1
|
Pokharel PV, Newchurch AM, Overby SC, Spease CA, Darzi LG, Kraemer BR. LM11a-31 Inhibits p75 Neurotrophin Receptor (p75 NTR ) Cleavage and is Neuroprotective in a Cell Culture Model of Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612299. [PMID: 39314373 PMCID: PMC11419115 DOI: 10.1101/2024.09.10.612299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The p75 Neurotrophin Receptor (p75 NTR ) is a multifunctional transmembrane protein that mediates neuronal responses to pathological conditions in specific regions of the nervous system. In many biological contexts, p75 NTR signaling is initiated through sequential cleavage of the receptor by α- and γ-secretases, which releases receptor fragments for downstream signaling. Our previous work demonstrated that proteolytic processing of p75 NTR in this manner is stimulated by oxidative stress in Lund Human Mesencephalic (LUHMES) cells, a dopaminergic neuronal cell line derived from human mesencephalic tissue. Considering the vulnerability of dopaminergic neurons in the ventral mesencephalon to oxidative stress and neurodegeneration associated with Parkinson's disease (PD), we investigated the role of this signaling cascade in neurodegeneration and explored cellular processes that govern oxidative stress-induced p75 NTR signaling. In the present study, we provide evidence that oxidative stress induces cleavage of p75 NTR by promoting c-Jun N-terminal Kinase (JNK)-dependent internalization of p75 NTR from the cell surface. This activation of p75 NTR signaling is counteracted by tropomyosin-related kinase (Trk) receptor signaling; however, oxidative stress leads to Trk receptor downregulation, thereby enhancing p75 NTR processing. Importantly, we demonstrate that this pathway can be inhibited by LM11a-31, a small molecule modulator of p75 NTR , thereby conferring protection against neurodegeneration. Treatment with LM11a-31 significantly reduced p75 NTR cleavage and neuronal death associated with oxidative stress. These findings reveal novel mechanisms underlying activation of p75 NTR in response to oxidative stress, underscore a key role for p75 NTR in dopaminergic neurodegeneration, and highlight p75 NTR as a potential therapeutic target for reducing neurodegeneration in PD.
Collapse
|
2
|
Mei F, Zhao C, Li S, Xue Z, Zhao Y, Xu Y, Ye R, You H, Yu P, Han X, Carr GV, Weinberger DR, Yang F, Lu B. Ngfr + cholinergic projection from SI/nBM to mPFC selectively regulates temporal order recognition memory. Nat Commun 2024; 15:7342. [PMID: 39187496 PMCID: PMC11347598 DOI: 10.1038/s41467-024-51707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
Acetylcholine regulates various cognitive functions through broad cholinergic innervation. However, specific cholinergic subpopulations, circuits and molecular mechanisms underlying recognition memory remain largely unknown. Here we show that Ngfr+ cholinergic neurons in the substantia innominate (SI)/nucleus basalis of Meynert (nBM)-medial prefrontal cortex (mPFC) circuit selectively underlies recency judgements. Loss of nerve growth factor receptor (Ngfr-/- mice) reduced the excitability of cholinergic neurons in the SI/nBM-mPFC circuit but not in the medial septum (MS)-hippocampus pathway, and impaired temporal order memory but not novel object and object location recognition. Expression of Ngfr in Ngfr-/- SI/nBM restored defected temporal order memory. Fiber photometry revealed that acetylcholine release in mPFC not only predicted object encounters but also mediated recency judgments of objects, and such acetylcholine release was absent in Ngfr-/- mPFC. Chemogenetic and optogenetic inhibition of SI/nBM projection to mPFC in ChAT-Cre mice diminished mPFC acetylcholine release and deteriorated temporal order recognition. Impaired cholinergic activity led to a depolarizing shift of GABAergic inputs to mPFC pyramidal neurons, due to disturbed KCC2-mediated chloride gradients. Finally, potentiation of acetylcholine signaling upregulated KCC2 levels, restored GABAergic driving force and rescued temporal order recognition deficits in Ngfr-/- mice. Thus, NGFR-dependent SI/nBM-mPFC cholinergic circuit underlies temporal order recognition memory.
Collapse
Affiliation(s)
- Fan Mei
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Chen Zhao
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Shangjin Li
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Zeping Xue
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- School of Basic Medicine, Capital Medical University, Beijing, China
- Laboratory of Cognitive and Behavioral Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yueyang Zhao
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yihua Xu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Rongrong Ye
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - He You
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Peng Yu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Xinyu Han
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Gregory V Carr
- Department of Pharmacology and Molecular Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Department of Pharmacology and Molecular Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Feng Yang
- Basic and Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
- Laboratory of Cognitive and Behavioral Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Yang XP, Dan-Dai, Chen RX, Li YX, Rui Lv X, Li Y. The expression of ProBDNF and its high affinity receptor P75NTR in the neurons of emotion-related brain regions of post-stroke depression rats. Brain Res 2024; 1831:148829. [PMID: 38423239 DOI: 10.1016/j.brainres.2024.148829] [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: 12/21/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE To investigate the expression of the precursor of brain-derived neurotrophic factor (proBDNF) and its high-affinity receptor p75NTR in neurons of emotion-related brain areas (prefrontal cortex, hippocampus, and amygdala) in rats with post-stroke depression (PSD), and to explore the expression levels of proBDNF and p75NTR in neurons of emotion-related brain areas by injecting tissue plasminogen activator (t-PA) into the lateral ventricle of PSD rats, this significantly improved the stress-induced depression-like behavior,thus further validating the above results. METHODS Rats were randomly divided into four groups: a normal control group (n = 8), a depression group (n = 8), a stroke group (n = 8), and a PSD group (n = 8). The rat model of stroke was established by thread embolism, and the PSD animal model was induced by chronic unpredictable mild stress (CUMS) and solitary feeding. Behavioral tests were conducted, including weight measurement, open field tests, and sucrose preference tests. Immunofluorescence double labeling was used to detect the expression of proBDNF and p75NTR in neurons of emotion-related brain regions in the PSD rat model. Four weeks after CUMS treatment, the PSD group was selected. Rats were infused with t-PA (3 μg dissolved in 6 μL saline, Boehringer Ingelheim), proBDNF (3 μg dissolved in 6 μL saline, Abcam), or equal-volume NS once per day for 7 consecutive days using the syringe pump connecting to injection needles. After 7 days of continuous administration, animal behavior was assessed through scoring, and the expression of proBDNF and p75NTR in the emotion-related brain regions of the PSD rat model was detected using immunofluorescence double labeling. RESULTS Compared with the normal control group and the stroke group, the body weight, sucrose water consumption, and vertical movement distance in the PSD group were significantly lower (P < 0.05). In contrast, when compared with the proBDNF injection group and saline injection group, the weight, sucrose water consumption, field horizontal movement, and vertical movement distance of the t-PA injection group significantly increased after PSD lateral ventricle intubation.Double immunofluorescence revealed a higher neuronal expression of proBDNF as well as p75NTR in the prefrontal cortex and hippocampus of PSD rats compared to control animals (P < 0.05). In the amygdala, the expression levels of proBDNF and P75NTR were significantly reduced in the PSD group compared to the control group (P < 0.05). The results of the expression levels of proBDNF and P75NTR in the emotion-related brain regions of PSD rats injected with t-PA showed that proBDNF and P75NTR was significantly reduced in the prefrontal cortex, hippocampus, and amygdala of PSD rats compared to those of the NS and proBDNF groups (P < 0.05). CONCLUSIONS The increased expression of the brain-derived neurotrophic factor precursor proBDNF and its receptor p75NTR in neurons of emotion-related brain regions may play an important role in the pathogenesis of PSD.t-PA reduced the expression of proBDNF and its receptor p75NTR in neurons emotion-related brain regions and significantly improved the stress-induced depression-like behavior. Therefore, it is reasonable to assume that exogenous injection of t-PA may alleviate the depressive symptoms of PSD patients.Reducing the expression of proBDNF by injecting t-PA may provide a novel therapeutic approach for the treatment of stress-related mood disorders.
Collapse
Affiliation(s)
- Xue-Ping Yang
- Clinical Medical School, Dali University, Dali, China
| | - Dan-Dai
- Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, Jiangsu Province, China
| | - Ruo-Xia Chen
- Clinical Medical School, Dali University, Dali, China
| | - Yu-Xuan Li
- Clinical Medical School, Dali University, Dali, China
| | - Xue Rui Lv
- Clinical Medical School, Dali University, Dali, China
| | - Yun Li
- Clinical Medical School, Dali University, Dali, China; Changshu Hospital Affiliated to Soochow University, Changshu No.1 People's Hospital, Changshu 215500, Jiangsu Province, China.
| |
Collapse
|
4
|
Jo D, Ahn SY, Choi SY, Choi Y, Lee DH, Song J. Positive Effects of Adiponectin, BDNF, and GLP-1 on Cortical Neurons Counteracting Palmitic Acid Induced Neurotoxicity. Clin Nutr Res 2024; 13:121-129. [PMID: 38784850 PMCID: PMC11109930 DOI: 10.7762/cnr.2024.13.2.121] [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: 03/20/2024] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024] Open
Abstract
The prevalence of metabolic syndrome caused by diets containing excessive fatty acids is increasing worldwide. Patients with metabolic syndrome exhibit abnormal lipid profiles, chronic inflammation, increased levels of saturated fatty acids, impaired insulin sensitivity, excessive fat accumulation, and neuropathological issues such as memory deficits. In particular, palmitic acid (PA) in saturated fatty acids aggravates inflammation, insulin resistance, impaired glucose tolerance, and synaptic failure. Recently, adiponectin, brain-derived neurotrophic factor (BDNF), and glucose-like peptide-1 (GLP-1) have been investigated to find therapeutic solutions for metabolic syndrome, with findings suggesting that they are involved in insulin sensitivity, enhanced lipid profiles, increased neuronal survival, and improved synaptic plasticity. We investigated the effects of adiponectin, BDNF, and GLP-1 on neurite outgrowth, length, and complexity in PA-treated primary cortical neurons using Sholl analysis. Our findings demonstrate the therapeutic potential of adiponectin, BDNF, and GLP-1 in enhancing synaptic plasticity within brains affected by metabolic imbalance. We underscore the need for additional research into the mechanisms by which adiponectin, BDNF, and GLP-1 influence neural complexity in brains with metabolic imbalances.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Seo Yeon Ahn
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| | - Seo Yoon Choi
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| | - Yoonjoo Choi
- Department of MRC, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Hwasun 58128, Korea
| |
Collapse
|
5
|
Yang CR, Liang R, Liu Y, Meng FJ, Zhou F, Zhang XY, Ning L, Wang ZQ, Liu S, Zhou XF. Upregulation of proBDNF/p75NTR signaling in immune cells and its correlation with inflammatory markers in patients with major depression. FASEB J 2024; 38:e23312. [PMID: 38161282 DOI: 10.1096/fj.202301140rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 01/03/2024]
Abstract
ProBDNF is the precursor protein of brain-derived neurotrophic factor (BDNF) expressed in the central nervous system and peripheral tissues. Previous studies showed that the blood levels of both proBDNF and p75 neurotrophic receptors (p75NTR) in major depressive disorder (MDD) were increased, but which blood cell types express proBDNF and its receptors is not known. Furthermore, the relationship between proBDNF/p75NTR and inflammatory cytokines in peripheral blood of MDD is unclear. Peripheral blood mononuclear cells (PBMCs) and serum were obtained from depressive patients (n = 32) and normal donors (n = 20). We examined the expression of proBDNF and inflammatory markers and their correlative relationship in patients with major depression. Using flow cytometry analysis, we examined which blood cells express proBDNF and its receptors. Finally, the role of proBDNF/p75NTR signal in inflammatory immune activity of PBMCs was verified in vitro experiments. Inflammatory cytokines in PBMC from MDD patients were increased and correlated with the major depression scores. The levels of IL-1β and IL-10 were also positively correlated with the major depression scores, while the levels of TNF-α and IL-6 were negatively correlated with the major depression scores. Intriguingly, the levels of sortilin were positively correlated with IL-1β. Q-PCR and Western blots showed proBDNF, p75NTR, and sortilin levels were significantly increased in PBMCs from MDD patients compared with that from the normal donors. Flow cytometry studies showed that proBDNF and p75NTR were present mainly in CD4+ and CD8+ T cells. The number of proBDNF and p75NTR positive CD4+ and CD8+ T cells from MDD patients was increased and subsequently reversed after therapeutic management. Exogenous proBDNF protein or p75ECD-Fc treatment of cultured PBMC affected the release of inflammatory cytokines in vitro. ProBDNF promoted the expression of inflammatory cytokines, while p75ECD-Fc inhibited the expression of inflammatory cytokines. Given there was an inflammatory response of lymphocytes to proBDNF, it is suggested that proBDNF/p75NTR signaling may upstream inflammatory cytokines in MDD. Our data suggest that proBDNF/p75NTR signaling may not only serve as biomarkers but also may be a potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Chun-Rui Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, P. R. China
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, P. R. China
| | - Rui Liang
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, P. R. China
| | - Yuan Liu
- Tianjin Anding Hospital, Tianjin, P. R. China
| | - Fan-Jie Meng
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P. R. China
| | - Fiona Zhou
- Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, Australia
- Garvan Institute of Medical Research, St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia
| | - Xiao-Yang Zhang
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, P. R. China
| | - Li Ning
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, P. R. China
| | - Zhi-Qiang Wang
- Department of Anorectal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, P. R. China
| | - Shuang Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, P. R. China
| | - Xin-Fu Zhou
- Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, Australia
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
- Suzhou Auzone Biotech Ltd, Suzhou International Park, Suzhou, Jiangsu Province, P.R. China
| |
Collapse
|
6
|
Otsuka N, Takaesu Y, Zamami Y, Ota K, Kurihara K, Shinzato H, Kondo T. Elevated Brain-Derived Neurotrophic Factor Levels During Depressive Mixed States. Psychiatry Investig 2023; 20:1027-1033. [PMID: 37997330 PMCID: PMC10678153 DOI: 10.30773/pi.2023.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/23/2023] [Accepted: 08/06/2023] [Indexed: 11/25/2023] Open
Abstract
OBJECTIVE Neurotrophin-like brain-derived neurotrophic factor (BDNF) and pro-inflammatory cytokines may modulate the pathophysiology of mood disorders. Although several studies show alterations in these biomarkers during the depressive, manic, and euthymic states of mood disorders, evidence is lacking for those in a mixed state. Therefore, this study aimed to investigate the relationship between the depressive mixed state (DMX) and peripheral neurobiological factors. METHODS We enrolled 136 patients with major depressive episodes. Depressive symptoms were assessed using the Quick Inventory of Depressive Symptomatology Self-Report Japanese version (QIDS-SR-J). The severity of DMX was assessed using the self-administered 12-item questionnaire (DMX-12). Categorical screening as DMX-positive (n=54) was determined by a cutoff score of 13 or more in the specific eight symptoms from the DMX-12; the remaining were DMX-negative (n=82). Serum BDNF, tumor necrosis factor-α, highsensitivity C-reactive protein, and interleukin-6 levels were measured. RESULTS When comparing biomarkers between the DMX-positive and DMX-negative groups, higher serum BDNF concentration in the DMX-positive group than in the DMX-negative group was the only significant finding (p=0.009). A positive correlation existed between the total score of the eight specific symptoms of DMX-12 and the BDNF concentration (r=0.190, p=0.027). After adjustment for confounders, logistic regression analysis revealed that BDNF (odds ratio [OR]=1.07, 95% confidence interval [CI]=1.00-1.14, p=0.045), bipolar diagnosis (OR=3.43, 95% CI=1.36-8.66, p=0.009), and total QIDS-SR-J score (OR=1.29, 95% CI=1.15-1.43, p<0.001) were significantly associated with DMX positivity. CONCLUSION BDNF was positively associated with DMX severity, suggesting that higher BDNF concentrations may be involved in the pathophysiology of DMX.
Collapse
Affiliation(s)
- Naoaki Otsuka
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yoshikazu Takaesu
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yu Zamami
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kazuki Ota
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kazuhiro Kurihara
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hotaka Shinzato
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Tsuyoshi Kondo
- Department of Neuropsychiatry, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
7
|
Wu Y, Dong Z, Jiang X, Qu L, Zhou W, Sun X, Hou J, Xu H, Cheng M. Gut Microbiota Taxon-Dependent Transformation of Microglial M1/M2 Phenotypes Underlying Mechanisms of Spatial Learning and Memory Impairment after Chronic Methamphetamine Exposure. Microbiol Spectr 2023; 11:e0030223. [PMID: 37212669 PMCID: PMC10269813 DOI: 10.1128/spectrum.00302-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/06/2023] [Indexed: 05/23/2023] Open
Abstract
Methamphetamine (METH) exposure may lead to cognitive impairment. Currently, evidence suggests that METH exposure alters the configuration of the gut microbiota. However, the role and mechanism of the gut microbiota in cognitive impairment after METH exposure are still largely unknown. Here, we investigated the impact of the gut microbiota on the phenotype status of microglia (microglial phenotypes M1 and microglial M2) and their secreting factors, the subsequent hippocampal neural processes, and the resulting influence on spatial learning and memory of chronically METH-exposed mice. We determined that gut microbiota perturbation triggered the transformation of microglial M2 to M1 and a subsequent change of pro-brain-derived neurotrophic factor (proBDNF)-p75NTR-mature BDNF (mBDNF)-TrkB signaling, which caused reduction of hippocampal neurogenesis and synaptic plasticity-related proteins (SYN, PSD95, and MAP2) and, consequently, deteriorated spatial learning and memory. More specifically, we found that Clostridia, Bacteroides, Lactobacillus, and Muribaculaceae might dramatically affect the homeostasis of microglial M1/M2 phenotypes and eventually contribute to spatial learning and memory decline after chronic METH exposure. Finally, we found that fecal microbial transplantation could protect against spatial learning and memory decline by restoring the microglial M1/M2 phenotype status and the subsequent proBDNF-p75NTR/mBDNF-TrkB signaling in the hippocampi of chronically METH-exposed mice. IMPORTANCE Our study indicated that the gut microbiota contributes to spatial learning and memory dysfunction after chronic METH exposure, in which microglial phenotype status plays an intermediary role. The elucidated "specific microbiota taxa-microglial M1/M2 phenotypes-spatial learning and memory impairment" pathway would provide a novel mechanism and elucidate potential gut microbiota taxon targets for the no-drug treatment of cognitive deterioration after chronic METH exposure.
Collapse
Affiliation(s)
- Yulong Wu
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Zhouyan Dong
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Xinze Jiang
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Lei Qu
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Wei Zhou
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Xu Sun
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Jiangshan Hou
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Hongmei Xu
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Mei Cheng
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| |
Collapse
|
8
|
Cade S, Zhou XF, Bobrovskaya L. An Imbalance in the Pro/mature BDNF Ratio Occurs in Multiple Brain Regions During Normal Ageing in Wild-Type Mice. J Mol Neurosci 2023; 73:469-484. [PMID: 37314606 PMCID: PMC10432372 DOI: 10.1007/s12031-023-02131-0] [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: 02/23/2023] [Accepted: 05/27/2023] [Indexed: 06/15/2023]
Abstract
The early transition to Alzheimer's disease is characterized by a period of accelerated brain atrophy that exceeds normal ageing. Identifying the molecular basis of this atrophy could facilitate the discovery of novel drug targets. The precursor of brain-derived neurotrophic factor, a well characterized neurotrophin, is increased in the hippocampus of aged rodents, while its mature isoform is relatively stable. This imbalance could increase the risk of Alzheimer's disease by precipitating its pathological hallmarks. However, less is known about how relative levels of these isoforms change in middle-aged mice. In addition, the underlying mechanisms that might cause an imbalance are unknown. The main aim of this study was to determine how precursor brain-derived neurotrophic factor changes relative to its mature isoform with normal brain ageing in wild type mice. A secondary aim was to determine if signaling through the neurotrophin receptor, p75 influences this ratio. An increasing ratio was identified in several brain regions, except the hippocampus, suggesting a neurotrophic imbalance occurs as early as middle age. Some changes in receptors that mediate the isoforms effects were also identified, but these did not correspond with trends in the isoforms. Relative amounts of precursor brain-derived neurotrophic factor were mostly unchanged in mutant p75 mice. The lack of changes suggested that signaling through the receptor had no influence on the ratio.
Collapse
Affiliation(s)
- Shaun Cade
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia.
| |
Collapse
|
9
|
Li Q, Hu YZ, Gao S, Wang PF, Hu ZL, Dai RP. ProBDNF and its receptors in immune-mediated inflammatory diseases: novel insights into the regulation of metabolism and mitochondria. Front Immunol 2023; 14:1155333. [PMID: 37143663 PMCID: PMC10151479 DOI: 10.3389/fimmu.2023.1155333] [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: 01/31/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs) consist of a common and clinically diverse group of diseases. Despite remarkable progress in the past two decades, no remission is observed in a large number of patients, and no effective treatments have been developed to prevent organ and tissue damage. Brain-derived neurotrophic factor precursor (proBDNF) and receptors, such as p75 neurotrophin receptor (p75NTR) and sortilin, have been proposed to mediate intracellular metabolism and mitochondrial function to regulate the progression of several IMIDs. Here, the regulatory role of proBDNF and its receptors in seven typical IMIDs, including multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, allergic asthma, type I diabetes, vasculitis, and inflammatory bowel diseases, was investigated.
Collapse
Affiliation(s)
- Qiao Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Anesthesia Medical Research Center, Central South University, Changsha, Hunan, China
| | - Yue-Zi Hu
- Clinical Laboratory, The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shan Gao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Anesthesia Medical Research Center, Central South University, Changsha, Hunan, China
| | - Peng-Fei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Anesthesia Medical Research Center, Central South University, Changsha, Hunan, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Anesthesia Medical Research Center, Central South University, Changsha, Hunan, China
- *Correspondence: Ru-Ping Dai, ; Zhao-Lan Hu,
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Anesthesia Medical Research Center, Central South University, Changsha, Hunan, China
- *Correspondence: Ru-Ping Dai, ; Zhao-Lan Hu,
| |
Collapse
|
10
|
Sun W, Li J, Li X, Chen X, Mei Y, Yang Y, An L. Aluminium oxide nanoparticles compromise spatial memory performance and proBDNF-mediated neuronal function in the hippocampus of rats. Part Fibre Toxicol 2022; 19:34. [PMID: 35538555 PMCID: PMC9087928 DOI: 10.1186/s12989-022-00477-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
Background Alumina nanoparticles (aluminaNPs), which are widely used in a range of daily and medical fields, have been shown to penetrate blood-brain barrier, and distribute and accumulate in different brain areas. Although oral treatment of aluminaNPs induces hippocampus-dependent learning and memory impairments, characteristic effects and exact mechanisms have not been fully elucidated. Here, male adult rats received a single bilateral infusion of aluminaNPs (10 or 20 µg/kg of body weight) into the hippocampal region, and their behavioral performance and neural function were assessed. Results The results indicated that the intra-hippocampus infusions at both doses of aluminaNPs did not cause spatial learning inability but memory deficit in the water maze task. This impairment was attributed to the effects of aluminaNP on memory consolidation phase through activation of proBDNF/RhoA pathway. Inhibition of the increased proBDNF by hippocampal infusions of p75NTR antagonist could effectively rescue the memory impairment. Incubation of aluminaNPs exaggerated GluN2B-dependent LTD induction with no effects on LTD expression in hippocampal slices. AluminaNP could also depress the amplitude of NMDA-GluN2B EPSCs. Meanwhile, increased reactive oxygen specie production was reduced by blocking proBDNF-p75NTR pathway in the hippocampal homogenates. Furthermore, the neuronal correlate of memory behavior was drastically weakened in the aluminaNP-infused groups. The dysfunction of synaptic and neuronal could be obviously mitigated by blocking proBDNF receptor p75NTR, implying the involvement of proBDNF signaling in aluminaNP-impaired memory process. Conclusions Taken together, our findings provide the first evidence that the accumulation of aluminaNPs in the hippocampus exaggeratedly activates proBDNF signaling, which leads to neural and memory impairments.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Jia Li
- College of Acupuncture and Orthopedics, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xiaoliang Li
- Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China
| | - Xiao Chen
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China
| | - Yazi Mei
- Graduate School of Guangzhou, University of Chinese Medicine, Guangzhou, 510006, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Lei An
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China. .,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China. .,Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China. .,Graduate School of Guangzhou, University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
11
|
BDNF and Pro-BDNF in Amyotrophic Lateral Sclerosis: A New Perspective for Biomarkers of Neurodegeneration. Brain Sci 2022; 12:brainsci12050617. [PMID: 35625004 PMCID: PMC9139087 DOI: 10.3390/brainsci12050617] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is characterized by the progressive degeneration of upper or lower motor neurons, leading to muscle wasting and paralysis, resulting in respiratory failure and death. The precise ALS aetiology is poorly understood, mainly due to clinical and genetic heterogeneity. Thus, the identification of reliable biomarkers of disease could be helpful in clinical practice. In this study, we investigated whether the levels of brain-derived neurotrophic factor (BDNF) and its precursor Pro-BDNF in serum and cerebrospinal fluid (CSF) may reflect the pathological changes related to ALS. We found higher BDNF and lower Pro-BDNF levels in ALS sera compared to healthy controls. BDNF/Pro-BDNF ratio turned out to be accurate in distinguishing ALS patients from controls. Then, the correlations of these markers with several ALS clinical variables were evaluated. This analysis revealed three statistically significant associations: (1) Patients carrying the C9orf72 expansion significantly differed from non-carrier patients and showed serum BDNF levels comparable to control subjects; (2) BDNF levels in CSF were significantly higher in ALS patients with faster disease progression; (3) lower serum levels of Pro-BDNF were associated with a shorter survival. Therefore, we suggest that BDNF and Pro-BDNF, alone or in combination, might be used as ALS prognostic biomarkers.
Collapse
|
12
|
Omar NA, Kumar J, Teoh SL. Neurotrophin-3 and neurotrophin-4: The unsung heroes that lies behind the meninges. Neuropeptides 2022; 92:102226. [PMID: 35030377 DOI: 10.1016/j.npep.2022.102226] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/06/2021] [Accepted: 01/04/2022] [Indexed: 11/17/2022]
Abstract
Neurotrophin is a growth factor that regulates the development and repair of the nervous system. From all factors, two pioneer groups, the nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF), have been widely explored for their role in disease pathogenesis and potential use as therapeutic agents. Nonetheless, neurotrophin-3 (NT3) and neurotrophin-4 (NT4) also have promising potential, albeit less popular than their counterparts. This review focuses on the latter two factors and their roles in the pathogenesis of brain disorders and potential therapies. An extensive literature search of NT3 and NT4 with their receptors, the TrkB and TrkC on the nervous system were extracted and analyzed. We found that NT3 and NT4 are not only involved in the pathogenesis of some neurodegenerative diseases, but also have promising therapeutic potential on injury- and vascular-related nervous system disease, neuropsychiatry, neurodegeneration and peripheral nerve diseases. In conclusion, the role of NT3 and NT4 should be further emphasized, and more studies could be explored on the potential use of these neurotrophins in the human study.
Collapse
Affiliation(s)
- Noor Azzizah Omar
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia; Department of Medical Sciences, Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, 71800 Nilai, Negeri Sembilan, Malaysia.
| | - Jaya Kumar
- Department of Physiology, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia.
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia.
| |
Collapse
|
13
|
Yang CR, Ding HJ, Yu M, Zhou FH, Han CY, Liang R, Zhang XY, Zhang XL, Meng FJ, Wang S, Li DD, Sun WZ, Meng B, Zhou XF. proBDNF/p75NTR promotes rheumatoid arthritis and inflammatory response by activating proinflammatory cytokines. FASEB J 2022; 36:e22180. [PMID: 35129860 DOI: 10.1096/fj.202101558r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 11/11/2022]
Abstract
P75 pan-neurotrophin receptor (p75NTR) is an important receptor for the role of neurotrophins in survival and death of neurons during development and after nerve injury. Our previous research found that the precursor of brain-derived neurotrophic factor (proBDNF) regulates pain as an inflammatory mediator. The current understanding of the role of proBDNF/p75NTR signaling pathway in inflammatory arthritis pain and rheumatoid arthritis (RA) is unclear. We recruited 20 RA patients, 20 healthy donors (HDs), and 10 osteoarthritis (OA) patients. Hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) of proBDNF and p75NTR in synovial membrane were performed and evaluated. We next examined the mRNA and protein expression of proBDNF/p75NTR signaling pathway in peripheral blood mononuclear cells (PBMCs) and synovial tissue. ELISA and flow cytometry were assessed between the blood of RA patients and HD. To induce RA, collagen-induced arthritis (CIA) were induced in mice. We found over-synovitis of RA synovial membrane compared to OA controls in histologic sections. P75NTR and sortilin mRNA, and proBDNF protein level were significantly increased in PBMCs of RA patients compared with the HD. Consistently, ELISA showed that p75NTR, sortilin, tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-10 (IL-10) levels in the serum of RA patients were increased compared with HD and p75NTR, sortilin were positively correlated with Disease Activity Score in 28 joints (DAS28). In addition, using flow cytometry we showed that the increased levels of proBDNF and p75NTR characterized in CD4+ and CD8+ T cells of RA patients were subsequently reversed with methotrexate (MTX) treatment. Furthermore, we found pathological changes, inflammatory pain, upregulation of the mRNA and protein expression of proBDNF/p75NTR signaling pathway, and upregulation of inflammatory cytokines in spinal cord using a well-established CIA mouse model. We showed intravenous treatment of recombinant p75ECD-Fc that biologically blocked all inflammatory responses and relieved inflammatory pain of animals with CIA. Our findings showed the involvement of proBDNF/p75NTR pathway in the RA inflammatory response and how blocking it with p75ECD-Fc may be a promising therapeutic treatment for RA.
Collapse
Affiliation(s)
- Chun-Rui Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, P. R. China.,Department of Pathology, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Hong-Jun Ding
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, P. R. China.,Tianjin Public Security Profession College, Tianjin, P. R. China
| | - Miao Yu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China
| | - Fiona-H Zhou
- Faculty of Health Sciences, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Chen-Yang Han
- Pain Treatment Center, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Rui Liang
- Department of Pathology, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Xiao-Yang Zhang
- Department of Pathology, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Xiang-Lian Zhang
- Department of Pathology, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Fan-Jie Meng
- Department of Thoracic Surgery, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Shuo Wang
- Department of Thoracic Surgery, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - De-Dong Li
- Department of Anesthesiology, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Wei-Zong Sun
- Department of Orthopedics, Tianjin Medical University Second Hospital, Tianjin, P. R. China
| | - Bin Meng
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Cade S, Zhou XF, Bobrovskaya L. The role of brain-derived neurotrophic factor and the neurotrophin receptor p75NTR in age-related brain atrophy and the transition to Alzheimer's disease. Rev Neurosci 2022; 33:515-529. [PMID: 34982865 DOI: 10.1515/revneuro-2021-0111] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/11/2021] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease is a neurodegenerative condition that is potentially mediated by synaptic dysfunction before the onset of cognitive impairments. The disease mostly affects elderly people and there is currently no therapeutic which halts its progression. One therapeutic strategy for Alzheimer's disease is to regenerate lost synapses by targeting mechanisms involved in synaptic plasticity. This strategy has led to promising drug candidates in clinical trials, but further progress needs to be made. An unresolved problem of Alzheimer's disease is to identify the molecular mechanisms that render the aged brain susceptible to synaptic dysfunction. Understanding this susceptibility may identify drug targets which could halt, or even reverse, the disease's progression. Brain derived neurotrophic factor is a neurotrophin expressed in the brain previously implicated in Alzheimer's disease due to its involvement in synaptic plasticity. Low levels of the protein increase susceptibility to the disease and post-mortem studies consistently show reductions in its expression. A desirable therapeutic approach for Alzheimer's disease is to stimulate the expression of brain derived neurotrophic factor and potentially regenerate lost synapses. However, synthesis and secretion of the protein are regulated by complex activity-dependent mechanisms within neurons, which makes this approach challenging. Moreover, the protein is synthesised as a precursor which exerts the opposite effect of its mature form through the neurotrophin receptor p75NTR. This review will evaluate current evidence on how age-related alterations in the synthesis, processing and signalling of brain derived neurotrophic factor may increase the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Shaun Cade
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
15
|
Molecular Mechanism for PACAP 38-Induced Neurite Outgrowth in PC12 Cells. Neural Plast 2021; 2021:2522454. [PMID: 34422037 PMCID: PMC8371652 DOI: 10.1155/2021/2522454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/08/2021] [Accepted: 07/17/2021] [Indexed: 11/17/2022] Open
Abstract
The present research investigates the molecular mechanism of neurite outgrowth (protrusion elongation) under pituitary adenylate cyclase-activating polypeptide (PACAP) 38 treatments using a rat adrenal-derived pheochromocytoma cell line-PC12. This study specifically looks into the regulation of PACAP38-induced collapsing response mediator protein 2 (CRMP2) previously identified in a mouse brain ischemia model and which could be recovered by PACAP38 treatment. Previously, DNA microarray analysis revealed that PACAP 38-mediated neuroprotection involved not only CRMP2 but also pathways related to glycogen synthase kinase-3β (GSK-3β) and other signaling components. Thus, to clarify whether CRMP2 acts directly on PACAP38 or through GSK-3β as part of the mechanism of PACAP38-induced neurite outgrowth, we observed neurite outgrowth in the presence of GSK-3β inhibitors and activators. PC12 cells were treated with PACAP38 being added to the cell culture medium at concentrations of 10-7 M, 10-8 M, and 10-9 M. Post PACAP38 treatment, immunostaining was used to confirm protrusion elongation of the PC12 cells, while RT-PCR, two-dimensional gel electrophoresis in conjunction with Western blotting, and inhibition experiments were performed to confirm the expression of the PACAP gene, its receptors, and downstream signaling components. Our data show that neurite protrusion elongation by PACAP38 (10-7 M) in PC12 cells is mediated through the PAC1-R receptor as demonstrated by its suppression by a specific inhibitor PA-8. Inhibitor experiments suggested that PACAP38-triggered neurite protrusion follows a GSK-3β-regulated pathway, where the AKT and cAMP/ERK pathways are involved and where the inhibition of Rho/Roc could enhance neurite protrusion under PACAP38 stimulation. Although we could not yet confirm the exact role and position of CRMP2 in PACAP38-mediated PC12 cell elongation, it appears that its phosphorylation and dephosphorylation have a correlation with the neurite protrusion elongation through the interplay of CDK5, which needs to be investigated further.
Collapse
|
16
|
Cui YH, Zhou SF, Liu Y, Wang S, Li F, Dai RP, Hu ZL, Li CQ. Injection of Anti-proBDNF Attenuates Hippocampal-Dependent Learning and Memory Dysfunction in Mice With Sepsis-Associated Encephalopathy. Front Neurosci 2021; 15:665757. [PMID: 34354558 PMCID: PMC8329425 DOI: 10.3389/fnins.2021.665757] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/26/2021] [Indexed: 12/14/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a risk factor for cognitive and memory dysfunction; however, the mechanism remains unclear. Brain-derived neurotrophic factor (BDNF) was reported to have a positive effect on cognition and emotion regulation, but the study of its precursor, proBDNF, has been limited. This study aimed to elucidate the effects and associated mechanisms of hippocampal proBDNF in a lipopolysaccharide (LPS)-induced SAE mouse model. In this study, we found that the mice exhibited cognitive dysfunction on day 7 after LPS injection. The expression of proBDNF and its receptor, p75NTR, was also increased in the hippocampus, while the levels of BDNF and its receptor, TrkB, were decreased. A co-localization study showed that proBDNF and p75NTR were mainly co-localized with neurons. Furthermore, LPS treatment reduced the expression of NeuN, Nissl bodies, GluR4, NR1, NR2A, and NR2B in the hippocampus of SAE mice. Furthermore, an intrahippocampal or intraperitoneal injection of anti-proBDNF antibody was able to ameliorate LPS-induced cognitive dysfunction and restore the expression of NeuN, Nissl bodies, GluR4, NR1, NR2A, NR2B, and PSD95. These results indicated that treatment with brain delivery by an intrahippocampal and systemic injection of mAb-proBDNF may represent a potential therapeutic strategy for treating patients with SAE.
Collapse
Affiliation(s)
- Yan-Hui Cui
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China.,Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Shi-Fen Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Yu Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China.,Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Wang
- Department of Medical Research Center and Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Qi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| |
Collapse
|
17
|
Sun W, Cheng H, Yang Y, Tang D, Li X, An L. Requirements of Postnatal proBDNF in the Hippocampus for Spatial Memory Consolidation and Neural Function. Front Cell Dev Biol 2021; 9:678182. [PMID: 34336832 PMCID: PMC8319730 DOI: 10.3389/fcell.2021.678182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mature brain-derived neurotrophic factor (BDNF) and its downstream signaling pathways have been implicated in regulating postnatal development and functioning of rodent brain. However, the biological role of its precursor pro-brain-derived neurotrophic factor (proBDNF) in the postnatal brain remains unknown. The expression of hippocampal proBDNF was blocked in postnatal weeks, and multiple behavioral tests, Western blot and morphological techniques, and neural recordings were employed to investigate how proBDNF played a role in spatial cognition in adults. The peak expression and its crucial effects were found in the fourth but not in the second or eighth postnatal week. Blocking proBDNF expression disrupted spatial memory consolidation rather than learning or memory retrieval. Structurally, blocking proBDNF led to the reduction in spine density and proportion of mature spines. Although blocking proBDNF did not affect N-methyl-D-aspartate (NMDA) receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, the learning-induced phosphorylation of the GluN2B subunit level declined significantly. Functionally, paired-pulse facilitation, post-low-frequency stimulation (LFS) transiently enhanced depression, and GluN2B-dependent short-lasting long-term depression in the Schaffer collateral-CA1 pathway were weakened. The firing rate of pyramidal neurons was significantly suppressed around the target region during the memory test. Furthermore, the activation of GluN2B-mediated signaling could effectively facilitate neural function and mitigate memory impairment. The findings were consistent with the hypothesis that postnatal proBDNF played an essential role in synaptic and cognitive functions.
Collapse
Affiliation(s)
- Wei Sun
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hong Cheng
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dongxin Tang
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaolian Li
- Department of Neurology, Jinan Geriatric Hospital, Jinan, China
| | - Lei An
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
18
|
Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins in Adult Neurogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:95-115. [PMID: 34453295 DOI: 10.1007/978-3-030-74046-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell survival during adult neurogenesis and the modulation of each step, namely, proliferation, lineage differentiation, migration, maturation, and functional integration of the newborn cells into the existing circuitry, is regulated by intrinsic and extrinsic factors. Transduction of extracellular niche signals triggers the activation of intracellular mechanisms that regulate adult neurogenesis by affecting gene expression. While the intrinsic factors include transcription factors and epigenetic regulators, the extrinsic factors are molecular signals that are present in the neurogenic niche microenvironment. These include morphogens, growth factors, neurotransmitters, and signaling molecules secreted as soluble factors or associated to the extracellular matrix. Among these molecular mechanisms are neurotrophins and neurotrophin receptors which have been implicated in the regulation of adult neurogenesis at different levels, with brain-derived neurotrophic factor (BDNF) being the most studied neurotrophin. In this chapter, we review the current knowledge about the role of neurotrophins in the regulation of adult neurogenesis in both the subventricular zone (SVZ) and the hippocampal subgranular zone (SGZ).
Collapse
|
19
|
Hu ZL, Luo C, Hurtado PR, Li H, Wang S, Hu B, Xu JM, Liu Y, Feng SQ, Hurtado-Perez E, Chen K, Zhou XF, Li CQ, Dai RP. Brain-derived neurotrophic factor precursor in the immune system is a novel target for treating multiple sclerosis. Am J Cancer Res 2021; 11:715-730. [PMID: 33391501 PMCID: PMC7738849 DOI: 10.7150/thno.51390] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/17/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: Brain-derived neurotrophic factor precursor (proBDNF) is expressed in the central nervous system (CNS) and the immune system. However, the role of proBDNF in the pathogenesis of multiple sclerosis (MS) is unknown. Methods: Peripheral blood and post-mortem brain and spinal cord specimens were obtained from multiple sclerosis patients to analyze proBDNF expression in peripheral lymphocytes and infiltrating immune cells in the lesion site. The proBDNF expression profile was also examined in the experimental autoimmune encephalomyelitis (EAE) mouse model, and polyclonal and monoclonal anti-proBDNF antibodies were used to explore their therapeutic effect in EAE. Finally, the role of proBDNF in the inflammatory immune activity of peripheral blood mononuclear cells (PBMCs) was verified in vitro experiments. Results: High proBDNF expression was detected in the circulating lymphocytes and infiltrated inflammatory cells at the lesion sites of the brain and spinal cord in MS patients. In the EAE mouse model, proBDNF was upregulated in CNS and in circulating and splenic lymphocytes. Systemic but not intracranial administration of anti-proBDNF blocking antibodies attenuated clinical scores, limited demyelination, and inhibited proinflammatory cytokines in EAE mice. Immuno-stimulants treatment increased the proBDNF release and upregulated the expression of p75 neurotrophic receptors (p75NTR) in lymphocytes. The monoclonal antibody against proBDNF inhibited the inflammatory response of PBMCs upon stimulations. Conclusion: The findings suggest that proBDNF from immune cells promotes the immunopathogenesis of MS. Monoclonal Ab-proB may be a promising therapeutic agent for treating MS.
Collapse
|
20
|
Lin L, Zhou XF, Bobrovskaya L. Blockage of p75 NTR ameliorates depressive-like behaviours of mice under chronic unpredictable mild stress. Behav Brain Res 2021; 396:112905. [PMID: 32926907 DOI: 10.1016/j.bbr.2020.112905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/15/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
The precursor of brain derived neurotrophic factor (proBDNF) and its receptor p75NTR are upregulated in depressive patients and chronic stress-induced depressive animals, suggesting that activation of p75NTR signalling may underlie pathogenesis of depression. In the present study we hypothesize that the blockade of p75NTR may have therapeutic effect on depressive mice under chronic stress. The treatment of mice with the recombinant fusion protein of p75NTR extracellular domain and fragment C of immunoglobulin (p75ECD-Fc) significantly reduced the immobility time in the forced swim test and tail suspension test, and increased the time spent in the central zone in the open field test in mice exposed to chronic unpredictable mild stress (CUMS). p75ECD-Fc treatment also significantly increased the length and density of neuronal dendritic spines in the dentate gyrus and amygdala. Our data indicate that blocking p75NTR signalling can alleviate depressive and anxiety-like behaviours of chronically stressed mice and improve the dendritic spinogenesis of neurons under stress.
Collapse
Affiliation(s)
- Liying Lin
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.
| |
Collapse
|
21
|
Kazana W, Zabłocka A. Brain-derived neurotrophic factor as a potential therapeutic
tool in the treatment of nervous system disorders. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.5678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in the proper functioning
of the nervous system. It regulates the growth and survival of nerve cells, and is crucial
in processes related to the memory, learning and synaptic plasticity. Abnormalities related
to the distribution and secretion of BDNF protein accompany many diseases of the nervous
system, in the course of which a significant decrease in BDNF level in the brain is observed.
Impairments of BDNF transport may occur, for example, in the event of a single nucleotide
polymorphism in the Bdnf (Val66Met) coding gene or due to the dysfunctions of the proteins
involved in intracellular transport, such as huntingtin (HTT), huntingtin-associated protein
1 (HAP1), carboxypeptidase E (CPE) or sortilin 1 (SORT1). One of the therapeutic goals in the treatment of diseases of the central nervous system may be the regulation of expression and
secretion of BDNF protein by nerve cells. Potential therapeutic strategies are based on direct
injection of the protein into the specific region of the brain, the use of viral vectors expressing
the Bdnf gene, transplantation of BDNF-producing cells, the use of substances of natural
origin that stimulate the cells of the central nervous system for BDNF production, or the
use of molecules activating the main receptor for BDNF – tyrosine receptor kinase B (TrkB).
In addition, an appropriate lifestyle that promotes physical activity helps to increase BDNF
level in the body. This paper summarizes the current knowledge about the biological role of
BDNF protein and proteins involved in intracellular transport of this neurotrophin. Moreover,
it presents contemporary research trends to develop therapeutic methods, leading to an
increase in the level of BDNF protein in the brain.
Collapse
Affiliation(s)
- Wioletta Kazana
- Laboratorium Immunobiologii Mikrobiomu, Instytut Immunologii i Terapii Doświadczalnej Polskiej Akademii Nauk im. L. Hirszfelda we Wrocławiu
| | - Agnieszka Zabłocka
- Laboratorium Immunobiologii Mikrobiomu, Instytut Immunologii i Terapii Doświadczalnej Polskiej Akademii Nauk im. L. Hirszfelda we Wrocławiu
| |
Collapse
|
22
|
Tanaka S, Miyashita M, Wakabayashi N, O'Hashi K, Tani T, Ribot J. Development and Reorganization of Orientation Representation in the Cat Visual Cortex: Experience-Dependent Synaptic Rewiring in Early Life. Front Neuroinform 2020; 14:41. [PMID: 32973480 PMCID: PMC7468406 DOI: 10.3389/fninf.2020.00041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/28/2020] [Indexed: 11/13/2022] Open
Abstract
To date, numerous mathematical models have been proposed on the basis of some types of Hebbian synaptic learning to account for the activity-dependent development of orientation maps as well as neuronal orientation selectivity. These models successfully reproduced orientation map-like spatial patterns. Nevertheless, we still have questions: (1) How does synaptic rewiring occur in the visual cortex during the formation of orderly orientation maps in early life? (2) How does visual experience contribute to the maturation of orientation selectivity of visual cortical neurons and reorganize orientation maps? (3) How does the sensitive period for orientation plasticity end? In this study, we performed animal experiments and mathematical modeling to understand the mechanisms underlying synaptic rewiring for experience-dependent formation and reorganization of orientation maps. At first, we visualized orientation maps from the intrinsic signal optical imaging in area 17 of kittens reared under single-orientation exposure through cylindrical-lens-fitted goggles. The experiments revealed that the degree of expansion of cortical domains representing the experienced orientation depends on the age at which the single-orientation exposure starts. As a result, we obtained the sensitive period profile for orientation plasticity. Next, we refined our previously proposed mathematical model for the activity-dependent self-organization of thalamo-cortical inputs on the assumption that rewiring is caused by the competitive interactions among transient synaptic contacts on the same dendritic spine. Although various kinds of molecules have been reported to be involved in such interactions, we attempt to build a mathematical model to describe synaptic rewiring focusing on brain-derived neurotrophic factor (BDNF) and its related molecules. Performing computer simulations based on the refined model, we successfully reproduced orientation maps reorganized in kittens reared under single-orientation exposure as well as normal visual experience. We also reproduced the experimentally obtained sensitive period profile for orientation plasticity. The excellent agreement between experimental observations and theoretical reproductions suggests that the BDNF-induced competitive interaction among synaptic contacts from different axons on the same spine is an important factor for the experience-dependent formation and reorganization of orientation selectivity and orientation maps.
Collapse
Affiliation(s)
- Shigeru Tanaka
- Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, Chofu, Japan
| | - Masanobu Miyashita
- Department of Control and Computer Engineering, National Institute of Technology, Numazu College, Numazu, Japan
| | - Nodoka Wakabayashi
- Power Plant Engineering, Engineering & Maintenance Center, All Nippon Airways Co., Ltd., Tokyo, Japan
| | - Kazunori O'Hashi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Toshiki Tani
- Laboratory for Molecular Analysis of Higher Brain Functions, RIKEN Center for Brain Science, Wako, Japan
| | - Jérôme Ribot
- Centre for Interdisciplinary Research in Biology, Collège de France, Paris, France
| |
Collapse
|
23
|
Sun W, Che H, Li J, Tang D, Liu X, Liu W, An L. Dorsolateral Striatal proBDNF Improves Reversal Learning by Enhancing Coordination of Neural Activity in Rats. Mol Neurobiol 2020; 57:4642-4656. [DOI: 10.1007/s12035-020-02051-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022]
|
24
|
Dexmedetomidine Attenuates Neurotoxicity in Developing Rats Induced by Sevoflurane through Upregulating BDNF-TrkB-CREB and Downregulating ProBDNF-P75NRT-RhoA Signaling Pathway. Mediators Inflamm 2020; 2020:5458061. [PMID: 32655312 PMCID: PMC7322616 DOI: 10.1155/2020/5458061] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/23/2020] [Indexed: 12/22/2022] Open
Abstract
To investigate the mechanism dexmedetomidine in relieving the neurotoxicity of a developing brain induced by sevoflurane. Sprague-Dawley rats, 6 days old, were randomly divided into three groups. Rats in the control group were inhaled with air after injection of normal saline; rats in the sevoflurane group were injected with normal saline and inhaled with 3% sevoflurane for 2 h in three consecutive day; rats in the dexmedetomidine group were inhaled with 3% sevoflurane after intraperitoneal injection of dexmedetomidine 25 μg/kg. WB results showed that mBDNF, pTrkB/TrkB, and CREB were significantly decreased in the hippocampus of the sevoflurane group, which are significantly upregulated in the dexmedetomidine group. In the sevoflurane group, proBDNF, P75NRT, and RhoA were significantly increased, which were significantly lower than those in the dexmedetomidine group than those in the sevoflurane group. The expression BDNF was downregulated in the sevoflurane group, while the proBDNF was upregulated in the sevoflurane group. In the Morris water maze test, the escape latency of the sevoflurane group was significantly prolonged. In sevoflurane groups, the number of crossing platform was significantly reduced, the synaptic protein decreased significantly, and this effect was reversed in rats of the dexmedetomidine group. Dexmedetomidine could reduce synaptic plasticity decline in developing rats induced by sevoflurane, through downregulating the proBDNF-p75NTR-RhoA pathway and upregulating BDNF-TrkB-CREB.
Collapse
|
25
|
Song HD, Kim SN, Saha A, Ahn SY, Akindehin S, Son Y, Cho YK, Kim M, Park JH, Jung YS, Lee YH. Aging-Induced Brain-Derived Neurotrophic Factor in Adipocyte Progenitors Contributes to Adipose Tissue Dysfunction. Aging Dis 2020; 11:575-587. [PMID: 32489703 PMCID: PMC7220283 DOI: 10.14336/ad.2019.0810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/10/2019] [Indexed: 12/22/2022] Open
Abstract
Aging-related adipose tissue dysfunction contributes to the progression of chronic metabolic diseases. We investigated the role of age-dependent expression of a neurotrophin, brain-derived neurotrophic factor (BDNF) in adipose tissue. Pro-BDNF expression was elevated in epididymal white adipose tissue (eWAT) with advanced age, which was associated with the reduction in sympathetic innervation. Interestingly, BDNF expression was enriched in PDGFRα+ adipocyte progenitors isolated from eWAT, with age-dependent increase in expression. In vitro pro-BDNF treatment caused apoptosis in adipocytes differentiated from C3H10T1/2 cells, and siRNA knockdown of sortilin mitigated these effects. Tamoxifen-inducible PDGFRα+ cell-specific deletion of BDNF (BDNFPdgfra KO) reduced pro-BDNF expression in eWAT, prevented age-associated declines in sympathetic innervation and mitochondrial content in eWAT, and improved insulin sensitivity. Moreover, BDNFPdgfra KO mice showed reduced expression of aging-induced inflammation and senescence markers in eWAT. Collectively, these results identified the upregulation of pro-BDNF expression in adipocyte progenitors as a feature of visceral white adipose tissue aging and suggested that inhibition of BDNF expression in adipocyte progenitors is potentially beneficial to prevent aging-related adipose tissue dysfunction.
Collapse
Affiliation(s)
- Hyun-Doo Song
- 1College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Sang Nam Kim
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Abhirup Saha
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Yeop Ahn
- 1College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Seun Akindehin
- 1College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Yeonho Son
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Keun Cho
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - MinSu Kim
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hyun Park
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Suk Jung
- 3College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yun-Hee Lee
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Liu Y, Zou GJ, Tu BX, Hu ZL, Luo C, Cui YH, Xu Y, Li F, Dai RP, Bi FF, Li CQ. Corticosterone Induced the Increase of proBDNF in Primary Hippocampal Neurons Via Endoplasmic Reticulum Stress. Neurotox Res 2020; 38:370-384. [PMID: 32378057 DOI: 10.1007/s12640-020-00201-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/28/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
Major depression disorder is one of the most common psychiatric disorders that greatly threaten the mental health of a large population worldwide. Previous studies have shown that endoplasmic reticulum (ER) stress plays an important role in the pathophysiology of depression, and current research suggests that brain-derived neurotrophic factor precursor (proBDNF) is involved in the development of depression. However, the relationship between ER and proBDNF in the pathophysiology of depression is not well elucidated. Here, we treated primary hippocampal neurons of mice with corticosterone (CORT) and evaluated the relationship between proBDNF and ERS. Our results showed that CORT induced ERS and upregulated the expression of proBDNF and its receptor, Follistatin-like protein 4 (FSTL4), which contributed to significantly decreased neuronal viability and expression of synaptic-related proteins including NR2A, PSD95, and SYN. Anti-proBDNF neutralization and ISRIB (an inhibitor of the ERS) treatment, respective ly, protected neuronal viabilities and increased the expression of synaptic-related proteins in corticosterone-exposed neurons. ISRIB treatment reduced the expression of proBDNF and FSTL4, whereas anti-proBDNF treatment did not affect ERS markers (Grp78, p-PERK, ATF4) expression. Our study presented evidence that CORT-induced ERS negatively regulated the neuronal viability and the level of synaptic-related protein of primary neurons via the proBDNF/FSTL4 pathway.
Collapse
Affiliation(s)
- Yu Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410013, Hunan, China
| | - Guang-Jing Zou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410013, Hunan, China
| | - Bo-Xuan Tu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410013, Hunan, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Cong Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yan-Hui Cui
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410013, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410013, Hunan, China
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Fang-Fang Bi
- Department of Neurology, Xiang Ya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Chang-Qi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410013, Hunan, China.
| |
Collapse
|
27
|
Wang X, Ma W, Wang T, Yang J, Wu Z, Liu K, Dai Y, Zang C, Liu W, Liu J, Liang Y, Guo J, Li L. BDNF-TrkB and proBDNF-p75NTR/Sortilin Signaling Pathways are Involved in Mitochondria-Mediated Neuronal Apoptosis in Dorsal Root Ganglia after Sciatic Nerve Transection. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:66-82. [PMID: 31957620 DOI: 10.2174/1871527319666200117110056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/12/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023]
Abstract
Background:
Brain-Derived Neurotrophic Factor (BDNF) plays critical roles during development
of the central and peripheral nervous systems, as well as in neuronal survival after injury.
Although proBDNF induces neuronal apoptosis after injury in vivo, whether it can also act as a death
factor in vitro and in vivo under physiological conditions and after nerve injury, as well as its mechanism
of inducing apoptosis, is still unclear.
Objective:
In this study, we investigated the mechanisms by which proBDNF causes apoptosis in sensory
neurons and Satellite Glial Cells (SGCs) in Dorsal Root Ganglia (DRG) After Sciatic Nerve
Transection (SNT).
Methods:
SGCs cultures were prepared and a scratch model was established to analyze the role of
proBDNF in sensory neurons and SGCs in DRG following SNT. Following treatment with proBDNF
antiserum, TUNEL and immunohistochemistry staining were used to detect the expression of Glial
Fibrillary Acidic Protein (GFAP) and Calcitonin Gene-Related Peptide (CGRP) in DRG tissue; immunocytochemistry
and Cell Counting Kit-8 (CCK8) assay were used to detect GFAP expression and
cell viability of SGCs, respectively. RT-qPCR, western blot, and ELISA were used to measure mRNA
and protein levels, respectively, of key factors in BDNF-TrkB, proBDNF-p75NTR/sortilin, and apoptosis
signaling pathways.
Results:
proBDNF induced mitochondrial apoptosis of SGCs and neurons by modulating BDNF-TrkB
and proBDNF-p75NTR/sortilin signaling pathways. In addition, neuroprotection was achieved by inhibiting
the biological activity of endogenous proBDNF protein by injection of anti-proBDNF serum. Furthermore,
the anti-proBDNF serum inhibited the activation of SGCs and promoted their proliferation.
Conclusion:
proBDNF induced apoptosis in SGCs and sensory neurons in DRG following SNT. The
proBDNF signaling pathway is a potential novel therapeutic target for reducing sensory neuron and
SGCs loss following peripheral nerve injury.
Collapse
Affiliation(s)
- Xianbin Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Tongtong Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jinwei Yang
- Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Zhen Wu
- Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Kuangpin Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yunfei Dai
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Chenghao Zang
- Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Wei Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jie Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yu Liang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jianhui Guo
- Second Department of General Surgery, First People’s Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Liyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650500, China
| |
Collapse
|
28
|
Yang CR, Zhang XY, Liu Y, Du JY, Liang R, Yu M, Zhang FQ, Mu XF, Li F, Zhou L, Zhou FH, Meng FJ, Wang S, Ming D, Zhou XF. Antidepressant Drugs Correct the Imbalance Between proBDNF/p75NTR/Sortilin and Mature BDNF/TrkB in the Brain of Mice with Chronic Stress. Neurotox Res 2020; 37:171-182. [PMID: 31493120 DOI: 10.1007/s12640-019-00101-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 02/08/2023]
Abstract
Depression is a worldwide problem with a great social and economic burden in many countries. In our previous research, we found that the expression of proBDNF/p75NTR/sortilin is upregulated in patients with major depressive disorder. In addition, the treatment of proBDNF antibodies reversed both the depressive behaviors and the reduced BDNF mRNA detected in our rodent chronic stress models. Antidepressant drugs are usually only effective in a subpopulation of patients with major depression with a delayed time window of 2-4 weeks to exert their efficacy. The mechanism underlying such delayed response is not known. In this study, we hypothesize that antidepressant drugs exert their therapeutic effect by modulating proBDNF/p75NTR and mature BDNF/TrkB signaling pathways. To test the hypothesis, C57 mice were randomly divided into normal control, chronic unpredictable mild stress (CUMS), vehicle (VEH), fluoxetine (FLU), and clozapine (CLO) groups. Behavioral tests (sucrose preference, open field, and tail suspension tests) were performed before and after 4 weeks of CUMS. The gene and protein expression of proBDNF, the neurotrophin receptor (p75NTR), sortilin, and TrkB in the cortex and hippocampus were examined. At the protein level, CUMS induced a significant increase in proBDNF, p75NTR, and sortilin production while the TrkB protein level was found to be lower in the cortex and hippocampus compared with the control group. Consistently, at the mRNA level, p75NTR expression increased with reduced BDNF/TrkB mRNA in both cortex and hippocampus, while sortilin increased only in the hippocampus after CUMS. FLU and CLO treatments of CUMS mice reversed all protein and mRNA expression of the biomarkers in both cortex and hippocampus, except for sortilin mRNA in the cortex and proBDNF in the hippocampus, respectively. This study further confirms that the imbalance between proBDNF/p75NTR/sortilin and mBDNF/TrkB production is important in the pathogenesis of depression. It is likely that antidepressant FLU and antipsychotic CLO exert their antidepressant-like effect correcting the imbalance between proBDNF/p75NTR/sortilin and mBDNF/TrkB.
Collapse
Affiliation(s)
- C R Yang
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, People's Republic of China
| | - X Y Zhang
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Y Liu
- Tianjin Mental Health Center, Tianjin, People's Republic of China
| | - J Y Du
- Tianjin Mental Health Center, Tianjin, People's Republic of China
| | - R Liang
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - M Yu
- Department of Pathology, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - F Q Zhang
- Department of Neurology, Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - X F Mu
- Department of Clinical Laboratory, Qingdao Central Hospital, Qingdao, Shandong Province, People's Republic of China
| | - F Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, People's Republic of China
| | - L Zhou
- The Mental Hospital of Yunnan Province, Kunming, Yunnan Province, People's Republic of China
| | - F H Zhou
- School of Pharmacology and Medical Science, University of South Australia, Adelaide, 5000, Australia
| | - F J Meng
- Department of Thoracic Surgery, Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - S Wang
- Department of Thoracic Surgery, Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - D Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, People's Republic of China.
| | - X F Zhou
- School of Pharmacology and Medical Science, University of South Australia, Adelaide, 5000, Australia.
| |
Collapse
|
29
|
Shen WY, Luo C, Reinaldo Hurtado P, Hurtado-Perez E, Luo RY, Hu ZL, Li H, Xu JM, Zhou XF, Dai RP. The regulatory role of ProBDNF in monocyte function: Implications in Stanford type-A aortic dissection disease. FASEB J 2019; 34:2541-2553. [PMID: 31908023 DOI: 10.1096/fj.201901905rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
Brain-derived neurotrophic factor precursor (proBDNF) has been reported to strengthen the dysfunction of monocytes/macrophages in animal studies. However, it is still unknown the roles of proBDNF in the dysfunction of monocytes in the inflammatory diseases in humans. In the present study, we showed that proBDNF and pan neurotrophic receptor p75 were significantly upregulated in monocytes from healthy donors (HD) after lipopolysaccharide treatment. Exogenous proBDNF treatment upregulated CD40 and proinflammatory cytokines expression in monocytes including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. In Stanford type-A acute aortic dissection (AAD) patients, proBDNF was upregulated in CD14+ CD163+ CX3CR1+ M2- but not CD14+ CD68+ CCR2+ M1-like monocytes. In addition, sera from AAD patients activated gene expression of proinflammatory cytokines in cultured PBMCs from HD, which was attenuated by proBDNF monoclonal antibody (Ab-proB) treatment. These findings suggested that upregulation of proBDNF in M2-like monocytes may contribute to the proinflammatory response in the AAD.
Collapse
Affiliation(s)
- Wei-Yun Shen
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| | - Cong Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| | | | - Ernesto Hurtado-Perez
- Department of Renal Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Ru-Yi Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| | - Jun-Mei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha City, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha City, China
| |
Collapse
|
30
|
Sleep Deprivation Disrupts Acquisition of Contextual Fear Extinction by Affecting Circadian Oscillation of Hippocampal-Infralimbic proBDNF. eNeuro 2019; 6:ENEURO.0165-19.2019. [PMID: 31585927 PMCID: PMC6800296 DOI: 10.1523/eneuro.0165-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/22/2019] [Accepted: 09/27/2019] [Indexed: 12/25/2022] Open
Abstract
Extensive evidence showed that mature brain-derived neurotrophic factor (mBDNF) levels displayed a circadian pattern. Circadian disruption, for example, sleep deprivation (SD), induced functional and behavioral deficits. However, compared with that of mature form, the biological role of the pro-peptide, proBDNF, was poorly understood. Here, we found that proBDNF was expressed under circadian rhythm in the ventral hippocampus (vHPC). SD rats exhibited deficits in acquisition of conditioned extinction and damped rhythmicity in vHPC proBDNF activity that were accompanied by SD between zeitgeber time (ZT)0 and ZT4, but not the late stage of sleep period. Furthermore, SD affected fear extinction through vHPC-IL proBDNF signaling, which was associated with NR2B subunits of NMDA receptors. More importantly, infusion of proBDNF could mitigate SD-induced abnormal neural activity, by suppressing the enhanced basal firing rate of IL-RS and elevating the depressed neural response that evoked by acquisition of conditioned extinction. Therefore, this finding provided the first evidence that circadian oscillation of vHPC proBDNF activity contributed to the effects of SD on acquisition of conditioned fear extinction, and suggested a new therapeutic target to reverse the cognitive deficits in sleep-related mental disorder, such as post-traumatic stress disorder (PTSD).
Collapse
|
31
|
Buck JM, O'Neill HC, Stitzel JA. Developmental nicotine exposure elicits multigenerational disequilibria in proBDNF proteolysis and glucocorticoid signaling in the frontal cortices, striata, and hippocampi of adolescent mice. Biochem Pharmacol 2019; 168:438-451. [PMID: 31404529 DOI: 10.1016/j.bcp.2019.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/07/2019] [Indexed: 01/03/2023]
Abstract
Maternal smoking of conventional or vapor cigarettes during pregnancy, a form of developmental nicotine exposure (DNE), enhances the risk of neurodevelopmental disorders such as ADHD, autism, and schizophrenia in children. Modeling the multigenerational effects of smoking during pregnancy and nursing in the first- (F1) and second- (F2) generation adolescent offspring of oral nicotine-treated female C57BL/6J mice, we have previously reported that DNE precipitates intergenerational transmission of nicotine preference, hyperactivity and impulsivity-like behaviors, altered rhythmicity of home cage activity, corticostriatal nicotinic acetylcholine receptor and dopamine transporter dysfunction, and corticostriatal global DNA methylome deficits. In aggregate, these DNE-evoked behavioral, neuropharmacological, and epigenomic anomalies mirror fundamental etiological aspects of neurodevelopmental disorders including ADHD, autism, and schizophrenia. Expanding this line of research, the current study profiled the multigenerational neurotrophic and neuroendocrine consequences of DNE. Results reveal impaired proBDNF proteolysis as indicated by proBDNF-BDNF imbalance, downregulation of the proBDNF processing enzyme furin, atypical glucocorticoid receptor (GR) activity as implied by decreased relative nuclear GR localization, and deficient basal plasma corticosterone (CORT) levels in adolescent DNE offspring and grandoffspring. Collectively, these data recapitulate the BDNF deficits and HPA axis dysregulation characteristic of neurodevelopmental disorders such as ADHD, autism, and schizophrenia as well as the children of maternal smokers. Notably, as BDNF is a quintessential mediator of neurodevelopment, our prior findings of multigenerational DNE-induced behavioral and neuropharmacological abnormalities may stem from neurodevelopmental insults conferred by the proBDNF-BDNF imbalance detected in DNE mice. Similarly, our findings of multigenerational GR hypoactivity may contribute to the increased risk-taking behaviors and aberrant circadian rhythmicity of home cage activity that we previously documented in first- and second-generation DNE mice.
Collapse
Affiliation(s)
- Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado, Boulder, United States; Department of Integrative Physiology, University of Colorado, Boulder, United States.
| | - Heidi C O'Neill
- Institute for Behavioral Genetics, University of Colorado, Boulder, United States
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, United States; Department of Integrative Physiology, University of Colorado, Boulder, United States
| |
Collapse
|
32
|
Houlton J, Abumaria N, Hinkley SFR, Clarkson AN. Therapeutic Potential of Neurotrophins for Repair After Brain Injury: A Helping Hand From Biomaterials. Front Neurosci 2019; 13:790. [PMID: 31427916 PMCID: PMC6688532 DOI: 10.3389/fnins.2019.00790] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022] Open
Abstract
Stroke remains the leading cause of long-term disability with limited options available to aid in recovery. Significant effort has been made to try and minimize neuronal damage following stroke with use of neuroprotective agents, however, these treatments have yet to show clinical efficacy. Regenerative interventions have since become of huge interest as they provide the potential to restore damaged neural tissue without being limited by a narrow therapeutic window. Neurotrophins, such as brain-derived neurotrophic factor (BDNF), and their high affinity receptors are actively produced throughout the brain and are involved in regulating neuronal activity and normal day-to-day function. Furthermore, neurotrophins are known to play a significant role in both protection and recovery of function following neurodegenerative diseases such as stroke and traumatic brain injury (TBI). Unfortunately, exogenous administration of these neurotrophins is limited by a lack of blood-brain-barrier (BBB) permeability, poor half-life, and rapid degradation. Therefore, we have focused this review on approaches that provide a direct and sustained neurotrophic support using pharmacological therapies and mimetics, physical activity, and potential drug delivery systems, including discussion around advantages and limitations for use of each of these systems. Finally, we discuss future directions of biomaterial drug-delivery systems, including the incorporation of heparan sulfate (HS) in conjunction with neurotrophin-based interventions.
Collapse
Affiliation(s)
- Josh Houlton
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai, China
- Department of Laboratory Animal Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Simon F. R. Hinkley
- The Ferrier Research Institute, Victoria University of Wellington, Petone, New Zealand
| | - Andrew N. Clarkson
- Brain Health Research Centre, Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
33
|
Upregulation of proBDNF in the Mesenteric Lymph Nodes in Septic Mice. Neurotox Res 2019; 36:540-550. [PMID: 31278527 DOI: 10.1007/s12640-019-00081-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 01/27/2023]
Abstract
The immune status in the lymphatic system, especially mesenteric lymph nodes (MLNs), is critical to regulate the septic shock. Brain-derived neurotrophic factor (BDNF) in the enteric system has been reported to regulate enteric immunity. However, the role of its precursor, proBDNF, in the immune status of MLNs under sepsis condition is still unclear. This study aimed to characterize the expression pattern of proBDNF in MLNs after lipopolysaccharide (LPS) stimulation, and to investigate the association of pathogenesis of sepsis. LPS (20 mg/kg) was intraperitoneally injected to induce sepsis in mice. Survival curve analysis, routine blood tests, and liver and kidney function tests were performed to evaluate the severity of sepsis. QPCR and histological staining were performed to assess the mRNA levels of proinflammatory cytokines and degree of immune-inflammatory response in the MLNs. Furthermore, Western blotting, flow cytometry, and immunofluorescence were performed to examine the key molecules expression of proBDNF signaling. Intraperitoneal LPS injection significantly decreased the number of lymphocytes in blood but increased the number of T lymphocytes in MLNs. Serum alanine transaminase, aspartate transaminase, and blood urea nitrogen levels were increased in LPS-challenged mice compared to control mice. LPS administration upregulated proinflammatory cytokine gene expression and induced histological changes in the MLNs. LPS injection increased BDNF, proBDNF, and its receptor pan neutrophin receptor 75 (p75NTR) expression in MLNs. The increased proBDNF was mainly localized on CD3+ and CD4+ T cells in the medulla of MLNs. LPS-induced sepsis upregulated proBDNF expression in medulla T cells of MLNs. ProBDNF upregulation may be involved in the pathogenesis of septic shock.
Collapse
|
34
|
Baho E, Chattopadhyaya B, Lavertu-Jolin M, Mazziotti R, Awad PN, Chehrazi P, Groleau M, Jahannault-Talignani C, Vaucher E, Ango F, Pizzorusso T, Baroncelli L, Di Cristo G. p75 Neurotrophin Receptor Activation Regulates the Timing of the Maturation of Cortical Parvalbumin Interneuron Connectivity and Promotes Juvenile-like Plasticity in Adult Visual Cortex. J Neurosci 2019; 39:4489-4510. [PMID: 30936240 PMCID: PMC6554620 DOI: 10.1523/jneurosci.2881-18.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/22/2019] [Accepted: 03/14/2019] [Indexed: 01/09/2023] Open
Abstract
By virtue of their extensive axonal arborization and perisomatic synaptic targeting, cortical inhibitory parvalbumin (PV) cells strongly regulate principal cell output and plasticity and modulate experience-dependent refinement of cortical circuits during development. An interesting aspect of PV cell connectivity is its prolonged maturation time course, which is completed only by end of adolescence. The p75 neurotrophin receptor (p75NTR) regulates numerous cellular functions; however, its role on cortical circuit development and plasticity remains elusive, mainly because localizing p75NTR expression with cellular and temporal resolution has been challenging. By using RNAscope and a modified version of the proximity ligation assay, we found that p75NTR expression in PV cells decreases between the second and fourth postnatal week, at a time when PV cell synapse numbers increase dramatically. Conditional knockout of p75NTR in single PV neurons in vitro and in PV cell networks in vivo causes precocious formation of PV cell perisomatic innervation and perineural nets around PV cell somata, therefore suggesting that p75NTR expression modulates the timing of maturation of PV cell connectivity in the adolescent cortex. Remarkably, we found that PV cells still express p75NTR in adult mouse cortex of both sexes and that its activation is sufficient to destabilize PV cell connectivity and to restore cortical plasticity following monocular deprivation in vivo Together, our results show that p75NTR activation dynamically regulates PV cell connectivity, and represent a novel tool to foster brain plasticity in adults.SIGNIFICANCE STATEMENT In the cortex, inhibitory, GABA-releasing neurons control the output and plasticity of excitatory neurons. Within this diverse group, parvalbumin-expressing (PV) cells form the larger inhibitory system. PV cell connectivity develops slowly, reaching maturity only at the end of adolescence; however, the mechanisms controlling the timing of its maturation are not well understood. We discovered that the expression of the neurotrophin receptor p75NTR in PV cells inhibits the maturation of their connectivity in a cell-autonomous fashion, both in vitro and in vivo, and that p75NTR activation in adult PV cells promotes their remodeling and restores cortical plasticity. These results reveal a new p75NTR function in the regulation of the time course of PV cell maturation and in limiting cortical plasticity.
Collapse
Affiliation(s)
- Elie Baho
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Bidisha Chattopadhyaya
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Marisol Lavertu-Jolin
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Raffaele Mazziotti
- Institute of Neuroscience Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
| | - Patricia N Awad
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Pegah Chehrazi
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Marianne Groleau
- École d'Optométrie, Université de Montréal, Montréal, Québec H3T 1P1, Canada
| | - Celine Jahannault-Talignani
- Institut de Génomique Fonctionnelle, université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé Et de la Recherche Médicale, 34090 Montpellier, France, and
| | - Elvire Vaucher
- École d'Optométrie, Université de Montréal, Montréal, Québec H3T 1P1, Canada
| | - Fabrice Ango
- Institut de Génomique Fonctionnelle, université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé Et de la Recherche Médicale, 34090 Montpellier, France, and
| | - Tommaso Pizzorusso
- Institute of Neuroscience Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research and Child Health Neurofarba, University of Florence, 50139 Firenze, Italy
| | - Laura Baroncelli
- Institute of Neuroscience Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
| | - Graziella Di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada,
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| |
Collapse
|
35
|
Panax notoginsenoside Rb1 Restores the Neurotrophic Imbalance Following Photothrombotic Stroke in Rats. Neurotox Res 2019; 36:441-451. [DOI: 10.1007/s12640-019-00058-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/27/2019] [Accepted: 05/02/2019] [Indexed: 01/13/2023]
|
36
|
The Long-Term Effects of Ethanol and Corticosterone on the Mood-Related Behaviours and the Balance Between Mature BDNF and proBDNF in Mice. J Mol Neurosci 2019; 69:60-68. [PMID: 31127538 DOI: 10.1007/s12031-019-01328-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 04/16/2019] [Indexed: 12/21/2022]
Abstract
In this study, we aimed to establish the effects of chronic corticosterone (CORT) and ethanol administration on mood-related behaviour and the levels of mature brain-derived neurotrophic factor (mBDNF) and its precursor protein proBDNF in mice. C57BL6 male and female mice received drinking water (n = 22), 1% ethanol in drinking water (n = 16) or 100 μg/ml corticosterone in drinking water (containing 1% ethanol, n = 18) for 4.5 weeks. At the end of experimental protocol, the open field test (OFT) and elevated plus maze test were performed. Brain and adrenal tissues were collected and mBDNF and proBDNF were measured by ELISA assays. We found that the mice fed with corticosterone and ethanol developed anxiety-like behaviours as evidenced by reduced time in the central zone in the OFT compared with the control group. Both proBDNF and mBDNF were significantly decreased in the corticosterone and ethanol groups compared with the control group in the prefrontal cortex, hippocampus, hypothalamus and adrenal. The ratio of proBDNF/mBDNF in prefrontal cortex in the corticosterone group was increased compared with the ethanol group. Our data suggest that the ratio of proBDNF/mBDNF is differentially regulated in different tissues. Ethanol and corticosterone downregulate both mBDNF and proBDNF and alter the balance of proBDNF/mBDNF in some tissues. In conclusion, the ethanol and corticosterone may cause abnormal regulation of mBDNF and proBDNF which may lead to mood disorders.
Collapse
|
37
|
Marton S, González B, Rodríguez-Bottero S, Miquel E, Martínez-Palma L, Pazos M, Prieto JP, Rodríguez P, Sames D, Seoane G, Scorza C, Cassina P, Carrera I. Ibogaine Administration Modifies GDNF and BDNF Expression in Brain Regions Involved in Mesocorticolimbic and Nigral Dopaminergic Circuits. Front Pharmacol 2019; 10:193. [PMID: 30890941 PMCID: PMC6411846 DOI: 10.3389/fphar.2019.00193] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/14/2019] [Indexed: 01/07/2023] Open
Abstract
Ibogaine is an atypical psychedelic alkaloid, which has been subject of research due to its reported ability to attenuate drug-seeking behavior. Recent work has suggested that ibogaine effects on alcohol self-administration in rats are related to the release of Glial cell Derived Neurotrophic Factor (GDNF) in the Ventral Tegmental Area (VTA), a mesencephalic region which hosts the soma of dopaminergic neurons. Although previous reports have shown ibogaine’s ability to induce GDNF expression in rat midbrain, there are no studies addressing its effect on the expression of GDNF and other neurotrophic factors (NFs) such as Brain Derived Neurotrophic Factor (BDNF) or Nerve Growth Factor (NGF) in distinct brain regions containing dopaminergic neurons. In this work, we examined the effect of ibogaine acute administration on the expression of these NFs in the VTA, Prefrontal Cortex (PFC), Nucleus Accumbens (NAcc) and the Substantia Nigra (SN). Rats were i.p. treated with ibogaine 20 mg/kg (I20), 40 mg/kg (I40) or vehicle, and NFs expression was analyzed after 3 and 24 h. At 24 h an increase of the expression of the NFs transcripts was observed in a site and dose dependent manner. Only for I40, GDNF was selectively upregulated in the VTA and SN. Both doses elicited a large increase in the expression of BDNF transcripts in the NAcc, SN and PFC, while in the VTA a significant effect was found only for I40. Finally, NGF mRNA was upregulated in all regions after I40, while I20 showed a selective upregulation in PFC and VTA. Regarding protein levels, an increase of GDNF was observed in the VTA only for I40 but no significant increase for BDNF was found in all the studied areas. Interestingly, an increase of proBDNF was detected in the NAcc for both doses. These results show for the first time a selective increase of GDNF specifically in the VTA for I40 but not for I20 after 24 h of administration, which agrees with the effective dose found in previous self-administration studies in rodents. Further research is needed to understand the contribution of these changes to ibogaine’s ability to attenuate drug-seeking behavior.
Collapse
Affiliation(s)
- Soledad Marton
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Bruno González
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Rodríguez-Bottero
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Ernesto Miquel
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laura Martínez-Palma
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mariana Pazos
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - José Pedro Prieto
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Paola Rodríguez
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY, United States
| | - Gustavo Seoane
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Scorza
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Patricia Cassina
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Ignacio Carrera
- Laboratorio de Síntesis Orgánica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
38
|
Montroull LE, Danelon V, Cragnolini AB, Mascó DH. Loss of TrkB Signaling Due to Status Epilepticus Induces a proBDNF-Dependent Cell Death. Front Cell Neurosci 2019; 13:4. [PMID: 30800056 PMCID: PMC6375841 DOI: 10.3389/fncel.2019.00004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/08/2019] [Indexed: 01/25/2023] Open
Abstract
Neurotrophins (NTs) are secretory proteins that bind to target receptors and influence many cellular functions, such as cell survival and cell death in neurons. The mammalian NT brain-derived neurotrophic factor (matBDNF) is the C-terminal mature form released by cleavage from the proBDNF precursor. The binding of matBDNF to the tyrosine kinase receptor B (TrkB) activates different signaling cascades and leads to neuron survival and plasticity, while the interaction of proBDNF with the p75 NT receptor (p75NTR)/sortilin receptor complex has been highly involved in apoptosis. Many studies have demonstrated that prolonged seizures such as status epilepticus (SE) induce changes in the expression of NT, pro-NT, and their receptors. We have previously described that the blockage of both matBDNF and proBDNF signaling reduces neuronal death after SE in vivo (Unsain et al., 2008). We used an in vitro model as well as an in vivo model of SE to determine the specific role of TrkB and proBDNF signaling during neuronal cell death. We found that the matBDNF sequestering molecule TrkB-Fc induced an increase in neuronal death in both models of SE, and it also prevented a decrease in TrkB levels. Moreover, SE triggered the interaction between proBDNF and p75NTR, which was not altered by sequestering matBDNF. The intra-hippocampal administration of TrkB-Fc, combined with an antibody against proBDNF, prevented neuronal degeneration. In addition, we demonstrated that proBDNF binding to p75NTR exacerbates neuronal death when matBDNF signaling is impaired through TrkB. Our results indicated that both the mature and the precursor forms of BDNF may have opposite effects depending on the scenario in which they function and the signaling pathways they activate.
Collapse
Affiliation(s)
- Laura Ester Montroull
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Víctor Danelon
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea Beatriz Cragnolini
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Daniel Hugo Mascó
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba; Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
39
|
Barfield ET, Gourley SL. Prefrontal cortical trkB, glucocorticoids, and their interactions in stress and developmental contexts. Neurosci Biobehav Rev 2018; 95:535-558. [PMID: 30477984 PMCID: PMC6392187 DOI: 10.1016/j.neubiorev.2018.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/14/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
The tropomyosin/tyrosine receptor kinase B (trkB) and glucocorticoid receptor (GR) regulate neuron structure and function and the hormonal stress response. Meanwhile, disruption of trkB and GR activity (e.g., by chronic stress) can perturb neuronal morphology in cortico-limbic regions implicated in stressor-related illnesses like depression. Further, several of the short- and long-term neurobehavioral consequences of stress depend on the developmental timing and context of stressor exposure. We review how the levels and activities of trkB and GR in the prefrontal cortex (PFC) change during development, interact, are modulated by stress, and are implicated in depression. We review evidence that trkB- and GR-mediated signaling events impact the density and morphology of dendritic spines, the primary sites of excitatory synapses in the brain, highlighting effects in adolescents when possible. Finally, we review the role of neurotrophin and glucocorticoid systems in stress-related metaplasticity. We argue that better understanding the long-term effects of developmental stressors on PFC trkB, GR, and related factors may yield insights into risk for chronic, remitting depression and related neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Elizabeth T Barfield
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| | - Shannon L Gourley
- Department of Pediatrics, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Graduate Program in Neuroscience, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Department of Psychiatry and Behavioral Sciences, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA; Molecular and Systems Pharmacology Program, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
40
|
Angelucci F, Čechová K, Průša R, Hort J. Amyloid beta soluble forms and plasminogen activation system in Alzheimer's disease: Consequences on extracellular maturation of brain-derived neurotrophic factor and therapeutic implications. CNS Neurosci Ther 2018; 25:303-313. [PMID: 30403004 PMCID: PMC6488905 DOI: 10.1111/cns.13082] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/18/2022] Open
Abstract
Soluble oligomeric forms of amyloid beta (Aβ) play an important role in causing the cognitive deficits in Alzheimer’s disease (AD) by targeting and disrupting synaptic pathways. Thus, the present research is directed toward identifying the neuronal pathways targeted by soluble forms and, accordingly, develops alternative therapeutic strategies. The neurotrophin brain‐derived neurotrophic factor (BDNF) is synthesized as a precursor (pro‐BDNF) which is cleaved extracellularly by plasmin to release the mature form. The conversion from pro‐BDNF to BDNF is an important process that regulates neuronal activity and memory processes. Plasmin‐dependent maturation of BDNF in the brain is regulated by plasminogen activator inhibitor‐1 (PAI‐1), the natural inhibitor of tissue‐type plasminogen activator (tPA). Therefore, tPA/PAI‐1 system represents an important regulator of extracellular BDNF/pro‐BDNF ratio. In this review, we summarize the data on the components of the plasminogen activation system and on BDNF in AD. Moreover, we will hypothesize a possible pathogenic mechanism caused by soluble Aβ forms based on the effects on tPA/PAI‐1 system and on the consequence of an altered conversion from pro‐BDNF to the mature BDNF in the brain of AD patients. Translation into clinic may include a better characterization of the disease stage and future direction on therapeutic targets.
Collapse
Affiliation(s)
- Francesco Angelucci
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Kateřina Čechová
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Richard Průša
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jakub Hort
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
41
|
Notaras M, van den Buuse M. Brain-Derived Neurotrophic Factor (BDNF): Novel Insights into Regulation and Genetic Variation. Neuroscientist 2018; 25:434-454. [DOI: 10.1177/1073858418810142] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since its discovery, brain-derived neurotrophic factor (BDNF) has spawned a literature that now spans 35 years of research. While all neurotrophins share considerable overlap in sequence homology and their processing, BDNF has become the most widely studied neurotrophin because of its broad roles in brain homeostasis, health, and disease. Although research on BDNF has produced thousands of articles, there remain numerous long-standing questions on aspects of BDNF molecular biology and signaling. Here we provide a comprehensive review, including both a historical narrative and a forward-looking perspective on advances in the actions of BDNF within the brain. We specifically review BDNF’s gene structure, peptide composition (including domains, posttranslational modifications and putative motif sites), mechanisms of transport, signaling pathway recruitment, and other recent developments including the functional effects of genetic variation and the discovery of a new BDNF prodomain ligand. This body of knowledge illustrates a highly conserved and complex role for BDNF within the brain, that promotes the idea that the neurotrophin biology of BDNF is diverse and that any disease involvement is likely to be equally multifarious.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
The Structure of the Pro-domain of Mouse proNGF in Contact with the NGF Domain. Structure 2018; 27:78-89.e3. [PMID: 30393051 DOI: 10.1016/j.str.2018.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/14/2018] [Accepted: 09/25/2018] [Indexed: 01/19/2023]
Abstract
Nerve growth factor (NGF) is an important neurotrophic factor involved in the regulation of cell differentiation and survival of target neurons. Expressed as a proNGF precursor, NGF is matured by furin-mediated protease cleavage. Increasing evidence suggests that NGF and proNGF have distinct functional roles. While the structure of mature NGF is available, little is known about that of the pro-domain because of its dynamical structural features. We exploited an ad hoc hybrid strategy based on nuclear magnetic resonance and modeling validated by small-angle X-ray scattering to gain novel insights on the pro-domain, both in isolation and in the context of proNGF. We show that the isolated pro-domain is intrinsically unstructured but forms transient intramolecular contacts with mature NGF and has per se the ability to induce growth cone collapse, indicating functional independence. Our data represent an important step toward the structural and functional characterization of the properties of proNGF.
Collapse
|
43
|
Chong PN, Sangu M, Huat TJ, Reza F, Begum T, Yusoff AAM, Jaafar H, Abdullah JM. Trkb-IP3 Pathway Mediating Neuroprotection in Rat Hippocampal Neuronal Cell Culture Following Induction of Kainic Acid. Malays J Med Sci 2018; 25:28-45. [PMID: 30914877 PMCID: PMC6422567 DOI: 10.21315/mjms2018.25.6.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/17/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Following brain injury, development of hippocampal sclerosis often led to the temporal lobe epilepsy which is sometimes resistant to common anti-epileptic drugs. Cellular and molecular changes underlying epileptogenesis in animal models were studied, however, the underlying mechanisms of kainic acid (KA) mediated neuronal damage in rat hippocampal neuron cell culture alone has not been elucidated yet. METHODS Embryonic day 18 (E-18) rat hippocampus neurons were cultured with poly-L-lysine coated glass coverslips. Following optimisation, KA (0.5 μM), a chemoconvulsant agent, was administered at three different time-points (30, 60 and 90 min) to induce seizure in rat hippocampal neuronal cell culture. We examined cell viability, neurite outgrowth density and immunoreactivity of the hippocampus neuron culture by measuring brain derived neurotrophic factor (BDNF), γ-amino butyric acid A (GABAA) subunit α-1 (GABRA1), tyrosine receptor kinase B (TrkB), and inositol trisphosphate receptor (IP3R/IP3) levels. RESULTS The results revealed significantly decreased and increased immunoreactivity changes in TrkB (a BDNF receptor) and IP3R, respectively, at 60 min time point. CONCLUSION The current findings suggest that TrkB and IP3 could have a neuroprotective role which could be a potential pharmacological target for anti-epilepsy drugs.
Collapse
Affiliation(s)
- Pei Nei Chong
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Muthuraju Sangu
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Tee Jong Huat
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Faruque Reza
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Tahamina Begum
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Abdul Aziz Mohamed Yusoff
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Hasnan Jaafar
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Jafri Malin Abdullah
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
- Centre for Neuroscience Services and Research, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
44
|
Hippocampal proBDNF facilitates place learning strategy associated with neural activity in rats. Brain Struct Funct 2018; 223:4099-4113. [PMID: 30151608 DOI: 10.1007/s00429-018-1742-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/23/2018] [Indexed: 12/30/2022]
Abstract
Mature brain-derived neurotrophic factor has been shown to have a promotive effect on synaptic plasticity and spatial memory. The precursor of BDNF (proBDNF) has emerged as a protein against its mature form. However, it is unknown whether and how proBDNF regulates neural excitability and spatial behavior. Through infusion of cleavage-resistant proBDNF or its antibody into HPC, we sought evidence for the influences by employing multiple behavioral tests and recording hippocampal single-unit activity. Our behavioral findings showed that proBDNF induced beneficial effects on spatial learning by facilitating the use of the place strategy and inhibiting the response strategy, including (1) using more place search strategies but less response strategies, and (2) increasing the number of rats in choosing place strategies but not response strategies. Intriguingly, infusion of an anti-proBDNF antibody did not affect rats' training process but rendered the adaption to learning reversal training more difficult, indicating deficits in choosing the proper learning strategy. The training-induced increase in proBDNF promoted the firing rate of pyramidal neurons but not fast-spiking (FS) interneurons. Importantly, endogenous proBDNF facilitated the neural correlate of spatial, but not response, learning behavior. However, the anti-proBDNF antibody effectively reversed the strategy preference and inhibited neural activity. We herein propose that proBDNF exerts pivotal effects on neural excitability and the use of cognitive strategies to facilitate the spatial learning process.
Collapse
|
45
|
Liu S, Guo W, Zhou H, Tang L, Feng S, Zhong JH, Zhou XF. proBDNF inhibits the proliferation and migration of OLN‑93 oligodendrocytes. Mol Med Rep 2018; 18:3809-3817. [PMID: 30132570 PMCID: PMC6131500 DOI: 10.3892/mmr.2018.9407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 05/03/2018] [Indexed: 01/11/2023] Open
Abstract
In contrast with mature brain-derived neurotrophic factor (mBDNF), proBDNF induces cell apoptosis. However, the function of proBDNF in oligodendrocytes remains unclear. In the present study, the OLN-93 oligodendroglia cell line was utilized as an in vitro model to analyse the functions of proBDNF in oligodendroglia. p75NTR, sortilin and proBDNF were expressed in cultured OLN-93 cells. It was indicated that proBDNF inhibited OLN-93 cell proliferation in a dose-dependent manner as determined using the MTT assay and BrdU staining. Furthermore, proBDNF suppressed the migration of OLN-93 cells as demonstrated using the scratch assay. proBDNF also decreased cell viability and promoted apoptosis as indicated by activated cysteine-aspartic acid protease-3 (caspase-3) immunocytochemistry. Notably, anti-proBDNF treatment neutralized the effect of proBDNF and resulted in increased cell proliferation and migration and decreased apoptosis. However, these effects were not observed in the presence of recombinant p75NTR extracellular domain-human FC fusion protein and p75NTR antibody, indicating that proBDNF imparts its inhibitory effects on oligodendrocytes through the p75NTR signal pathway.
Collapse
Affiliation(s)
- Shen Liu
- School of Pharmacy and Medical Science and Sansom Institute, Division of Health Sciences, University of South Australia, Adelaide SA5000, Australia
| | - Wei Guo
- Orthopaedic Department of Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hengxing Zhou
- Orthopaedic Department of Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Liang Tang
- Orthopaedic Department of Tianjin Haihe Hospital, Tianjin 300000, P.R. China
| | - Shiqing Feng
- Orthopaedic Department of Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jin-Hua Zhong
- School of Pharmacy and Medical Science and Sansom Institute, Division of Health Sciences, University of South Australia, Adelaide SA5000, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Science and Sansom Institute, Division of Health Sciences, University of South Australia, Adelaide SA5000, Australia
| |
Collapse
|
46
|
Giza JI, Kim J, Meyer HC, Anastasia A, Dincheva I, Zheng CI, Lopez K, Bains H, Yang J, Bracken C, Liston C, Jing D, Hempstead BL, Lee FS. The BDNF Val66Met Prodomain Disassembles Dendritic Spines Altering Fear Extinction Circuitry and Behavior. Neuron 2018; 99:163-178.e6. [PMID: 29909994 DOI: 10.1016/j.neuron.2018.05.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/18/2018] [Accepted: 05/15/2018] [Indexed: 11/16/2022]
Abstract
A human variant in the BDNF gene (Val66Met; rs6265) is associated with impaired fear extinction. Using super-resolution imaging, we demonstrate that the BDNF Met prodomain disassembles dendritic spines and eliminates synapses in hippocampal neurons. In vivo, ventral CA1 (vCA1) hippocampal neurons undergo similar morphological changes dependent on their transient co-expression of a SorCS2/p75NTR receptor complex during peri-adolescence. BDNF Met prodomain infusion into the vCA1 during this developmental time frame reduces dendritic spine density and prelimbic (PL) projections, impairing cued fear extinction. Adolescent BdnfMet/Met mice display similar spine and PL innervation deficits. Using fiber photometry, we found that, in wild-type mice, vCA1 neurons projecting to the PL encode extinction by enhancing neural activity in threat anticipation and rapidly subsiding their response. This adaptation is absent in BDNFMet/Met mice. We conclude that the BDNF Met prodomain renders vCA1-PL projection neurons underdeveloped, preventing their capacity for subsequent circuit modulation necessary for fear extinction. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Joanna I Giza
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jihye Kim
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heidi C Meyer
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Agustin Anastasia
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Iva Dincheva
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Crystal I Zheng
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Katherine Lopez
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrietta Bains
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Key Laboratory of Shaanxi Province Department for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Clay Bracken
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Conor Liston
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Deqiang Jing
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA; Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
47
|
Ohja K, Gozal E, Fahnestock M, Cai L, Cai J, Freedman JH, Switala A, El-Baz A, Barnes GN. Neuroimmunologic and Neurotrophic Interactions in Autism Spectrum Disorders: Relationship to Neuroinflammation. Neuromolecular Med 2018; 20:161-173. [PMID: 29691724 PMCID: PMC5942347 DOI: 10.1007/s12017-018-8488-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022]
Abstract
Autism spectrum disorders (ASD) are the most prevalent set of pediatric neurobiological disorders. The etiology of ASD has both genetic and environmental components including possible dysfunction of the immune system. The relationship of the immune system to aberrant neural circuitry output in the form of altered behaviors and communication characterized by ASD is unknown. Dysregulation of neurotrophins such as BDNF and their signaling pathways have been implicated in ASD. While abnormal cortical formation and autistic behaviors in mouse models of immune activation have been described, no one theory has been described to link activation of the immune system to specific brain signaling pathways aberrant in ASD. In this paper we explore the relationship between neurotrophin signaling, the immune system and ASD. To this effect we hypothesize that an interplay of dysregulated immune system, synaptogenic growth factors and their signaling pathways contribute to the development of ASD phenotypes.
Collapse
Affiliation(s)
- Kshama Ohja
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Evelyne Gozal
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Lu Cai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jun Cai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jonathan H Freedman
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Andy Switala
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Gregory Neal Barnes
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA. .,Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA. .,Spafford Ackerly Chair in Child and Adolescent Psychiatry, University of Louisville Autism Center, 1405 East Burnett Avenue, Louisville, KY, 40217, USA.
| |
Collapse
|
48
|
Pearn ML, Schilling JM, Jian M, Egawa J, Wu C, Mandyam CD, Fannon-Pavlich MJ, Nguyen U, Bertoglio J, Kodama M, Mahata SK, DerMardirossian C, Lemkuil BP, Han R, Mobley WC, Patel HH, Patel PM, Head BP. Inhibition of RhoA reduces propofol-mediated growth cone collapse, axonal transport impairment, loss of synaptic connectivity, and behavioural deficits. Br J Anaesth 2018; 120:745-760. [PMID: 29576115 PMCID: PMC6200100 DOI: 10.1016/j.bja.2017.12.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/28/2017] [Accepted: 12/26/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Exposure of the developing brain to propofol results in cognitive deficits. Recent data suggest that inhibition of neuronal apoptosis does not prevent cognitive defects, suggesting mechanisms other than neuronal apoptosis play a role in anaesthetic neurotoxicity. Proper neuronal growth during development is dependent upon growth cone morphology and axonal transport. Propofol modulates actin dynamics in developing neurones, causes RhoA-dependent depolymerisation of actin, and reduces dendritic spines and synapses. We hypothesised that RhoA inhibition prevents synaptic loss and subsequent cognitive deficits. The present study tested whether RhoA inhibition with the botulinum toxin C3 (TAT-C3) prevents propofol-induced synapse and neurite loss, and preserves cognitive function. METHODS RhoA activation, growth cone morphology, and axonal transport were measured in neonatal rat neurones (5-7 days in vitro) exposed to propofol. Synapse counts (electron microscopy), dendritic arborisation (Golgi-Cox), and network connectivity were measured in mice (age 28 days) previously exposed to propofol at postnatal day 5-7. Memory was assessed in adult mice (age 3 months) previously exposed to propofol at postnatal day 5-7. RESULTS Propofol increased RhoA activation, collapsed growth cones, and impaired retrograde axonal transport of quantum dot-labelled brain-derived neurotrophic factor, all of which were prevented with TAT-C3. Adult mice previously treated with propofol had decreased numbers of total hippocampal synapses and presynaptic vesicles, reduced hippocampal dendritic arborisation, and infrapyramidal mossy fibres. These mice also exhibited decreased hippocampal-dependent contextual fear memory recall. All anatomical and behavioural changes were prevented with TAT-C3 pre-treatment. CONCLUSION Inhibition of RhoA prevents propofol-mediated hippocampal neurotoxicity and associated cognitive deficits.
Collapse
Affiliation(s)
- M L Pearn
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - J M Schilling
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - M Jian
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA; Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - J Egawa
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - C Wu
- Department of Neurosciences, UCSD, San Diego, CA, USA
| | - C D Mandyam
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - M J Fannon-Pavlich
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - U Nguyen
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - J Bertoglio
- INSERM U749, Institut Gustave Roussy, Universite Paris-sud, Paris, France
| | - M Kodama
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA; Metabolic Physiology and Ultrastructural Biology Laboratory, UCSD, San Diego CA, USA; Department of Anesthesiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - S K Mahata
- Metabolic Physiology and Ultrastructural Biology Laboratory, UCSD, San Diego CA, USA
| | - C DerMardirossian
- Department of Immunology and Microbial Sciences, TSRI, La Jolla, CA, USA; Department of Cell and Molecular Biology, TSRI, La Jolla, CA, USA
| | - B P Lemkuil
- Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - R Han
- Department of Anesthesiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - W C Mobley
- Department of Neurosciences, UCSD, San Diego, CA, USA
| | - H H Patel
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - P M Patel
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA
| | - B P Head
- Veterans Affairs San Diego Healthcare System, UCSD, San Diego CA, USA; Department of Anesthesiology, UCSD, San Diego, CA, USA.
| |
Collapse
|
49
|
Abstract
Alcohol dependence is a worldwide problem with a great social and economic burden in many countries. A number of studies have suggested that BDNF (mature BDNF) and its precursor (proBDNF) play important roles in the alcohol dependence. However, what roles of the mBDNF/proBDNF pathways play during the pathological process of alcohol dependence are not clearly understood. In our clinical study, peripheral blood was sampled from 30 male patients with alcohol dependence and 50 healthy males (as control). The protein levels of proBDNF, p75NTR, sortilin, mBDNF, TrkB and mRNA levels of BDNF, p75NTR, sortilin, and TrkB were detected in the peripheral blood in our study. We found that the protein levels of proBDNF and p75NTR were increased, but not the sortilin protein level; while the TrkB protein level was decreased in the alcohol dependence patients compared with healthy controls. Moreover, the mRNA levels of p75NTR and sortilin from the lymphocytes were slightly increased; while BDNF and TrkB were significantly decreased. The ELISA results of mBDNF and TrkB were declined in the alcohol dependence group. The levels of mBDNF and TrkB were negatively correlated with the average amount of daily ethanol consumption, and the levels of proBDNF, p75NTR and sortilin were positively correlated with the average amount of ethanol consumption per day. The ratio of proBDNF to mBDNF was altered in alcohol dependence patients. The balance between the proBDNF/p75NTR and mBDNF/TrkB signalling pathways appeared dysregulated in alcohol dependence. Our results suggested that both pathways may participate in the complex processes of alcohol dependence.
Collapse
|
50
|
Saadipour K, Mañucat-Tan NB, Lim Y, Keating DJ, Smith KS, Zhong JH, Liao H, Bobrovskaya L, Wang YJ, Chao MV, Zhou XF. p75 neurotrophin receptor interacts with and promotes BACE1 localization in endosomes aggravating amyloidogenesis. J Neurochem 2018; 144:302-317. [PMID: 28869759 DOI: 10.1111/jnc.14206] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/17/2017] [Accepted: 08/29/2017] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive deposition of amyloid beta (Aβ) and dysregulation of neurotrophic signaling, causing synaptic dysfunction, loss of memory, and cell death. The expression of p75 neurotrophin receptor is elevated in the brain of AD patients, suggesting its involvement in this disease. However, the exact mechanism of its action is not yet clear. Here, we show that p75 interacts with beta-site amyloid precursor protein cleaving enzyme-1 (BACE1), and this interaction is enhanced in the presence of Aβ. Our results suggest that the colocalization of BACE1 and amyloid precursor protein (APP) is increased in the presence of both Aβ and p75 in cortical neurons. In addition, the localization of APP and BACE1 in early endosomes is increased in the presence of Aβ and p75. An increased phosphorylation of APP-Thr668 and BACE1-Ser498 by c-Jun N-terminal kinase (JNK) in the presence of Aβ and p75 could be responsible for this localization. In conclusion, our study proposes a potential involvement in amyloidogenesis for p75, which may represent a future therapeutic target for AD. Cover Image for this Issue: doi. 10.1111/jnc.14163.
Collapse
Affiliation(s)
- Khalil Saadipour
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia
- Department of Human Physiology, Centre for Neuroscience, Flinders University, Adelaide, South Australia
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Noralyn B Mañucat-Tan
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia
| | - Yoon Lim
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia
| | - Damien J Keating
- Department of Human Physiology, Centre for Neuroscience, Flinders University, Adelaide, South Australia
| | - Kevin S Smith
- Department of Human Physiology, Centre for Neuroscience, Flinders University, Adelaide, South Australia
| | - Jin-Hua Zhong
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia
| | - Hong Liao
- New Drug Screening Centre, China Pharmaceutical University, Nanjing, China
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia
| |
Collapse
|