1
|
El-Kadi RA, AbdelKader NF, Zaki HF, Kamel AS. Vilazodone Alleviates Neurogenesis-Induced Anxiety in the Chronic Unpredictable Mild Stress Female Rat Model: Role of Wnt/β-Catenin Signaling. Mol Neurobiol 2024; 61:9060-9077. [PMID: 38584231 PMCID: PMC11496359 DOI: 10.1007/s12035-024-04142-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
Defective β-catenin signaling is accompanied with compensatory neurogenesis process that may pave to anxiety. β-Catenin has a distinct role in alleviating anxiety in adolescence; however, it undergoes degradation by the degradation complex Axin and APC. Vilazodone (VZ) is a fast, effective antidepressant with SSRI activity and 5-HT1A partial agonism that amends somatic and/or psychic symptoms of anxiety. Yet, there is no data about anxiolytic effect of VZ on anxiety-related neurogenesis provoked by stress-reduced β-catenin signaling. Furthermore, females have specific susceptibility toward psychopathology. The aim of the present study is to uncover the molecular mechanism of VZ relative to Wnt/β-catenin signaling in female rats. Stress-induced anxiety was conducted by subjecting the rats to different stressful stimuli for 21 days. On the 15th day, stressed rats were treated with VZ(10 mg/kg, p.o.) alone or concomitant with the Wnt inhibitor: XAV939 (0.1 mg/kg, i.p.). Anxious rats showed low β-catenin level turned over by Axin-1 with unanticipated reduction of APC pursued with elevated protein levels of neurogenesis-stimulating proteins: c-Myc and pThr183-Erk likewise gene expressions of miR-17-5p and miR-18. Two weeks of VZ treatment showed anxiolytic effect figured by alleviation of hippocampal histological examination. VZ protected β-catenin signal via reduction in Axin-1 and elevation of APC conjugated with modulation of β-catenin downstream targets. The cytoplasmic β-catenin turnover by Axin-1 was restored by XAV939. Herein, VZ showed anti-anxiety effect, which may be in part through regaining the balance of the reduced β-catenin and its subsequent exaggerated response of p-Erk, c-Myc, Dicer-1, miR-17-5p, and miR-18.
Collapse
Affiliation(s)
- Rana A El-Kadi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo, 11562, Egypt
- Alexandria University Hospitals, Champollion Street, El-Khartoum Square, El Azareeta, Alexandria, 21131, Egypt
| | - Noha F AbdelKader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo, 11562, Egypt
| | - Hala F Zaki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo, 11562, Egypt
| | - Ahmed S Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo, 11562, Egypt.
| |
Collapse
|
2
|
Wei X, Browning JL, Olsen ML. Neuron and astrocyte specific 5mC and 5hmC signatures of BDNF's receptor, TrkB. Front Mol Neurosci 2024; 17:1463437. [PMID: 39268252 PMCID: PMC11390696 DOI: 10.3389/fnmol.2024.1463437] [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: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Brain derived neurotrophic factor (BDNF) is the most studied trophic factor in the central nervous system (CNS), and its role in the maturation of neurons, including synapse development and maintenance has been investigated intensely for over three decades. The primary receptor for BDNF is the tropomyosin receptor kinase B (TrkB), which is broadly expressed as two primary isoforms in the brain; the full length TrkB (TrkB.FL) receptor, expressed mainly in neurons and the truncated TrkB (TrkB.T1) receptor. We recently demonstrated that TrkB.T1 is predominately expressed in astrocytes, and appears critical for astrocyte morphological maturation. Given the critical role of BDNF/TrkB pathway in healthy brain development and mature CNS function, we aimed to identify molecular underpinnings of cell-type specific expression of each TrkB isoform. Using Nanopore sequencing which enables direct, long read sequencing of native DNA, we profiled DNA methylation patterns of the entire TrkB gene, Ntrk2, in both neurons and astrocytes. Here, we identified robust differences in cell-type specific isoform expression associated with significantly different methylation patterns of the Ntrk2 gene in each cell type. Notably, astrocytes demonstrated lower 5mC methylation, and higher 5hmC across the entire gene when compared to neurons, including differentially methylated sites (DMSs) found in regions flanking the unique TrkB.T1 protein coding sequence (CDS). These data suggest DNA methylation patterns may provide instruction for isoform specific TrkB expression across unique CNS cell types.
Collapse
Affiliation(s)
- Xiaoran Wei
- Biomedical and Veterinary Sciences Graduate Program, Virginia Tech, Blacksburg, VA, United States
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| | - Jack L. Browning
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
- Genetics, Bioinformatics and Computational Biology Graduate Program, Virginia Tech, Blacksburg, VA, United States
| | - Michelle L. Olsen
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
3
|
Liu K, Tronstad O, Flaws D, Churchill L, Jones AYM, Nakamura K, Fraser JF. From bedside to recovery: exercise therapy for prevention of post-intensive care syndrome. J Intensive Care 2024; 12:11. [PMID: 38424645 PMCID: PMC10902959 DOI: 10.1186/s40560-024-00724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/17/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND As advancements in critical care medicine continue to improve Intensive Care Unit (ICU) survival rates, clinical and research attention is urgently shifting toward improving the quality of survival. Post-Intensive Care Syndrome (PICS) is a complex constellation of physical, cognitive, and mental dysfunctions that severely impact patients' lives after hospital discharge. This review provides a comprehensive and multi-dimensional summary of the current evidence and practice of exercise therapy (ET) during and after an ICU admission to prevent and manage the various domains of PICS. The review aims to elucidate the evidence of the mechanisms and effects of ET in ICU rehabilitation and highlight that suboptimal clinical and functional outcomes of ICU patients is a growing public health concern that needs to be urgently addressed. MAIN BODY This review commences with a brief overview of the current relationship between PICS and ET, describing the latest research on this topic. It subsequently summarises the use of ET in ICU, hospital wards, and post-hospital discharge, illuminating the problematic transition between these settings. The following chapters focus on the effects of ET on physical, cognitive, and mental function, detailing the multi-faceted biological and pathophysiological mechanisms of dysfunctions and the benefits of ET in all three domains. This is followed by a chapter focusing on co-interventions and how to maximise and enhance the effect of ET, outlining practical strategies for how to optimise the effectiveness of ET. The review next describes several emerging technologies that have been introduced/suggested to augment and support the provision of ET during and after ICU admission. Lastly, the review discusses future research directions. CONCLUSION PICS is a growing global healthcare concern. This review aims to guide clinicians, researchers, policymakers, and healthcare providers in utilising ET as a therapeutic and preventive measure for patients during and after an ICU admission to address this problem. An improved understanding of the effectiveness of ET and the clinical and research gaps that needs to be urgently addressed will greatly assist clinicians in their efforts to rehabilitate ICU survivors, improving patients' quality of survival and helping them return to their normal lives after hospital discharge.
Collapse
Affiliation(s)
- Keibun Liu
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.
- Non-Profit Organization ICU Collaboration Network (ICON), Tokyo, Japan.
| | - Oystein Tronstad
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Physiotherapy Department, The Prince Charles Hospital, Brisbane, Australia
| | - Dylan Flaws
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Metro North Mental Health, Caboolture Hospital, Caboolture, Australia
- School of Clinical Science, Queensland University of Technology, Brisbane, Australia
| | - Luke Churchill
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Physiotherapy Department, The Prince Charles Hospital, Brisbane, Australia
- School of Health & Rehabilitation Sciences, The University of Queensland, Brisbane, Australia
| | - Alice Y M Jones
- School of Health & Rehabilitation Sciences, The University of Queensland, Brisbane, Australia
| | - Kensuke Nakamura
- Department of Critical Care Medicine, Yokohama City University Hospital, Kanagawa, Japan
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Chermside, QLD, 4032, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Queensland University of Technology, Brisbane, Australia
- St. Andrews War Memorial Hospital, Brisbane, Australia
| |
Collapse
|
4
|
Wang Y, Liang J, Xu B, Yang J, Wu Z, Cheng L. TrkB/BDNF signaling pathway and its small molecular agonists in CNS injury. Life Sci 2024; 336:122282. [PMID: 38008209 DOI: 10.1016/j.lfs.2023.122282] [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: 02/18/2023] [Revised: 10/19/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023]
Abstract
As one of the most prevalent neurotrophic factors in the central nervous system (CNS), brain-derived neurotrophic factor (BDNF) plays a significant role in CNS injury by binding to its specific receptor Tropomyosin-related kinase receptor B (TrkB). The BDNF/TrkB signaling pathway is crucial for neuronal survival, structural changes, and plasticity. BDNF acts as an axonal growth and extension factor, a pro-survival factor, and a synaptic modulator in the CNS. BDNF also plays an important role in the maintenance and plasticity of neuronal circuits. Several studies have demonstrated the importance of BDNF in the treatment and recovery of neurodegenerative and neurotraumatic disorders. By undertaking in-depth study on the mechanism of BDNF/TrkB function, important novel therapeutic strategies for treating neuropsychiatric disorders have been discovered. In this review, we discuss the expression patterns and mechanisms of the TrkB/BDNF signaling pathway in CNS damage and introduce several intriguing small molecule TrkB receptor agonists produced over the previous several decades.
Collapse
Affiliation(s)
- Yujin Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jing Liang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; School of Stomatology, Tongji University, Shanghai 200072, China
| | - Boyu Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jin Yang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| |
Collapse
|
5
|
Carrion SA, Michal JJ, Jiang Z. Alternative Transcripts Diversify Genome Function for Phenome Relevance to Health and Diseases. Genes (Basel) 2023; 14:2051. [PMID: 38002994 PMCID: PMC10671453 DOI: 10.3390/genes14112051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Manipulation using alternative exon splicing (AES), alternative transcription start (ATS), and alternative polyadenylation (APA) sites are key to transcript diversity underlying health and disease. All three are pervasive in organisms, present in at least 50% of human protein-coding genes. In fact, ATS and APA site use has the highest impact on protein identity, with their ability to alter which first and last exons are utilized as well as impacting stability and translation efficiency. These RNA variants have been shown to be highly specific, both in tissue type and stage, with demonstrated importance to cell proliferation, differentiation and the transition from fetal to adult cells. While alternative exon splicing has a limited effect on protein identity, its ubiquity highlights the importance of these minor alterations, which can alter other features such as localization. The three processes are also highly interwoven, with overlapping, complementary, and competing factors, RNA polymerase II and its CTD (C-terminal domain) chief among them. Their role in development means dysregulation leads to a wide variety of disorders and cancers, with some forms of disease disproportionately affected by specific mechanisms (AES, ATS, or APA). Challenges associated with the genome-wide profiling of RNA variants and their potential solutions are also discussed in this review.
Collapse
Affiliation(s)
| | | | - Zhihua Jiang
- Department of Animal Sciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164-7620, USA; (S.A.C.); (J.J.M.)
| |
Collapse
|
6
|
Gramatiuk SM, Ivanova YV, Hudyma AA, Sargsyan K, Kryvoruchko IA, Puliaieva IS. Differentiation of neurosphere after transplantation into the damaged spinal cord. J Med Life 2023; 16:689-698. [PMID: 37520471 PMCID: PMC10375341 DOI: 10.25122/jml-2022-0346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/26/2023] [Indexed: 08/01/2023] Open
Abstract
This study aimed to compare the differentiation and survival of human neural stem/progenitor cells of various origins in vitro and after transplantation into the injured spinal cord of laboratory animals. Rats with simulated spinal cord injury were transplanted with neurosphere cells obtained by directed differentiation of HUES6 cell lines. Fluorescence microscopy was used to visualize the obtained results. HUES6#1 and iPSC#1 neurospheres showed a wide range of markers associated with glial differentiation. The expression of the proliferation marker Ki67 did not exceed 25%, both in the lines of early and late neurospheres. Although neurospheres did not fully differentiate into astrocytes in vitro, they massively approached the GFAP+ astrocyte phenotype when exposed to the transplanted environment. PSC-derived neurospheres transplanted into the site of SM injury without additional growth factors showed only moderate survival, a significant degree of differentiation into astrocytes, and moderate differentiation into neurons. The difference in the survival and differentiation of HUES6#1 and iPSC#1 neurospheres, both in vitro and in vivo, can be explained by the difference in the regulatory behavior of signaling molecules corresponding to the source of origin of PSCs. Derivatives of human PSCs of various origins obtained according to the described differentiation protocol did not mature into astrocytic populations, nor did the glycogenic transition of PSC-derived NSCs occur in vitro. The study demonstrated the impact of the injured spinal cord microenvironment on the differentiation of transplanted HUES6#1 and iPSC#1 into astrocytes. The results showed that HUES6-derived neurospheres generated 90% of GFAP+ astrocytes and 5-10% of early neurons, while iPSC-derived neurospheres generated an average of 74% GFAP+ astrocytes and 5% of early neurons in vivo.
Collapse
Affiliation(s)
- Svetlana Mykolaiivna Gramatiuk
- Department of Biotechnology, Institute of Bio-Stem Cell Rehabilitation of the Ukrainian Association of Biobanks, Kharkiv, Ukraine
- Department of Biotechnology, Louisiana State University, Baton Rouge, Louisiana, USA
- International Biobanking and Education, Medical University of Graz, Graz, Austria
| | - Yulia Viktorovna Ivanova
- Department of Biotechnology, Institute of Bio-Stem Cell Rehabilitation of the Ukrainian Association of Biobanks, Kharkiv, Ukraine
- Department of Surgery No.1, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Arsen Arsenievich Hudyma
- Emergency Medical Care, Ternopil National Medical University named after I. Ya. Gorbachevsky, Ternopil, Ukraine
| | - Karine Sargsyan
- International Biobanking and Education, Medical University of Graz, Graz, Austria
- Department of Medical Genetics, Yerevan State Medical University, Yerevan, Armenia
| | | | - Inna Sergeevna Puliaieva
- Department of Biotechnology, Institute of Bio-Stem Cell Rehabilitation of the Ukrainian Association of Biobanks, Kharkiv, Ukraine
| |
Collapse
|
7
|
Bazzari AH, Bazzari FH. BDNF Therapeutic Mechanisms in Neuropsychiatric Disorders. Int J Mol Sci 2022; 23:ijms23158417. [PMID: 35955546 PMCID: PMC9368938 DOI: 10.3390/ijms23158417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain and functions as both a primary neurotrophic signal and a neuromodulator. It serves essential roles in neuronal development, maintenance, transmission, and plasticity, thereby influencing aging, cognition, and behavior. Accumulating evidence associates reduced central and peripheral BDNF levels with various neuropsychiatric disorders, supporting its potential utilization as a biomarker of central pathologies. Subsequently, extensive research has been conducted to evaluate restoring, or otherwise augmenting, BDNF transmission as a potential therapeutic approach. Promising results were indeed observed for genetic BDNF upregulation or exogenous administration using a multitude of murine models of neurological and psychiatric diseases. However, varying mechanisms have been proposed to underlie the observed therapeutic effects, and many findings indicate the engagement of disease-specific and other non-specific mechanisms. This is because BDNF essentially affects all aspects of neuronal cellular function through tropomyosin receptor kinase B (TrkB) receptor signaling, the disruptions of which vary between brain regions across different pathologies leading to diversified consequences on cognition and behavior. Herein, we review the neurophysiology of BDNF transmission and signaling and classify the converging and diverging molecular mechanisms underlying its therapeutic potentials in neuropsychiatric disorders. These include neuroprotection, synaptic maintenance, immunomodulation, plasticity facilitation, secondary neuromodulation, and preservation of neurovascular unit integrity and cellular viability. Lastly, we discuss several findings suggesting BDNF as a common mediator of the therapeutic actions of centrally acting pharmacological agents used in the treatment of neurological and psychiatric illness.
Collapse
Affiliation(s)
- Amjad H. Bazzari
- Faculty of Medicine, Arab American University, 13 Zababdeh, Jenin 240, Palestine
- Correspondence:
| | - Firas H. Bazzari
- Faculty of Pharmacy, Arab American University, 13 Zababdeh, Jenin 240, Palestine;
| |
Collapse
|
8
|
Hvid LG, Harwood DL, Eskildsen SF, Dalgas U. A Critical Systematic Review of Current Evidence on the Effects of Physical Exercise on Whole/Regional Grey Matter Brain Volume in Populations at Risk of Neurodegeneration. Sports Med 2021; 51:1651-1671. [PMID: 33861414 DOI: 10.1007/s40279-021-01453-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the intriguing potential of physical exercise being able to preserve or even restore brain volume (grey matter volume in particular)-a tissue essential for both cognitive and physical function-no reviews have so far synthesized the existing knowledge from randomized controlled trials investigating exercise-induced changes of the brain's grey matter volume in populations at risk of neurodegeneration. Our objective was to critically review the existing evidence regarding this topic. METHODS A systematic search was carried out in MEDLINE and EMBASE databases primo April 2020, to identify randomized controlled trials evaluating the effects of aerobic training, resistance training or concurrent training on brain grey volume changes (by MRI) in adult clinical or healthy elderly populations. RESULTS A total of 20 articles (from 19 RCTs) evaluating 3-12 months of aerobic, resistance, or concurrent training were identified and included, involving a total of 1662 participants (populations: healthy older adults, older adults with mild cognitive impairment or Alzheimer's disease, adults with schizophrenia or multiple sclerosis or major depression). While few studies indicated a positive effect-although modest-of physical exercise on certain regions of brain grey matter volume, the majority of study findings were neutral (i.e., no effects/small effect sizes) and quite divergent across populations. Meta-analyses showed that different exercise modalities failed to elicit any substantial effects on whole brain grey volume and hippocampus volume, although with rather large confidence interval width (i.e., variability). CONCLUSION Altogether, the current evidence on the effects of physical exercise on whole/regional grey matter brain volume appear sparse and inconclusive, and does not support that physical exercise is as potent as previously proposed when it comes to affecting brain grey matter volume.
Collapse
Affiliation(s)
- Lars G Hvid
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark.
| | - Dylan L Harwood
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ulrik Dalgas
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Roos BB, Teske JJ, Bhallamudi S, Pabelick CM, Sathish V, Prakash YS. Neurotrophin Regulation and Signaling in Airway Smooth Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:109-121. [PMID: 34019266 PMCID: PMC11042712 DOI: 10.1007/978-3-030-68748-9_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Structural and functional aspects of bronchial airways are key throughout life and play critical roles in diseases such as asthma. Asthma involves functional changes such as airway irritability and hyperreactivity, as well as structural changes such as enhanced cellular proliferation of airway smooth muscle (ASM), epithelium, and fibroblasts, and altered extracellular matrix (ECM) and fibrosis, all modulated by factors such as inflammation. There is now increasing recognition that disease maintenance following initial triggers involves a prominent role for resident nonimmune airway cells that secrete growth factors with pleiotropic autocrine and paracrine effects. The family of neurotrophins may be particularly relevant in this regard. Long recognized in the nervous system, classical neurotrophins such as brain-derived neurotrophic factor (BDNF) and nonclassical ligands such as glial-derived neurotrophic factor (GDNF) are now known to be expressed and functional in non-neuronal systems including lung. However, the sources, targets, regulation, and downstream effects are still under investigation. In this chapter, we discuss current state of knowledge and future directions regarding BDNF and GDNF in airway physiology and on pathophysiological contributions in asthma.
Collapse
Affiliation(s)
- Benjamin B Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jacob J Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sangeeta Bhallamudi
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
10
|
One-Week High-Intensity Interval Training Increases Hippocampal Plasticity and Mitochondrial Content without Changes in Redox State. Antioxidants (Basel) 2020; 9:antiox9050445. [PMID: 32455608 PMCID: PMC7278594 DOI: 10.3390/antiox9050445] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/20/2022] Open
Abstract
Evidence suggests that physical exercise has effects on neuronal plasticity as well as overall brain health. This effect has been linked to exercise capacity in modulating the antioxidant status, when the oxidative stress is usually linked to the neuronal damage. Although high-intensity interval training (HIIT) is the training-trend worldwide, its effect on brain function is still unclear. Thus, we aimed to assess the neuroplasticity, mitochondrial, and redox status after one-week HIIT training. Male (C57Bl/6) mice were assigned to non-trained or HIIT groups. The HIIT protocol consisted of three days with short bouts at 130% of maximum speed (Vmax), intercalated with moderate-intensity continuous exercise sessions of 30 min at 60% Vmax. The mass spectrometry analyses showed that one-week of HIIT increased minichromosome maintenance complex component 2 (MCM2), brain derived neutrophic factor (BDNF), doublecortin (DCX) and voltage-dependent anion-selective channel protein 2 (VDAC), and decreased mitochondrial superoxide dismutase 2 (SOD 2) in the hippocampus. In addition, one-week of HIIT promoted no changes in H2O2 production and carbonylated protein concentration in the hippocampus as well as in superoxide anion production in the dentate gyrus. In conclusion, our one-week HIIT protocol increased neuroplasticity and mitochondrial content regardless of changes in redox status, adding new insights into the neuronal modulation induced by new training models.
Collapse
|
11
|
Chen S, Zhu J, Li P, Xia Z, Tu M, Lin Z, Xu B, Fu X. 3'UTRs Regulate Mouse Ntrk2 mRNA Distribution in Cortical Neurons. J Mol Neurosci 2020; 70:1858-1870. [PMID: 32430868 PMCID: PMC7561570 DOI: 10.1007/s12031-020-01579-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/01/2020] [Indexed: 12/01/2022]
Abstract
There are two major isoforms of NTRK2 (neurotrophic receptor tyrosine kinase 2, or TrkB), full-length isoform with tyrosine kinase (TK) domain intact (+) and spliced isoform without tyrosine kinase domain (TK(−)). Within each isoform, there exist subtypes with minor modifications of the protein sequences. In human, the NTRK2 mRNA transcripts encoding TK(+) have same 3′UTRs, while the transcripts encoding subtypes of NTRK2 TK(−) have two completely different 3′UTRs. In mouse, the mRNA transcripts encoding same NTRK2 protein sequence for either TK(+) or TK(−) have long or short 3′UTRs, respectively. The physiological functions of these different 3′UTRs are still unknown. Pilocarpine stimulation increased Ntrk2 mRNA levels in soma, while the increase in synaptosome was smaller. FISH results further showed that mouse Ntrk2 transcripts with different 3′UTRs were distributed differently in cultured cortical neurons. The transcripts with long 3′UTR were distributed more in apical dendrites compared with transcripts with short 3′UTR. Our results provide evidence of non-coding 3′UTR function in regulating mRNA distribution in neurons.
Collapse
Affiliation(s)
- Shangqin Chen
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jinjin Zhu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhaonan Xia
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Mengjing Tu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhenlang Lin
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Xiaoqin Fu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
12
|
Hernandez-Echeagaray E. The role of the TrkB-T1 receptor in the neurotrophin-4/5 antagonism of brain-derived neurotrophic factor on corticostriatal synaptic transmission. Neural Regen Res 2020; 15:1973-1976. [PMID: 32394943 PMCID: PMC7716028 DOI: 10.4103/1673-5374.282224] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This manuscript reviews the function and fundamental characteristics of the neurotrophins and their receptors to introduce the reader to the differential effects exhibited by the neurotrophins; brain-derived neurotrophic factor and neurotrophin 4/5 when acted together after sequential presentation. The neurotrophin 4/5 exhibits an inhibitory action on the modulatory effect of brain-derived neurotrophic factor in corticostriatal synapses when they are administered sequentially (brain-derived neurotrophic factor to neurotrophin 4/5). This inhibitory effect has not been previously documented and is relevant for these neurotrophins as both of them stimulate the TrkB receptor. The additive effect of these neurotrophins is also discussed and occurs when neurotrophin 4/5 exposure is followed by brain-derived neurotrophic factor in a mouse model of striatal degeneration. Occlusive and additive effects of both neurotrophins are accompanied by changes in the expression of the TrkB receptor isoforms, specifically TrkB-T1 and TrkB-FL, as well as differences in phosphorylation levels of the TrkB receptor. The results of the experiments described raise several questions to inquire about the role that TrkB-T1 receptor plays in striatal physiology, as well as the functional relevance of the interaction of brain-derived neurotrophic factor and neurotrophin 4/5 in the brain and more specifically at the striatal circuits in normal as well as pathological conditions.
Collapse
|
13
|
Cao T, Matyas JJ, Renn CL, Faden AI, Dorsey SG, Wu J. Function and Mechanisms of Truncated BDNF Receptor TrkB.T1 in Neuropathic Pain. Cells 2020; 9:cells9051194. [PMID: 32403409 PMCID: PMC7290366 DOI: 10.3390/cells9051194] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a major focus for regenerative therapeutics, has been lauded for its pro-survival characteristics and involvement in both development and recovery of function within the central nervous system (CNS). However, studies of tyrosine receptor kinase B (TrkB), a major receptor for BDNF, indicate that certain effects of the TrkB receptor in response to disease or injury may be maladaptive. More specifically, imbalance among TrkB receptor isoforms appears to contribute to aberrant signaling and hyperpathic pain. A truncated isoform of the receptor, TrkB.T1, lacks the intracellular kinase domain of the full length receptor and is up-regulated in multiple CNS injury models. Such up-regulation is associated with hyperpathic pain, and TrkB.T1 inhibition reduces neuropathic pain in various experimental paradigms. Deletion of TrkB.T1 also limits astrocyte changes in vitro, including proliferation, migration, and activation. Mechanistically, TrkB.T1 is believed to act through release of intracellular calcium in astrocytes, as well as through interactions with neurotrophins, leading to cell cycle activation. Together, these studies support a potential role for astrocytic TrkB.T1 in hyperpathic pain and suggest that targeted strategies directed at this receptor may have therapeutic potential.
Collapse
Affiliation(s)
- Tuoxin Cao
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
| | - Jessica J. Matyas
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
| | - Cynthia L. Renn
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD 21201, USA; (C.L.R.); (S.G.D.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Alan I. Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Susan G. Dorsey
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD 21201, USA; (C.L.R.); (S.G.D.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-706-5189
| |
Collapse
|
14
|
Jeon MT, Moon GJ, Kim S, Choi M, Oh YS, Kim DW, Kim HJ, Lee KJ, Choe Y, Ha CM, Jang IS, Nakamura M, McLean C, Chung WS, Shin WH, Lee SG, Kim SR. Neurotrophic interactions between neurons and astrocytes following AAV1-Rheb(S16H) transduction in the hippocampus in vivo. Br J Pharmacol 2019; 177:668-686. [PMID: 31658360 PMCID: PMC7012949 DOI: 10.1111/bph.14882] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 12/28/2022] Open
Abstract
Background and Purpose We recently reported that AAV1‐Rheb(S16H) transduction could protect hippocampal neurons through the induction of brain‐derived neurotrophic factor (BDNF) in the rat hippocampus in vivo. It is still unclear how neuronal BDNF produced by AAV1‐Rheb(S16H) transduction induces neuroprotective effects in the hippocampus and whether its up‐regulation contributes to the enhance of a neuroprotective system in the adult brain. Experimental Approach To determine the presence of a neuroprotective system in the hippocampus of patients with Alzheimer's disease (AD), we examined the levels of glial fibrillary acidic protein, BDNF and ciliary neurotrophic factor (CNTF) and their receptors, tropomyocin receptor kinase B (TrkB) and CNTF receptor α(CNTFRα), in the hippocampus of AD patients. We also determined whether AAV1‐Rheb(S16H) transduction stimulates astroglial activation and whether reactive astrocytes contribute to neuroprotection in models of hippocampal neurotoxicity in vivo and in vitro. Key Results AD patients may have a potential neuroprotective system, demonstrated by increased levels of full‐length TrkB and CNTFRα in the hippocampus. Further AAV1‐Rheb(S16H) transduction induced sustained increases in the levels of full‐length TrkB and CNTFRα in reactive astrocytes and hippocampal neurons. Moreover, neuronal BDNF produced by Rheb(S16H) transduction of hippocampal neurons induced reactive astrocytes, resulting in CNTF production through the activation of astrocytic TrkB and the up‐regulation of neuronal BDNF and astrocytic CNTF which had synergistic effects on the survival of hippocampal neurons in vivo. Conclusions and Implications The results demonstrated that Rheb(S16H) transduction of hippocampal neurons could strengthen the neuroprotective system and this intensified system may have a therapeutic value against neurodegeneration in the adult brain.
Collapse
Affiliation(s)
- Min-Tae Jeon
- School of Life Sciences, Kyungpook National University, Daegu, Korea.,BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Gyeong Joon Moon
- School of Life Sciences, Kyungpook National University, Daegu, Korea.,BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Sehwan Kim
- School of Life Sciences, Kyungpook National University, Daegu, Korea.,BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Minji Choi
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Yong-Seok Oh
- Department of Brain-Cognitive Science, Daegu-Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Dong Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyung-Jun Kim
- Department of Neural Development and Disease, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Korea
| | - Kea Joo Lee
- Department of Neural Development and Disease, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Korea
| | - Youngshik Choe
- Department of Neural Development and Disease, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Korea
| | - Chang Man Ha
- Department of Neural Development and Disease, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Korea
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| | - Michiko Nakamura
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| | - Catriona McLean
- Victorian Brain Bank Network, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Department of Anatomical Pathology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Won-Ho Shin
- Predictive Model Research Center, Korea Institute of Toxicology, Daejeon, Korea
| | - Seok-Geun Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Korea
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu, Korea.,BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea.,Institute of Life Science and Biotechnology, Kyungpook National University, Daegu, Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| |
Collapse
|
15
|
Functional Group-Dependent Induction of Astrocytogenesis and Neurogenesis by Flavone Derivatives. Biomolecules 2019; 9:biom9120812. [PMID: 31810286 PMCID: PMC6995541 DOI: 10.3390/biom9120812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) differentiate into multiple cell types, including neurons, astrocytes, and oligodendrocytes, and provide an excellent platform to screen drugs against neurodegenerative diseases. Flavonoids exert a wide range of biological functions on several cell types and affect the fate of NSCs. In the present study, we investigated whether the structure-activity relationships of flavone derivatives influence NSC differentiation. As previously reported, we observed that PD98059 (2′-amino-3′-methoxy-flavone), compound 2 (3′-methoxy-flavone) induced astrocytogenesis. In the present study, we showed that compound 3 (2′-hydroxy-3′-methoxy-flavone), containing a 3′-methoxy group, and a non-bulky group at C2′ and C4′, induced astrocytogenesis through JAK-STAT3 signaling pathway. However, compound 1 and 7–12 without the methoxy group did not show such effects. Interestingly, the compounds 4 (2′,3′-dimethoxyflavone), 5 (2′-N-phenylacetamido-3′-methoxy-flavone), and 6 (3′,4′-dimethoxyflavone) containing 3′-methoxy could not promote astrocytic differentiation, suggesting that both the methoxy groups at C3′ and non-bulky group at C2′ and C4′ are required for the induction of astrocytogenesis. Notably, compound 6 promoted neuronal differentiation, whereas its 4′-demethoxylated analog, compound 2, repressed neurogenesis, suggesting an essential role of the methoxy group at C4′ in neurogenesis. These findings revealed that subtle structural changes of flavone derivatives have pronounced effects on NSC differentiation and can guide to design and develop novel flavone chemicals targeting NSCs fate regulation.
Collapse
|
16
|
Dedoni S, Marras L, Olianas MC, Ingianni A, Onali P. Downregulation of TrkB Expression and Signaling by Valproic Acid and Other Histone Deacetylase Inhibitors. J Pharmacol Exp Ther 2019; 370:490-503. [PMID: 31308194 DOI: 10.1124/jpet.119.258129] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/14/2019] [Indexed: 01/27/2023] Open
Abstract
Valproic acid (VPA) has been shown to regulate the levels of brain-derived neurotrophic factor (BDNF), but it is not known whether this drug can affect the neuronal responses to BDNF. In the present study, we show that in retinoic acid-differentiated SH-SY5Y human neuroblastoma cells, prolonged exposure to VPA reduces the expression of the BDNF receptor TrkB at the protein and mRNA levels and inhibits the intracellular signaling, neurotrophic activity, and prosurvival function of BDNF. VPA downregulates TrkB and curtails BDNF-induced signaling also in differentiated Kelly and LAN-1 neuroblastoma cells and primary mouse cortical neurons. The VPA effect is mimicked by several histone deacetylase (HDAC) inhibitors, including the class I HDAC inhibitors entinostat and romidepsin. Conversely, the class II HDAC inhibitor MC1568, the HDAC6 inhibitor tubacin, the HDAC8 inhibitor PCI-34051, and the VPA derivative valpromide have no effect. In neuroblastoma cells and primary neurons both VPA and entinostat increase the cellular levels of the transcription factor RUNX3, which negatively regulates TrkB gene expression. Treatment with RUNX3 siRNA attenuates VPA-induced RUNX3 elevation and TrkB downregulation. VPA, entinostat, HDAC1 depletion by siRNA, and 3-deazaneplanocin A (DZNep), an inhibitor of the polycomb repressor complex 2 (PRC2), decrease the PRC2 core component EZH2, a RUNX3 suppressor. Like VPA, HDAC1 depletion and DZNep increase RUNX3 and decrease TrkB expression. These results indicate that VPA downregulates TrkB through epigenetic mechanisms involving the EZH2/RUNX3 axis and provide evidence that this effect implicates relevant consequences with regard to BDNF efficacy in stimulating intracellular signaling and functional responses. SIGNIFICANCE STATEMENT: The tropomyosin-related kinase receptor B (TrkB) mediates the stimulatory effects of brain-derived neurotrophic factor (BDNF) on neuronal growth, differentiation, and survival and is highly expressed in aggressive neuroblastoma and other tumors. Here we show that exposure to valproic acid (VPA) downregulates TrkB expression and functional activity in retinoic acid-differentiated human neuroblastoma cell lines and primary mouse cortical neurons. The effects of VPA are mimicked by other histone deacetylase (HDAC) inhibitors and HDAC1 knockdown and appear to be mediated by an epigenetic mechanism involving the upregulation of RUNX3, a suppressor of TrkB gene expression. TrkB downregulation may have relevance for the use of VPA as a potential therapeutic agent in neuroblastoma and other pathologies characterized by an excessive BDNF/TrkB signaling.
Collapse
Affiliation(s)
- Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Luisa Marras
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Angela Ingianni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences, Department of Biomedical Sciences (S.D., M.C.O., P.O.) and Section of Microbiology, Department of Biomedical Sciences (L.M., A.I.), University of Cagliari, Cagliari, Italy
| |
Collapse
|
17
|
Fletcher JL, Murray SS, Xiao J. Brain-Derived Neurotrophic Factor in Central Nervous System Myelination: A New Mechanism to Promote Myelin Plasticity and Repair. Int J Mol Sci 2018; 19:ijms19124131. [PMID: 30572673 PMCID: PMC6321406 DOI: 10.3390/ijms19124131] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/27/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays vitally important roles in neural development and plasticity in both health and disease. Recent studies using mutant mice to selectively manipulate BDNF signalling in desired cell types, in combination with animal models of demyelinating disease, have demonstrated that BDNF not only potentiates normal central nervous system myelination in development but enhances recovery after myelin injury. However, the precise mechanisms by which BDNF enhances myelination in development and repair are unclear. Here, we review some of the recent progress made in understanding the influence BDNF exerts upon the myelinating process during development and after injury, and discuss the cellular and molecular mechanisms underlying its effects. In doing so, we raise new questions for future research.
Collapse
Affiliation(s)
- Jessica L Fletcher
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, 3010, VIC, Australia.
| | - Simon S Murray
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, 3010, VIC, Australia.
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, 3010, VIC, Australia.
| |
Collapse
|
18
|
Kowiański P, Lietzau G, Czuba E, Waśkow M, Steliga A, Moryś J. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell Mol Neurobiol 2018; 38:579-593. [PMID: 28623429 PMCID: PMC5835061 DOI: 10.1007/s10571-017-0510-4] [Citation(s) in RCA: 779] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/08/2017] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most widely distributed and extensively studied neurotrophins in the mammalian brain. Among its prominent functions, one can mention control of neuronal and glial development, neuroprotection, and modulation of both short- and long-lasting synaptic interactions, which are critical for cognition and memory. A wide spectrum of processes are controlled by BDNF, and the sometimes contradictory effects of its action can be explained based on its specific pattern of synthesis, comprising several intermediate biologically active isoforms that bind to different types of receptor, triggering several signaling pathways. The functions of BDNF must be discussed in close relation to the stage of brain development, the different cellular components of nervous tissue, as well as the molecular mechanisms of signal transduction activated under physiological and pathological conditions. In this review, we briefly summarize the current state of knowledge regarding the impact of BDNF on regulation of neurophysiological processes. The importance of BDNF for future studies aimed at disclosing mechanisms of activation of signaling pathways, neuro- and gliogenesis, as well as synaptic plasticity is highlighted.
Collapse
Affiliation(s)
- Przemysław Kowiański
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland.
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland.
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| | - Monika Waśkow
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland
| | - Aleksandra Steliga
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| |
Collapse
|
19
|
Kinboshi M, Mukai T, Nagao Y, Matsuba Y, Tsuji Y, Tanaka S, Tokudome K, Shimizu S, Ito H, Ikeda A, Inanobe A, Kurachi Y, Inoue S, Ohno Y. Inhibition of Inwardly Rectifying Potassium (Kir) 4.1 Channels Facilitates Brain-Derived Neurotrophic Factor (BDNF) Expression in Astrocytes. Front Mol Neurosci 2017; 10:408. [PMID: 29358904 PMCID: PMC5768989 DOI: 10.3389/fnmol.2017.00408] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/24/2017] [Indexed: 11/16/2022] Open
Abstract
Inwardly rectifying potassium (Kir) 4.1 channels in astrocytes regulate neuronal excitability by mediating spatial potassium buffering. Although dysfunction of astrocytic Kir4.1 channels is implicated in the development of epileptic seizures, the functional mechanisms of Kir4.1 channels in modulating epileptogenesis remain unknown. We herein evaluated the effects of Kir4.1 inhibition (blockade and knockdown) on expression of brain-derived neurotrophic factor (BDNF), a key modulator of epileptogenesis, in the primary cultures of mouse astrocytes. For blockade of Kir4.1 channels, we tested several antidepressant agents which reportedly bound to and blocked Kir4.1 channels in a subunit-specific manner. Treatment of astrocytes with fluoxetine enhanced BDNF mRNA expression in a concentration-dependent manner and increased the BDNF protein level. Other antidepressants (e.g., sertraline and imipramine) also increased the expression of BDNF mRNA with relative potencies similar to those for inhibition of Kir4.1 channels. In addition, suppression of Kir4.1 expression by the transfection of small interfering RNA (siRNA) targeting Kir4.1 significantly increased the mRNA and protein levels of BDNF. The BDNF induction by Kir4.1 siRNA transfection was suppressed by the MEK1/2 inhibitor U0126, but not by the p38 MAPK inhibitor SB202190 or the JNK inhibitor SP600125. The present results demonstrated that inhibition of Kir4.1 channels facilitates BDNF expression in astrocytes primarily by activating the Ras/Raf/MEK/ERK pathway, which may be linked to the development of epilepsy and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Masato Kinboshi
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan.,Department of Neurology, Wakayama Medical University, Wakayama, Japan.,Department of Epilepsy, Movement Disorders and Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Mukai
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Yuki Nagao
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Yusuke Matsuba
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Yoshimi Tsuji
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Shiho Tanaka
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Kentaro Tokudome
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan.,Department of Molecular and Cellular Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Saki Shimizu
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Akio Ikeda
- Department of Epilepsy, Movement Disorders and Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Inanobe
- Department of Molecular and Cellular Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshihisa Kurachi
- Department of Molecular and Cellular Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Seiji Inoue
- Education and Research Center for Fundamental Pharmaceutical Sciences, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Yukihiro Ohno
- Laboratory of Pharmacology, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| |
Collapse
|
20
|
Weissleder C, Kondo MA, Yang C, Fung SJ, Rothmond DA, Wong MW, Halliday GM, Herman MM, Kleinman JE, Webster MJ, Shannon Weickert C. Early-life decline in neurogenesis markers and age-related changes of TrkB splice variant expression in the human subependymal zone. Eur J Neurosci 2017; 46:1768-1778. [PMID: 28612959 DOI: 10.1111/ejn.13623] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 05/26/2017] [Accepted: 06/05/2017] [Indexed: 11/28/2022]
Abstract
Neurogenesis in the subependymal zone (SEZ) declines across the human lifespan, and reduced local neurotrophic support is speculated to be a contributing factor. While tyrosine receptor kinase B (TrkB) signalling is critical for neuronal differentiation, maturation and survival, little is known about subependymal TrkB expression changes during postnatal human life. In this study, we used quantitative PCR and in situ hybridisation to determine expression of the cell proliferation marker Ki67, the immature neuron marker doublecortin (DCX) and both full-length (TrkB-TK+) and truncated TrkB receptors (TrkB-TK-) in the human SEZ from infancy to middle age (n = 26-35, 41 days to 43 years). We further measured TrkB-TK+ and TrkB-TK- mRNAs in the SEZ from young adulthood into ageing (n = 50, 21-103 years), and related their transcript levels to neurogenic and glial cell markers. Ki67, DCX and both TrkB splice variant mRNAs significantly decreased in the SEZ from infancy to middle age. In contrast, TrkB-TK- mRNA increased in the SEZ from young adulthood into ageing, whereas TrkB-TK+ mRNA remained stable. TrkB-TK- mRNA positively correlated with expression of neural precursor (glial fibrillary acidic protein delta and achaete-scute homolog 1) and glial cell markers (vimentin and pan glial fibrillary acidic protein). TrkB-TK+ mRNA positively correlated with expression of neuronal cell markers (DCX and tubulin beta 3 class III). Our results indicate that cells residing in the human SEZ maintain their responsiveness to neurotrophins; however, this capability may change across postnatal life. We suggest that TrkB splice variants may differentially influence neuronal and glial differentiation in the human SEZ.
Collapse
Affiliation(s)
- Christin Weissleder
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Margarete Ainsworth Building, 139 Barker Street, Randwick, NSW, 2031, Australia.,Schizophrenia Research Institute, Randwick, NSW, Australia.,Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Mari A Kondo
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Margarete Ainsworth Building, 139 Barker Street, Randwick, NSW, 2031, Australia.,Schizophrenia Research Institute, Randwick, NSW, Australia.,Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Chunhui Yang
- Section on Neuropathology, Clinical Brain Disorders Branch, Intramural Research Program, NIMH, NIH, Bethesda, MD, USA.,Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Samantha J Fung
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Margarete Ainsworth Building, 139 Barker Street, Randwick, NSW, 2031, Australia.,Schizophrenia Research Institute, Randwick, NSW, Australia.,Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Debora A Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Margarete Ainsworth Building, 139 Barker Street, Randwick, NSW, 2031, Australia.,Schizophrenia Research Institute, Randwick, NSW, Australia
| | - Matthew W Wong
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Margarete Ainsworth Building, 139 Barker Street, Randwick, NSW, 2031, Australia.,Schizophrenia Research Institute, Randwick, NSW, Australia.,Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Glenda M Halliday
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia, Randwick, NSW, Australia
| | - Mary M Herman
- Section on Neuropathology, Clinical Brain Disorders Branch, Intramural Research Program, NIMH, NIH, Bethesda, MD, USA
| | - Joel E Kleinman
- Department of Psychiatry and Behavioral Sciences, Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, Chevy Chase, MD, USA
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Margarete Ainsworth Building, 139 Barker Street, Randwick, NSW, 2031, Australia.,Schizophrenia Research Institute, Randwick, NSW, Australia.,Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
21
|
Tang T, Rios-Pilier J, Krimm R. Taste bud-derived BDNF maintains innervation of a subset of TrkB-expressing gustatory nerve fibers. Mol Cell Neurosci 2017; 82:195-203. [PMID: 28600222 DOI: 10.1016/j.mcn.2017.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/08/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023] Open
Abstract
Taste receptor cells transduce different types of taste stimuli and transmit this information to gustatory neurons that carry it to the brain. Taste receptor cells turn over continuously in adulthood, requiring constant new innervation from nerve fibers. Therefore, the maintenance of innervation to taste buds is an active process mediated by many factors, including brain-derived neurotrophic factor (BDNF). Specifically, 40% of taste bud innervation is lost when Bdnf is removed during adulthood. Here we speculated that not all gustatory nerve fibers express the BDNF receptor, TrkB, resulting in subsets of neurons that vary in their response to BDNF. However, it is also possible that the partial loss of innervation occurred because the Bdnf gene was not effectively removed. To test these possibilities, we first determined that not all gustatory nerve fibers express the TrkB receptor in adult mice. We then verified the efficiency of Bdnf removal specifically in taste buds of K14-CreER:Bdnf mice and found that Bdnf expression was reduced to 1%, indicating efficient Bdnf gene recombination. BDNF removal resulted in a 55% loss of TrkB-expressing nerve fibers, which was greater than the loss of P2X3-positive fibers (39%), likely because taste buds were innervated by P2X3+/TrkB- fibers that were unaffected by BDNF removal. We conclude that gustatory innervation consists of both TrkB-positive and TrkB-negative taste fibers and that BDNF is specifically important for maintaining TrkB-positive innervation to taste buds. In addition, although taste bud size was not affected by inducible Bdnf removal, the expression of the γ subunit of the ENaC channel was reduced. So, BDNF may regulate expression of some molecular components of taste transduction pathways.
Collapse
Affiliation(s)
- Tao Tang
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Jennifer Rios-Pilier
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Robin Krimm
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
22
|
Pramanik S, Sulistio YA, Heese K. Neurotrophin Signaling and Stem Cells-Implications for Neurodegenerative Diseases and Stem Cell Therapy. Mol Neurobiol 2016; 54:7401-7459. [PMID: 27815842 DOI: 10.1007/s12035-016-0214-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Neurotrophins (NTs) are members of a neuronal growth factor protein family whose action is mediated by the tropomyosin receptor kinase (TRK) receptor family receptors and the p75 NT receptor (p75NTR), a member of the tumor necrosis factor (TNF) receptor family. Although NTs were first discovered in neurons, recent studies have suggested that NTs and their receptors are expressed in various types of stem cells mediating pivotal signaling events in stem cell biology. The concept of stem cell therapy has already attracted much attention as a potential strategy for the treatment of neurodegenerative diseases (NDs). Strikingly, NTs, proNTs, and their receptors are gaining interest as key regulators of stem cells differentiation, survival, self-renewal, plasticity, and migration. In this review, we elaborate the recent progress in understanding of NTs and their action on various stem cells. First, we provide current knowledge of NTs, proNTs, and their receptor isoforms and signaling pathways. Subsequently, we describe recent advances in the understanding of NT activities in various stem cells and their role in NDs, particularly Alzheimer's disease (AD) and Parkinson's disease (PD). Finally, we compile the implications of NTs and stem cells from a clinical perspective and discuss the challenges with regard to transplantation therapy for treatment of AD and PD.
Collapse
Affiliation(s)
- Subrata Pramanik
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Yanuar Alan Sulistio
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea.
| |
Collapse
|
23
|
Somkuwar SS, Staples MC, Fannon MJ, Ghofranian A, Mandyam CD. Evaluating Exercise as a Therapeutic Intervention for Methamphetamine Addiction-Like Behavior. Brain Plast 2015; 1:63-81. [PMID: 29765835 PMCID: PMC5928557 DOI: 10.3233/bpl-150007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The need for effective treatments for addiction and dependence to the illicit stimulant methamphetamine in primary care settings is increasing, yet no effective medications have been FDA approved to reduce dependence [1]. This is partially attributed to the complex and dynamic neurobiology underlying the various stages of addiction [2]. Therapeutic strategies to treat methamphetamine addiction, particularly the relapse stage of addiction, could revolutionize methamphetamine addiction treatment. In this context, preclinical studies demonstrate that voluntary exercise (sustained physical activity) could be used as an intervention to reduce methamphetamine addiction. Therefore, it appears that methamphetamine disrupts normal functioning in the brain and this disruption is prevented or reduced by engaging in exercise. This review discusses animal models of methamphetamine addiction and sustained physical activity and the interactions between exercise and methamphetamine behaviors. The review highlights how methamphetamine and exercise affect neuronal plasticity and neurotoxicity in the adult mammalian striatum, hippocampus, and prefrontal cortex, and presents the emerging mechanisms of exercise in attenuating intake and in preventing relapse to methamphetamine seeking in preclinical models of methamphetamine addiction.
Collapse
Affiliation(s)
- Sucharita S Somkuwar
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Miranda C Staples
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - McKenzie J Fannon
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Atoosa Ghofranian
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Chitra D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
24
|
Gao S, Zheng Y, Cai Q, Wu X, Yao W, Wang J. Different methods for inducing adipose-derived stem cells to differentiate into Schwann-like cells. Arch Med Sci 2015; 11:886-92. [PMID: 26322102 PMCID: PMC4548042 DOI: 10.5114/aoms.2015.53310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/12/2013] [Accepted: 08/26/2013] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION The aim of the study was to explore an effective method to induce adipose-derived stem cells (ADSCs) to differentiate into Schwann-like cells in vitro. MATERIAL AND METHODS Reagents were applied in two different ways (Dezawa inducing method and modified inducing method) in which inducers including β-mercaptoethanol (β-ME), all-trans-retinoic acid (ATRA), type I collagenase, forskolin, heregulin, basic fibroblast growth factor (BFGF) and brain-derived neurotrophic factor (BDNF) were used in different ways to induce ADSCs of rats to differentiate into Schwann-like cells. After induction, the cell morphologic characteristics and the cellular immunohistochemical staining positive rate of anti-S100 and anti-GFAP (glial fibrillary acidic protein) antibodies and the gray value of immunocytochemical dye with anti-S100 and anti-GFAP antibodies and cell activity measured by the MTT method were compared with each other to evaluate the induction effects. RESULTS Both methods can induce differentiation of ADSCs of rats into Schwann-like cells, but the cellular morphology of the modified method was more similar to Schwann cells than that of the Dezawa inducing method, there was a higher cellular immunohistochemical staining positive rate and staining grey value in immunocytochemical dye with anti-S100 and anti-GFAP antibodies, and less damage in the cell activity of the modified inducing method than that of the Dezawa inducing method. CONCLUSIONS The effect of the modified method to induce ADSCs to differentiate into Schwann-like cells in vitro is superior to that of the Dezawa inducing method.
Collapse
Affiliation(s)
- Songtao Gao
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Zheng
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiqing Cai
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejian Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weitao Yao
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaqiang Wang
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Razavi S, Khosravizadeh Z, Bahramian H, Kazemi M. Time-Dependent Effect of Encapsulating Alginate Hydrogel on Neurogenic Potential. CELL JOURNAL 2015. [PMID: 26199909 PMCID: PMC4503844 DOI: 10.22074/cellj.2016.3736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Due to the restricted potential of neural stem cells for regeneration of central nervous system (CNS) after injury, providing an alternative source for neural stem cells is essential. Adipose derived stem cells (ADSCs) are multipotent cells with properties suitable for tissue engineering. In addition, alginate hydrogel is a biocompatible polysaccharide polymer that has been used to encapsulate many types of cells. The aim of this study was to assess the proliferation rate and level of expression of neural markers; NESTIN, glial fibrillary acidic protein (GFAP) and microtubule-associated protein 2 (MAP2) in encapsulated human ADSCs (hADSCs) 10 and14 days after neural induction. MATERIALS AND METHODS In this experimental study, ADSCs isolated from human were cultured in neural induction media and seeded into alginate hydrogel. The rate of proliferation and differentiation of encapsulated cells were evaluated by 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl tetrazolium bromide (MTT) assay, immunocytoflourescent and realtime reverse transcriptase polymerase chain reaction (RT-PCR) analyzes 10 and 14 days after induction. RESULTS The rate of proliferation of encapsulated cells was not significantly changed with time passage. The expression of NESTIN and GFAP significantly decreased on day 14 relative to day 10 (P<0.001) but MAP2 expression was increased. CONCLUSION Alginate hydrogel can promote the neural differentiation of encapsulated hADSCs with time passage.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Khosravizadeh
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Bahramian
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- Department of Genetic, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
26
|
Prakash YS, Martin RJ. Brain-derived neurotrophic factor in the airways. Pharmacol Ther 2014; 143:74-86. [PMID: 24560686 DOI: 10.1016/j.pharmthera.2014.02.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/10/2014] [Indexed: 12/13/2022]
Abstract
In addition to their well-known roles in the nervous system, there is increasing recognition that neurotrophins such as brain derived neurotrophic factor (BDNF) as well as their receptors are expressed in peripheral tissues including the lung, and can thus potentially contribute to both normal physiology and pathophysiology of several diseases. The relevance of this family of growth factors lies in emerging clinical data indicating altered neurotrophin levels and function in a range of diseases including neonatal and adult asthma, sinusitis, influenza, and lung cancer. The current review focuses on 1) the importance of BDNF expression and signaling mechanisms in early airway and lung development, critical to both normal neonatal lung function and also its disruption in prematurity and insults such as inflammation and infection; 2) how BDNF, potentially derived from airway nerves modulate neurogenic control of airway tone, a key aspect of airway reflexes as well as dysfunctional responses to allergic inflammation; 3) the emerging idea that local BDNF production by resident airway cells such as epithelium and airway smooth muscle can contribute to normal airway structure and function, and to airway hyperreactivity and remodeling in diseases such as asthma. Furthermore, given its pleiotropic effects in the airway, BDNF may be a novel and appealing therapeutic target.
Collapse
Affiliation(s)
- Y S Prakash
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN 55905, United States; Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905, United States.
| | - Richard J Martin
- Department of Pediatrics, Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH 44106, United States
| |
Collapse
|
27
|
Expression of full-length and truncated trkB in human striatum and substantia nigra neurons: implications for Parkinson's disease. J Mol Histol 2013; 45:349-61. [PMID: 24374887 DOI: 10.1007/s10735-013-9562-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/17/2013] [Indexed: 12/20/2022]
Abstract
Brain derived neurotrophic factor (BDNF) is a potent mediator of cell survival and differentiation and can reverse neuronal injury associated with Parkinson's disease (PD). Tropomyosin receptor kinase B (trkB) is the high affinity receptor for BDNF. There are two major trkB isoforms, the full-length receptor (trkB.tk(+)) and the truncated receptor (trkB.t1), that mediate the diverse, region specific functions of BDNF. Both trkB isoforms are widely distributed throughout the brain, but the isoform specific distribution of trkB.t1 and trkB.tk(+) to human neurons is not well characterized. Therefore, we report the regional and neuronal distribution of trkB.tk(+) and trkB.t1 in the striatum and substantia nigra pars compacta (SNpc) of human autopsy tissues from control and PD cases. In both PD and control tissues, we found abundant, punctate distribution of trkB.tk(+) and trkB.t1 proteins in striatum and SNpc neurons. In PD, trkB.tk(+) is decreased in striatal neurites, increased in striatal somata, decreased in SNpc somata and dendrites, and increased in SNpc axons. TrkB.t1 is increased in striatal somata, decreased in striatal axons, and increased in SNpc distal dendrites. We believe changes in trkB isoform distribution and expression levels may be markers of pathology and affect the neuronal response to BDNF.
Collapse
|
28
|
Permeability transition pore-mediated mitochondrial superoxide flashes mediate an early inhibitory effect of amyloid beta1-42 on neural progenitor cell proliferation. Neurobiol Aging 2013; 35:975-89. [PMID: 24325797 DOI: 10.1016/j.neurobiolaging.2013.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 08/22/2013] [Accepted: 11/05/2013] [Indexed: 01/04/2023]
Abstract
Cellular damage by reactive oxygen species and altered neurogenesis are implicated in the etiology of AD and the pathogenic actions of amyloid β-peptide (Aβ); the underlying mechanisms and the early oxidative intracellular events triggered by Aβ are not established. In the present study, we found that mouse embryonic cortical neural progenitor cells exhibit intermittent spontaneous mitochondrial superoxide (SO) flashes that require transient opening of mitochondrial permeability transition pores (mPTPs). The incidence of mitochondria SO flash activity in neural progenitor cells (NPCs) increased during the first 6-24 hours of exposure to aggregating amyloid β-peptide (Aβ1-42), indicating an increase in transient mPTP opening. Subsequently, the SO flash frequency progressively decreased and ceased between 48 and 72 hours of exposure to Aβ1-42, during which time global cellular reactive oxygen species increased, mitochondrial membrane potential decreased, cytochrome C was released from mitochondria and the cells degenerated. Inhibition of mPTPs and selective reduction in mitochondrial SO flashes significantly ameliorated the negative effects of Aβ1-42 on NPC proliferation and survival. Our findings suggest that mPTP-mediated bursts of mitochondrial SO production is a relatively early and pivotal event in the adverse effects of Aβ1-42 on NPCs. If Aβ inhibits NPC proliferation in the brains of AD patients by a similar mechanism, then interventions that inhibit mPTP-mediated superoxide flashes would be expected to protect NPCs against the adverse effects of Aβ.
Collapse
|
29
|
TrkB.T1 contributes to neuropathic pain after spinal cord injury through regulation of cell cycle pathways. J Neurosci 2013; 33:12447-63. [PMID: 23884949 DOI: 10.1523/jneurosci.0846-13.2013] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Spinal cord injury (SCI) frequently causes severe, persistent central neuropathic pain that responds poorly to conventional pain treatments. Brain-derived neurotrophic factor (BDNF) signaling appears to contribute to central sensitization and nocifensive behaviors in certain animal models of chronic pain through effects mediated in part by the alternatively spliced truncated isoform of the BDNF receptor tropomyosin-related kinase B.T1 (trkB.T1). Mechanisms linking trkB.T1 to SCI-induced chronic central pain are unknown. Here, we examined the role of trkB.T1 in central neuropathic pain after spinal cord contusion. Genetic deletion of trkB.T1 in mice significantly reduced post-SCI mechanical hyperesthesia, locomotor dysfunction, lesion volumes, and white matter loss. Whole genome analysis, confirmed at the protein level, revealed that cell cycle genes were upregulated in trkB.T1(+/+) but not trkB.T1(-/-) spinal cord after SCI. TGFβ-induced reactive astrocytes from WT mice showed increased cell cycle protein expression that was significantly reduced in astrocytes from trkB.T1(-/-) mice that express neither full-length trkB nor trkB.T1. Administration of CR8, which selectively inhibits cyclin-dependent kinases, reduced hyperesthesia, locomotor deficits, and dorsal horn (SDH) glial changes after SCI, similar to trkB.T1 deletion, without altering trkB.T1 protein expression. In trkB.T1(-/-) mice, CR8 had no effect. These data indicate that trkB.T1 contributes to the pathobiology of SCI and SCI pain through modulation of cell cycle pathways and suggest new therapeutic targets.
Collapse
|
30
|
Perreault ML, Fan T, O'Dowd BF, George SR. Enhanced brain-derived neurotrophic factor signaling in the nucleus accumbens of juvenile rats. Dev Neurosci 2013; 35:384-95. [PMID: 24021607 DOI: 10.1159/000351026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 03/28/2013] [Indexed: 11/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signaling through its receptor, tropomyosin receptor kinase B (TrkB), plays a critical role in neural plasticity and its dysregulation in striatum and prefrontal cortex (PFC) has been implicated in the etiology of mental health disorders such schizophrenia and drug addiction. In the present study, we characterized age-dependent differences in BDNF signaling and TrkB expression within the nucleus accumbens (NAc), caudate putamen (CP) and PFC in rats and determined the effects of administration of the dopamine agonist, SKF 83959, which activates the Gq-coupled dopamine receptors, the dopamine D5 receptor and the D1-D2 receptor heteromer. As proBDNF binds with high affinity to the p75 neurotrophin receptor (p75NTR), expression levels of these proteins were also assessed. The present findings showed that juvenile rats (aged 26-28 days) exhibited significantly elevated basal BDNF expression and activation of full-length TrkB (TrkBfull) in NAc compared to their adult counterparts, as evidenced by increased TrkBfull phosphorylation. These changes were concomitant with an increase in the relative expression of TrkBfull compared to the truncated isoform, TrkB.T1, in NAc and CP. Conversely, in PFC the basal expression of BDNF in juvenile rats was significantly lower than in adult rats with an elevated relative expression of TrkBfull. Acute administration of SKF 83959 to juvenile rats abolished the age-dependent differences in BDNF expression in NAc and PFC, and in the relative expression of TrkBfull in NAc and CP. Together these findings indicate that the expression and/or signaling of BDNF and TrkB in striatum and PFC of juvenile rats is fundamentally different from that of adult rats, a finding that may have implications in neuropsychiatric disorders that exhibit age-dependent susceptibility such as schizophrenia and drug addiction.
Collapse
Affiliation(s)
- Melissa L Perreault
- Centre for Addiction and Mental Health, Departments of Pharmacology, University of Toronto, Toronto, Ont., Canada
| | | | | | | |
Collapse
|
31
|
Hou Y, Ouyang X, Wan R, Cheng H, Mattson MP, Cheng A. Mitochondrial superoxide production negatively regulates neural progenitor proliferation and cerebral cortical development. Stem Cells 2013; 30:2535-47. [PMID: 22949407 DOI: 10.1002/stem.1213] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although high amounts of reactive oxygen species (ROS) can damage cells, ROS can also play roles as second messengers, regulating diverse cellular processes. Here, we report that embryonic mouse cerebral cortical neural progenitor cells (NPCs) exhibit intermittent spontaneous bursts of mitochondrial superoxide (SO) generation (mitochondrial SO flashes) that require transient opening of membrane permeability transition pores (mPTP). This quantal SO production negatively regulates NPC self-renewal. Mitochondrial SO scavengers and mPTP inhibitors reduce SO flash frequency and enhance NPC proliferation, whereas prolonged mPTP opening and SO generation increase SO flash incidence and decrease NPC proliferation. The inhibition of NPC proliferation by mitochondrial SO involves suppression of extracellular signal-regulated kinases. Moreover, mice lacking SOD2 (SOD2-/- mice) exhibit significantly fewer proliferative NPCs and differentiated neurons in the embryonic cerebral cortex at midgestation compared with wild-type littermates. Cultured SOD2-/- NPCs exhibit a significant increase in SO flash frequency and reduced NPC proliferation. Taken together, our findings suggest that mitochondrial SO flashes negatively regulate NPC self-renewal in the developing cerebral cortex.
Collapse
Affiliation(s)
- Yan Hou
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
32
|
Ying C, Hu W, Cheng B, Zheng X, Li S. Neural differentiation of rat adipose-derived stem cells in vitro. Cell Mol Neurobiol 2012; 32:1255-63. [PMID: 22569742 DOI: 10.1007/s10571-012-9850-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 04/17/2012] [Indexed: 01/23/2023]
Abstract
It is reported that adipose-derived stem cells (ADSCs) had multilineage differentiation potential, and could differentiate into neuron-like cells induced by special induction media, which may provide a new idea for restoration of erectile dysfunction (ED) after cavernous nerve injury. The aim of this research was to explore the neuronal differentiation potential of ADSCs in vitro. ADSCs isolated from inguinal adipose tissue of rat were characterized by flow cytometry, and results showed that ADSCs were positive for mesenchymal stem cell markers CD90 and CD44, but negative for hematopoietic stem cell markers. ADSCs maintained self-renewing capacity and could differentiate into adipocytes and neurocytes under special culture condition. In this research, two methods were used to induce ADSCs. In method 1, ADSCs were treated with the preinduction medium including epithelium growth factor, basic fibroblast growth factor, and brain derived neurotrophic factor (BDNF) for 3 days, then with the neurogenic induction medium containing isobutylmethylxanthine, indomethacin, and insulin. While in method 2, BDNF was not used to treat ADSCs. After induction, neuronal differentiation of ADSCs was evaluated. Neuronal markers, glial fibrillary acidic protein (GFAP), and β-tubulin III (Tuj-1) were detected by immunofluorescence and Western Blot analyses. The expressions of GFAP and Tuj-1 in method 1 were obviously higher then those in method 2. In addition, the positive rate of the neuron-like cells was higher in method 1. It suggested that ADSCs are able to differentiate into neural-like cells in vitro, and the administration of BDNF in the preinduction medium may provide a new way to modify the culture method for getting more neuron-like cells in vitro.
Collapse
Affiliation(s)
- Chengcheng Ying
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | | | | | | | | |
Collapse
|
33
|
Texel SJ, Camandola S, Ladenheim B, Rothman SM, Mughal MR, Unger EL, Cadet JL, Mattson MP. Ceruloplasmin deficiency results in an anxiety phenotype involving deficits in hippocampal iron, serotonin, and BDNF. J Neurochem 2011; 120:125-34. [PMID: 22035068 DOI: 10.1111/j.1471-4159.2011.07554.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ceruloplasmin (Cp) is a ferroxidase involved in iron metabolism by converting Fe(2+) to Fe(3+), and by regulating cellular iron efflux. In the ceruloplasmin knockout (CpKO) mouse, the deregulation of iron metabolism results in moderate liver and spleen hemosiderosis, but the impact of Cp deficiency on brain neurochemistry and behavior in this animal model is unknown. We found that in contrast to peripheral tissues, iron levels in the hippocampus are significantly reduced in CpKO mice. Although it does not cause any discernable deficits in motor function or learning and memory, Cp deficiency results in heightened anxiety-like behavior in the open field and elevated plus maze tests. This anxiety phenotype is associated with elevated levels of plasma corticosterone. Previous studies provided evidence that anxiety disorders and long-standing stress are associated with reductions in levels of serotonin (5HT) and brain-derived neurotrophic factor (BDNF) in the hippocampus. We found that levels of 5HT and norepinephrine (NE), and the expression of BDNF and its receptor trkB, are significantly reduced in the hippocampus of CpKO mice. Thus, Cp deficiency causes an anxiety phenotype by a mechanism that involves decreased levels of iron, 5HT, NE, and BDNF in the hippocampus.
Collapse
Affiliation(s)
- Sarah J Texel
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bath KG, Akins MR, Lee FS. BDNF control of adult SVZ neurogenesis. Dev Psychobiol 2011; 54:578-89. [PMID: 21432850 DOI: 10.1002/dev.20546] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 02/17/2011] [Indexed: 12/14/2022]
Abstract
The sensory processing of odorants is a dynamic process that requires plasticity at multiple levels. In the olfactory bulb (OB), inhibitory interneurons undergo lifelong replacement through a process known as adult neurogenesis. These newly born cells are incorporated in a learning-dependent fashion, a process which has led some to suggest this as a primary mechanism through which the OB retains a high degree of plasticity throughout life. A continued focus of researchers in this field has been to understand the molecular mechanisms controlling adult subventricular zone (SVZ) neurogenesis and the innate functional role of these cells. Brain-derived neurotrophic factor (BDNF) has been identified as a strong candidate molecule regulating adult OB neurogenesis. We review what is known regarding the functional role of newly born cells, highlight the role of BDNF in this process, and describe preliminary findings from our lab implicating BDNF in the process of selecting of newly born cells for survival.
Collapse
Affiliation(s)
- Kevin G Bath
- Department of Psychiatry, Weill Cornell Medical College, New York, New York 10065, USA.
| | | | | |
Collapse
|
35
|
Michaelsen K, Zagrebelsky M, Berndt-Huch J, Polack M, Buschler A, Sendtner M, Korte M. Neurotrophin receptors TrkB.T1 and p75NTR cooperate in modulating both functional and structural plasticity in mature hippocampal neurons. Eur J Neurosci 2010; 32:1854-65. [PMID: 20955473 DOI: 10.1111/j.1460-9568.2010.07460.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Tropomyosin-related kinase (Trk) receptors modulate neuronal structure and function both during development and in the mature nervous system. Interestingly, TrkB and TrkC are expressed as full-length and as truncated splice variants. The cellular function of the kinase-lacking isoforms remains so far unclear. We investigated the role of the truncated receptor TrkB.T1 in the hippocampus of transgenic mice overexpressing this splice variant by analyzing both neuronal structure and function. We observed an impairment in activity-dependent synaptic plasticity as indicated by deficits in long-term potentiation and long-term depression in acute hippocampal slices of transgenic TrkB.T1 mice. In addition, dendritic complexity and spine density were significantly altered in TrkB.T1-overexpressing CA1 neurons. We found that the effect of TrkB.T1 overexpression differs between subgroups of CA1 neurons. Remarkably, overexpression of p75(NTR) and its activation by chemical induction of long-term depression in slice cultures rescued the TrkB.T1-dependent morphological alterations specifically in one of the two subgroups observed. These findings suggest that the TrkB.T1 and p75(NTR) receptor signaling systems might be cross-linked. Our findings demonstrate that TrkB.T1 regulates the function and the structure of mature pyramidal neurons. In addition, we showed that the ratio of expression levels of p75(NTR) and TrkB.T1 plays an important role in modulating dendritic architecture and synaptic plasticity in the adult rodent hippocampus, and, indeed, that the endogenous expression patterns of both receptors change reciprocally over time. We therefore propose a new function of TrkB.T1 as being dominant-negative to p75(NTR).
Collapse
Affiliation(s)
- K Michaelsen
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Braunschweig, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Cheng A, Scott AL, Ladenheim B, Chen K, Ouyang X, Lathia JD, Mughal M, Cadet JL, Mattson MP, Shih JC. Monoamine oxidases regulate telencephalic neural progenitors in late embryonic and early postnatal development. J Neurosci 2010; 30:10752-62. [PMID: 20702706 PMCID: PMC2967387 DOI: 10.1523/jneurosci.2037-10.2010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 05/31/2010] [Accepted: 06/11/2010] [Indexed: 12/31/2022] Open
Abstract
Monoamine neurotransmitters play major roles in regulating a range of brain functions in adults and increasing evidence suggests roles for monoamines in brain development. Here we show that mice lacking the monoamine metabolic enzymes MAO A and MAO B (MAO AB-deficient mice) exhibit diminished proliferation of neural stem cells (NSC) in the developing telencephalon beginning in late gestation [embryonic day (E) 17.5], a deficit that persists in neonatal and adult mice. These mice showed significantly increased monoamine levels and anxiety-like behaviors as adults. Assessments of markers of intermediate progenitor cells (IPC) and mitosis showed that NSC in the subventricular zone (SVZ), but not in the ventricular zone, are reduced in MAO AB-deficient mice. A developmental time course of monoamines in frontal cortical tissues revealed increased serotonin levels as early as E14.5, and a further large increase was found between E17.5 and postnatal day 2. Administration of an inhibitor of serotonin synthesis (parachlorophenylalanine) between E14.5 and E19.5 restored the IPC numbers and SVZ thickness, suggesting the role of serotonin in the suppression of IPC proliferation. Studies of neurosphere cultures prepared from the telencephalon at different embryonic and postnatal ages showed that serotonin stimulates proliferation in wild-type, but not in MAO AB-deficient, NSC. Together, these results suggest that a MAO-dependent long-lasting alteration in the proliferation capacity of NSC occurs late in embryonic development and is mediated by serotonin. Our findings reveal novel roles for MAOs and serotonin in the regulation of IPC proliferation in the developing brain.
Collapse
Affiliation(s)
- Aiwu Cheng
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224
| | - Anna L. Scott
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, Baltimore, Maryland 21224
| | - Kevin Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089
| | - Xin Ouyang
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224
| | - Justin D. Lathia
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224
| | - Mohamed Mughal
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, Baltimore, Maryland 21224
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, and
| | - Jean C. Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
37
|
Abstract
Neurogenesis is the process by which cells divide, migrate, and subsequently differentiate into a neuronal phenotype. Significant rates of neurogenesis persist into adulthood in two brain regions, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) of the lateral ventricles. Cells of the SVZ divide and migrate via the rostral migratory stream (RMS) to the olfactory bulb (OB) where they differentiate into granule and periglomerular cells. With the discovery of large-scale neurogenesis in the adult brain, there have been significant efforts to identify the mechanisms that control this process as well as the role of these cells in neuronal functioning. Neurotrophic factors are a family of molecules that serve critical roles in the survival and differentiation of neurons during development, as well as contribute to continued plasticity throughout life. Several members of the neurotrophin family have been implicated in the control of adult postnatal SVZ neurogenesis. In this review we will address what is currently known regarding neurotrophic factor-dependent control of SVZ neurogenesis and place these findings in the context of what is known regarding other growth factors.
Collapse
Affiliation(s)
- Kevin G Bath
- Department of Psychiatry, Weill Cornell Medical College, New York, New York 10065, USA.
| | | |
Collapse
|
38
|
Gopalakrishna-Pillai S, Iverson LE. Astrocytes derived from trisomic human embryonic stem cells express markers of astrocytic cancer cells and premalignant stem-like progenitors. BMC Med Genomics 2010; 3:12. [PMID: 20423517 PMCID: PMC2873256 DOI: 10.1186/1755-8794-3-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 04/27/2010] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Trisomic variants of human embryonic stem cells (hESCs) arise spontaneously in culture. Although trisomic hESCs share many properties with diploid hESCs, they also exhibit features of cancer stem cells. Since most hESC-based therapies will utilize differentiated derivatives, it is imperative to investigate the potential of trisomic hESCs to undergo malignant transformation during differentiation prior to their use in the clinical setting. METHODS Diploid and trisomic hESCs were differentiated into astrocytic progenitors cells (APCs), RNA extracted and hybridized to human exon-specific microarrays. Global gene expression profiles of diploid and trisomic APCs were compared to that of an astrocytoma cell line and glioblastoma samples, analyzed by others, using the same microarray platform. RESULTS Bioinformatic analysis of microarray data indicates that differentiated trisomic APCs exhibit global expression profiles with similarities to the malignant astrocytoma cell line. An analogous trend is observed in comparison to glioblastoma samples indicating that trisomic APCs express markers of astrocytic cancer cells. The analysis also allowed identification of transcripts predicted to be differentially expressed in brain tumor stem cells. These data indicate that in vitro differentiation of trisomic hESCs along astrocytic pathways give rise to cells exhibiting properties of premalignant astrocytic stem/progenitor cells. CONCLUSIONS Given their occult nature, opportunities to study premalignant stem/progenitor cells in human have been few. The ability to propagate and direct the differentiation of aneuploid hESCs provides a powerful in vitro system for investigating biological properties of human cells exhibiting features of premalignant stem cells. This in vitro culture system can be used to elucidate changes in gene expression occurring enroute to malignant transformation and to identify molecular markers of cancer stem/progenitor cells. These markers are invaluable for diagnostic purposes and may be novel targets for therapeutic intervention.
Collapse
|
39
|
Ahn JI, Kim SY, Ko MJ, Chung HJ, Jeong HS. Analysis of Gene Expression in Mouse Spinal Cord-derived Neural Precursor Cells During Neuronal Differentiation. Genomics Inform 2009. [DOI: 10.5808/gi.2009.7.2.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
40
|
Functional and evolutionary insights into human brain development through global transcriptome analysis. Neuron 2009; 62:494-509. [PMID: 19477152 DOI: 10.1016/j.neuron.2009.03.027] [Citation(s) in RCA: 431] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 02/05/2009] [Accepted: 03/17/2009] [Indexed: 01/09/2023]
Abstract
Our understanding of the evolution, formation, and pathological disruption of human brain circuits is impeded by a lack of comprehensive data on the developing brain transcriptome. A whole-genome, exon-level expression analysis of 13 regions from left and right sides of the mid-fetal human brain revealed that 76% of genes are expressed, and 44% of these are differentially regulated. These data reveal a large number of specific gene expression and alternative splicing patterns, as well as coexpression networks, associated with distinct regions and neurodevelopmental processes. Of particular relevance to cognitive specializations, we have characterized the transcriptional landscapes of prefrontal cortex and perisylvian speech and language areas, which exhibit a population-level global expression symmetry. We show that differentially expressed genes are more frequently associated with human-specific evolution of putative cis-regulatory elements. These data provide a wealth of biological insights into the complex transcriptional and molecular underpinnings of human brain development and evolution.
Collapse
|
41
|
Luo Y, Lathia J, Mughal M, Mattson MP. SDF1alpha/CXCR4 signaling, via ERKs and the transcription factor Egr1, induces expression of a 67-kDa form of glutamic acid decarboxylase in embryonic hippocampal neurons. J Biol Chem 2008; 283:24789-800. [PMID: 18606818 PMCID: PMC2529007 DOI: 10.1074/jbc.m800649200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 06/06/2008] [Indexed: 02/04/2023] Open
Abstract
Stromal cell-derived factor alpha (SDF1alpha) and its cognate receptor CXCR4 play an important role in neuronal development in the hippocampus, but the genes directly regulated by SDF1alpha/CXCR4 signaling are unknown. To study the role of CXCR4 targeted genes in neuronal development, we used neuronal cultures established from embryonic day 18 rats. Hippocampal neurons express CXCR4 receptor proteins and are stimulated by SDF1alpha resulting in activation of extracellular signal-regulated kinase (ERK)1/2 and the transcription factor cAMP-response element-binding protein. SDF1alpha rapidly induces the expression of the early growth response gene Egr1, a transcription factor involved in activity-dependent neuronal responses, in a concentration-dependent manner. Gel-shift analysis showed that SDF1alpha enhances DNA binding activity to the Egr1-containing promoter for GAD67. Chromatin immunoprecipitation analysis using an Egr1 antibody indicated that SDF1alpha stimulation increases binding of Egr1 to a GAD67 promoter DNA sequence. SDF1alpha stimulation increases the expression of GAD67 at both the mRNA and protein levels, and increases the amount and neurite localization of gamma-aminobutyric acid (GABA) in neurons already expressing GABA. SDF1alpha-induced Egr1/GAD67 expression is mediated by the G protein-coupled CXCR4 receptor and activation of the ERK pathway. Reduction of Egr1 gene expression using small interfering RNA technology lowers the level of GAD67 transcripts and inhibits SDF1alpha-induced GABA production. Inhibition of CXCR4 activation in the developing mouse brain in utero greatly reduced Egr1 and GAD67 mRNA levels and GAD67 protein levels, suggesting a pivotal role for CXCR4 signaling in the development of GABAergic neurons in vivo. Our data suggest that SDF1alpha/CXCR4/G protein/ERK signaling induces the expression of the GAD67 system via Egr1 activation, a mechanism that may promote the maturation of GABAergic neurons during development.
Collapse
MESH Headings
- Animals
- Chemokine CXCL12/metabolism
- Chemokine CXCL12/pharmacology
- Dose-Response Relationship, Drug
- Early Growth Response Protein 1/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/enzymology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/physiology
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Glutamate Decarboxylase/biosynthesis
- Hippocampus/embryology
- Hippocampus/enzymology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Mice
- Neurites/enzymology
- Pregnancy
- RNA, Messenger/biosynthesis
- RNA, Small Interfering/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, CXCR4/agonists
- Receptors, CXCR4/metabolism
- Response Elements/physiology
- gamma-Aminobutyric Acid/biosynthesis
Collapse
Affiliation(s)
| | | | | | - Mark P. Mattson
- Laboratory of Neurosciences, NIA Intramural Research Program, National
Institutes of Health, Baltimore, Maryland 21224
| |
Collapse
|