administered with fCPF (10 mg/kg/day) three days a week, for 2 and 4 weeks, respectively. Behavioral and biochemical analyses were conducted 3-7, and 7.5 months after the last IN fCPF administration, respectively. Following a 6-month fCPF-free washout period, fur appearance and body injuries scores improved in the fCPF-treated groups. Notably, spatial learning and memory enhancement was observed 4 and 7 months after the last IN fCPF administration. Changes in oxidative stress markers and the activities of enzymes involved in cholinergic and glutamatergic pathways were observed in different brain areas from fCPF-treated mice, still after 7.5 months from fCPF application. Altogether, these neurochemical disturbances could be responsible for the described behavioral observations.
Collapse
Affiliation(s)
- Cristina Eugenia Gallegos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Fernanda Gumilar
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Mariana Bartos
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina
| | - Carlos Javier Baier
- Laboratorio de Toxicología, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Departamento de Biología, Bioquímica y Farmacia (DBByF), San Juan 670, B8000ICN Bahía Blanca, Argentina.
| |
Collapse
|
198
|
Yu Z, Iyer L, Swiercz AP, Paronett E, Ramadan M, Marvar PJ, Posnack NG. The Impact of Chronic Phthalate Exposure on Rodent Anxiety and Cognition. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:203-212. [PMID: 38298799 PMCID: PMC10829632 DOI: 10.1016/j.bpsgos.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 02/02/2024] Open
Abstract
Background There is a growing importance for environmental contributions to psychiatric disorders and understanding the impact of the exposome (i.e., pollutants and toxins). For example, increased biomonitoring and epidemiological studies suggest that daily phthalate chemical exposure contributes to neurological and behavioral abnormalities; however, these mechanisms remain poorly understood. Therefore, the current study was aimed at examining the effects of chronic phthalate exposure on rodent anxiety behaviors and cognition and the impact on hypothalamic-pituitary-adrenal axis function. Methods Adult male mice (C57BL6/J) were administered MEHP via drinking water (1 mg/mL), and anxiety-like behavior and cognition combined with hypothalamic-pituitary-adrenal axis and inflammatory assays were assessed after 3 weeks of MEHP exposure. Results MEHP-treated mice exhibited enhanced generalized anxiety-like behaviors, as demonstrated by reduced time spent in the open-arm of the elevated plus maze and center exploration in the open field. Tests of spatial memory and cognition were unchanged. Following MEHP administration, circulating levels of corticosterone and proinflammatory cytokines were significantly increased, while at the tissue level, there were MEHP-dependent reductions in glucocorticoid metabolism genes Hsd11b1 and Hsd11b2. Conclusions These data suggest that chronic MEHP exposure leads to enhanced generalized anxiety behaviors independent of rodent measures of cognition and memory, which may be driven by MEHP-dependent effects on hypothalamic-pituitary-adrenal axis and peripheral glucocorticoid metabolism function.
Collapse
Affiliation(s)
- Zhe Yu
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Laxmi Iyer
- Department of Anatomy, Physiology and Genetics, Uniformed Services University Health Sciences, Bethesda, Maryland
| | - Adam P. Swiercz
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Paronett
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Manelle Ramadan
- Children’s National Heart Institute, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC
| | - Paul J. Marvar
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC
| | - Nikki Gillum Posnack
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
- Children’s National Heart Institute, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC
| |
Collapse
|
199
|
Wang JH, Wu C, Lian YN, Cao XW, Wang ZY, Dong JJ, Wu Q, Liu L, Sun L, Chen W, Chen WJ, Zhang Z, Zhuo M, Li XY. Single-cell RNA sequencing uncovers the cell type-dependent transcriptomic changes in the retrosplenial cortex after peripheral nerve injury. Cell Rep 2023; 42:113551. [PMID: 38048224 DOI: 10.1016/j.celrep.2023.113551] [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: 12/10/2021] [Revised: 05/14/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023] Open
Abstract
The retrosplenial cortex (RSC) is a vital area for storing remote memory and has recently been found to undergo broad changes after peripheral nerve injury. However, little is known about the role of RSC in pain regulation. Here, we examine the involvement of RSC in the pain of mice with nerve injury. Notably, reducing the activities of calcium-/calmodulin-dependent protein kinase type II-positive splenial neurons chemogenetically increases paw withdrawal threshold and extends thermal withdrawal latency in mice with nerve injury. The single-cell or single-nucleus RNA-sequencing results predict enhanced excitatory synaptic transmissions in RSC induced by nerve injury. Local infusion of 1-naphthyl acetyl spermine into RSC to decrease the excitatory synaptic transmissions relieves pain and induces conditioned place preference. Our data indicate that RSC is critical for regulating physiological and neuropathic pain. The cell type-dependent transcriptomic information would help understand the molecular basis of neuropathic pain.
Collapse
Affiliation(s)
- Jing-Hua Wang
- Department of Psychiatry of the Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Cheng Wu
- Department of Psychiatry of the Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH8 9JU, UK
| | - Yan-Na Lian
- Department of Psychiatry of the Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiao-Wen Cao
- Department of Psychiatry of the Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zi-Yue Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jia-Jun Dong
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Qin Wu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Li Liu
- Core Facilities of the School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Li Sun
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Wen-Juan Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Zhi Zhang
- Key Laboratory of Brain Functions and Diseases, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Min Zhuo
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiang-Yao Li
- Department of Psychiatry of the Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain, Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH8 9JU, UK.
| |
Collapse
|
200
|
Su J, Huang F, Tian Y, Tian R, Qianqian G, Bello ST, Zeng D, Jendrichovsky P, Lau CG, Xiong W, Yu D, Tortorella M, Chen X, He J. Entorhinohippocampal cholecystokinin modulates spatial learning by facilitating neuroplasticity of hippocampal CA3-CA1 synapses. Cell Rep 2023; 42:113467. [PMID: 37979171 DOI: 10.1016/j.celrep.2023.113467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/01/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
The hippocampus is broadly impacted by neuromodulations. However, how neuropeptides shape the function of the hippocampus and the related spatial learning and memory remains unclear. Here, we discover the crucial role of cholecystokinin (CCK) in heterosynaptic neuromodulation from the medial entorhinal cortex (MEC) to the hippocampus. Systematic knockout of the CCK gene impairs CA3-CA1 LTP and space-related performance. The MEC provides most of the CCK-positive neurons projecting to the hippocampal region, which potentiates CA3-CA1 long-term plasticity heterosynaptically in a frequency- and NMDA receptor (NMDAR)-dependent manner. Selective inhibition of MEC CCKergic neurons or downregulation of their CCK mRNA levels also impairs CA3-CA1 LTP formation and animals' performance in the water maze. This excitatory extrahippocampal projection releases CCK upon high-frequency excitation and is active during animal exploration. Our results reveal the critical role of entorhinal CCKergic projections in bridging intra- and extrahippocampal circuitry at electrophysiological and behavioral levels.
Collapse
Affiliation(s)
- Junfeng Su
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Fengwen Huang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China.
| | - Yu Tian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Ran Tian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Gao Qianqian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Stephen Temitayo Bello
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China
| | - Dingxaun Zeng
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Peter Jendrichovsky
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - C Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Wenjun Xiong
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China
| | - Daiguan Yu
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P.R. China
| | - Micky Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P.R. China
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China.
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China.
| |
Collapse
|