1
|
Ghaffari LT, Welebob E, Boehringer A, Cyliax K, Pasinelli P, Trotti D, Haeusler AR. Neuronal Activity-Dependent Gene Dysregulation in C9orf72 i 3Neuronal Models of ALS/FTD Pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.632228. [PMID: 39975241 PMCID: PMC11838197 DOI: 10.1101/2025.01.27.632228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The GGGGCC nucleotide repeat expansion (NRE) mutation in the C9orf72 (C9) gene is the most common cause of ALS and FTD. Neuronal activity plays an essential role in shaping biological processes within both healthy and neurodegenerative disease scenarios. Here, we show that at baseline conditions, C9-NRE iPSC-cortical neurons display aberrations in several pathways, including synaptic signaling and transcriptional machinery, potentially priming diseased neurons for an altered response to neuronal stimulation. Indeed, exposure to two pathophysiologically relevant stimulation modes, prolonged membrane depolarization, or a blockade of K+ channels, followed by RNA sequencing, induces a temporally divergent activity-dependent transcriptome of C9-NRE cortical neurons compared to healthy controls. This study provides new insights into how neuronal activity influences the ALS/FTD-associated transcriptome, offering a dataset that enables further exploration of pathways necessary for conferring neuronal resilience or degeneration.
Collapse
Affiliation(s)
- Layla T. Ghaffari
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Emily Welebob
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ashley Boehringer
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kelly Cyliax
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aaron R. Haeusler
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
2
|
Hu H, Liu Y, Qiu C, Zhang L, Cui H, Gu J. LINC00894 inhibited neuron cellular apoptosis and regulated activating transcription factor 3 expression. Gene 2024; 927:148670. [PMID: 38857714 DOI: 10.1016/j.gene.2024.148670] [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: 02/29/2024] [Revised: 06/01/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
LINC00894 may be associated with synaptic function, but its biology function in neural cells is still unknown. In this study, LINC00894 knockdown decreased the EdU incorporated into newly synthesized DNA and cell viability in MTT or CCK-8 assay in HEK-293T and BE(2)-M17 (M17) neuroblastoma cells. And LINC00894 knockdown increased cellular apoptosis in Annexin V-FITC staining, the expression of activated Caspase3 and the level of reactive oxygen species (ROS) both in HEK-293T and M17 cells. Moreover, LINC00894 also protected cells from hydrogen peroxide induced apoptosis in in vitro models. Utilizing RNA sequencing (RNA-seq) integrated with quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunoblot, we identified that LINC00894 affected activating transcription factor 3 (ATF3) expression in HEK-293T, M17, and SH-SY5Y neuroblastoma cells. Finally, we found that ectopic expression of ATF3 restored cell proliferation and inhibited cell apoptosis in LINC00894 downregulated M17 cells. While knockdown of ATF3 also significantly increased the cell viability inhibition and apoptosis promotion induced by LINC00894 knockdown in M17 cells. Our results from in vitro models revealed that LINC00894 could promote neuronal cell proliferation and inhibit cellular apoptosis by affecting ATF3 expression.
Collapse
Affiliation(s)
- Hanjing Hu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Yuxiao Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Cheng Qiu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Liti Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Hengxiang Cui
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianlan Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
3
|
Hou Y, Sun L, LaFleur MW, Huang L, Lambden C, Thakore PI, Geiger-Schuller K, Kimura K, Yan L, Zang Y, Tang R, Shi J, Barilla R, Deng L, Subramanian A, Wallrapp A, Choi HS, Kye YC, Ashenberg O, Schiebinger G, Doench JG, Chiu IM, Regev A, Sharpe AH, Kuchroo VK. Neuropeptide signalling orchestrates T cell differentiation. Nature 2024; 635:444-452. [PMID: 39415015 PMCID: PMC11951087 DOI: 10.1038/s41586-024-08049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
The balance between T helper type 1 (TH1) cells and other TH cells is critical for antiviral and anti-tumour responses1-3, but how this balance is achieved remains poorly understood. Here we dissected the dynamic regulation of TH1 cell differentiation during in vitro polarization, and during in vivo differentiation after acute viral infection. We identified regulators modulating T helper cell differentiation using a unique TH1-TH2 cell dichotomous culture system and systematically validated their regulatory functions through multiple in vitro and in vivo CRISPR screens. We found that RAMP3, a component of the receptor for the neuropeptide CGRP (calcitonin gene-related peptide), has a cell-intrinsic role in TH1 cell fate determination. Extracellular CGRP signalling through the receptor RAMP3-CALCRL restricted the differentiation of TH2 cells, but promoted TH1 cell differentiation through the activation of downstream cAMP response element-binding protein (CREB) and activating transcription factor 3 (ATF3). ATF3 promoted TH1 cell differentiation by inducing the expression of Stat1, a key regulator of TH1 cell differentiation. After viral infection, an interaction between CGRP produced by neurons and RAMP3 expressed on T cells enhanced the anti-viral IFNγ-producing TH1 and CD8+ T cell response, and timely control of acute viral infection. Our research identifies a neuroimmune circuit in which neurons participate in T cell fate determination by producing the neuropeptide CGRP during acute viral infection, which acts on RAMP3-expressing T cells to induce an effective anti-viral TH1 cell response.
Collapse
Affiliation(s)
- Yu Hou
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Liangzhu Laboratory of Zhejiang University, Zhejiang University School of Medicine, Hangzhou, China
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Linyu Sun
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Martin W LaFleur
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Linglin Huang
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Conner Lambden
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Kimitoshi Kimura
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Longjun Yan
- Liangzhu Laboratory of Zhejiang University, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Zang
- Liangzhu Laboratory of Zhejiang University, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruihan Tang
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jingwen Shi
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Rocky Barilla
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Liwen Deng
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Ayshwarya Subramanian
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Antonia Wallrapp
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Hee Sun Choi
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Yoon-Chul Kye
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Orr Ashenberg
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Geoffrey Schiebinger
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - John G Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Genentech, South San Francisco, CA, USA.
| | - Arlene H Sharpe
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital and Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
4
|
Kang L, Piao M, Liu N, Gu W, Feng C. Sevoflurane Exposure Induces Neuronal Cell Ferroptosis Initiated by Increase of Intracellular Hydrogen Peroxide in the Developing Brain via ER Stress ATF3 Activation. Mol Neurobiol 2024; 61:2313-2335. [PMID: 37874483 DOI: 10.1007/s12035-023-03695-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/04/2023] [Indexed: 10/25/2023]
Abstract
Neuronal cell death is acknowledged as the primary pathological basis underlying developmental neurotoxicity in response to sevoflurane exposure, but the exact mechanism remains unclear. Ferroptosis is a form of programmed cell death characterized by iron-dependent lipid peroxidation that is driven by hydrogen peroxide (H2O2) and ferrous iron through the Fenton reaction and participates in the pathogenesis of multiple neurological diseases. As stress response factor, activating transcription factor 3 (ATF3) can be activated by the PERK/ATF4 pathway during endoplasmic reticulum (ER) stress, followed by increased intracellular H2O2, which is involved in regulation of apoptosis, autophagy, and ferroptosis. Here, we investigated whether ferroptosis and ATF3 activation were implicated in sevoflurane-induced neuronal cell death in the developing brain. The results showed that sevoflurane exposure induced neuronal death as a result of iron-dependent lipid peroxidation damage secondary to H2O2 accumulation and ferrous iron increase, which was consistent with the criteria for ferroptosis. Furthermore, we observed that increases in iron and H2O2 induced by sevoflurane exposure were associated with the upregulation and nuclear translocation of ATF3 in response to ER stress. Knockdown of ATF3 expression alleviated iron-dependent lipid peroxidation, which prevented sevoflurane-induced neuronal ferroptosis. Mechanistically, ATF3 promoted sevoflurane-induced H2O2 accumulation by activating NOX4 and suppressing catalase, GPX4, and SLC7A11 expression. Additionally, an increase in H2O2 was accompanied by the upregulation of TFR and TF and downregulation of FPN, which linked iron overload to ferroptosis induced by sevoflurane. Taken together, our results demonstrated that ER stress-mediated ATF3 activation contributed to sevoflurane-induced neuronal ferroptosis via H2O2 accumulation and the resultant iron overload.
Collapse
Affiliation(s)
- Liheng Kang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Nan Liu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Wanping Gu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Chunsheng Feng
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China.
| |
Collapse
|
5
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
6
|
Sharma R, Neupane C, Pham TL, Lee M, Lee S, Lee SY, Nam MH, Kim CS, Park JB. Tonic Activation of NR2D-Containing NMDARs Exacerbates Dopaminergic Neuronal Loss in MPTP-Injected Parkinsonian Mice. J Neurosci 2023; 43:7730-7744. [PMID: 37726169 PMCID: PMC10648527 DOI: 10.1523/jneurosci.1955-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
NR2D subunit-containing NMDA receptors (NMDARs) gradually disappear during brain maturation but can be recruited by pathophysiological stimuli in the adult brain. Here, we report that 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxication recruited NR2D subunit-containing NMDARs that generated an Mg2+-resistant tonic NMDA current (INMDA) in dopaminergic (DA) neurons in the midbrain of mature male mice. MPTP selectively generated an Mg2+-resistant tonic INMDA in DA neurons in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA). Consistently, MPTP increased NR2D but not NR2B expression in the midbrain regions. Pharmacological or genetic NR2D interventions abolished the generation of Mg2+-resistant tonic INMDA in SNpc DA neurons, and thus attenuated subsequent DA neuronal loss and gait deficits in MPTP-treated mice. These results show that extrasynaptic NR2D recruitment generates Mg2+-resistant tonic INMDA and exacerbates DA neuronal loss, thus contributing to MPTP-induced Parkinsonism. The state-dependent NR2D recruitment could be a novel therapeutic target for mitigating cell type-specific neuronal death in neurodegenerative diseases.SIGNIFICANCE STATEMENT NR2D subunit-containing NMDA receptors (NMDARs) are widely expressed in the brain during late embryonic and early postnatal development, and then downregulated during brain maturation and preserved at low levels in a few regions of the adult brain. Certain stimuli can recruit NR2D subunits to generate tonic persistent NMDAR currents in nondepolarized neurons in the mature brain. Our results show that MPTP intoxication recruits NR2D subunits in midbrain dopaminergic (DA) neurons, which leads to tonic NMDAR current-promoting dopaminergic neuronal death and consequent abnormal gait behavior in the MPTP mouse model of Parkinson's disease (PD). This is the first study to indicate that extrasynaptic NR2D recruitment could be a target for preventing neuronal death in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ramesh Sharma
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Chiranjivi Neupane
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Thuy Linh Pham
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Miae Lee
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Cuk-Seong Kim
- Department of Biomedicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Physiology, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin Bong Park
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08852, Republic of Korea
| |
Collapse
|
7
|
Reiners JC, Leopold L, Hallebach V, Sinske D, Meier P, Amoroso M, Langgartner D, Reber SO, Knöll B. Acute stress modulates the outcome of traumatic brain injury-associated gene expression and behavioral responses. FASEB J 2023; 37:e23218. [PMID: 37779443 DOI: 10.1096/fj.202301035r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023]
Abstract
Psychological stress and traumatic brain injury (TBI) result in long-lasting emotional and behavioral impairments in patients. So far, the interaction of psychological stress with TBI not only in the brain but also in peripheral organs is poorly understood. Herein, the impact of acute stress (AS) occurring immediately before TBI is investigated. For this, a mouse model of restraint stress and TBI was employed, and their influence on behavior and gene expression in brain regions, the hypothalamic-pituitary-adrenal (HPA) axis, and peripheral organs was analyzed. Results demonstrate that, compared to single AS or TBI exposure, mice treated with AS prior to TBI showed sex-specific alterations in body weight, memory function, and locomotion. The induction of immediate early genes (IEGs, e.g., c-Fos) by TBI was modulated by previous AS in several brain regions. Furthermore, IEG upregulation along the HPA axis (e.g., pituitary, adrenal glands) and other peripheral organs (e.g., heart) was modulated by AS-TBI interaction. Proteomics of plasma samples revealed proteins potentially mediating this interaction. Finally, the deletion of Atf3 diminished the TBI-induced induction of IEGs in peripheral organs but left them largely unaltered in the brain. In summary, AS immediately before brain injury affects the brain and, to a strong degree, also responses in peripheral organs.
Collapse
Affiliation(s)
| | - Laura Leopold
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Vera Hallebach
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Daniela Sinske
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Philip Meier
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Mattia Amoroso
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Bernd Knöll
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Liao W, Wen Y, Yang S, Duan Y, Liu Z. Research progress and perspectives of N-methyl-D-aspartate receptor in myocardial and cerebral ischemia-reperfusion injury: A review. Medicine (Baltimore) 2023; 102:e35490. [PMID: 37861505 PMCID: PMC10589574 DOI: 10.1097/md.0000000000035490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/13/2023] [Indexed: 10/21/2023] Open
Abstract
There is an urgent need to find common targets for precision therapy, as there are no effective preventive therapeutic measures for combined clinical heart-brain organ protection and common pathways associated with glutamate receptors are involved in heart-brain injury, but current glutamate receptor-related clinical trials have failed. Ischemia-reperfusion injury (IRI) is a common pathological condition that occurs in multiple organs, including the heart and brain, and can lead to severe morbidity and mortality. N-methyl-D-aspartate receptor (NMDAR), a type of ionotropic glutamate receptor, plays a crucial role in the pathogenesis of IRI. NMDAR activity is mainly regulated by endogenous activators, agonists, antagonists, and voltage-gated channels, and activation leads to excessive calcium influx, oxidative stress, mitochondrial dysfunction, inflammation, apoptosis, and necrosis in ischemic cells. In this review, we summarize current research advances regarding the role of NMDAR in myocardial and cerebral IRI and discuss potential therapeutic strategies to modulate NMDAR signaling to prevent and treat IRI.
Collapse
Affiliation(s)
- Wei Liao
- Department of Neurosurgery, First Affiliated of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yuehui Wen
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaochun Yang
- Department of Neurosurgery, First Affiliated of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yanyu Duan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- Heart Medical Centre, First Affiliated of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Ziyou Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- Heart Medical Centre, First Affiliated of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Cardiac Surgery, First Affiliated of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
9
|
Wang Y, Hong Q, Xia Y, Zhang Z, Wen B. The Lysine Demethylase KDM7A Regulates Immediate Early Genes in Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301367. [PMID: 37565374 PMCID: PMC10558696 DOI: 10.1002/advs.202301367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/11/2023] [Indexed: 08/12/2023]
Abstract
Lysine demethylase KDM7A removes histone modifications H3K9me1/2 and H3K27me1/2. KDM7A plays critical roles in gene expression and contribute to biological processes including tumorigenesis, metabolism, and embryonic development. However, the functions of KDM7A in mammalian nervous system are still poorly explored. In this study, functional roles of KDM7A are comprehensively investigated in neuronal cells by applying CUT&Tag-seq, RNA-seq and mice models. Knockdown of Kdm7a in N2A cells result in the alteration of histone modifications near transcription start sites (TSSs) and the expression changes of a large number of genes. In particular, the expression of immediate early genes (IEGs), a series of genes maintaining the function of the nervous system and associating with neurological disorders, are significantly decreased upon Kdm7a knockdown. Furthermore, in vivo knockdown of Kdm7a in dentate gyrus (DG) neuron of mice hippocampus, via Adeno-associated virus (AAV)-based stereotaxic microinjection, led to a significant decrease of the expression of c-Fos, a marker of neuron activity. Behavior assays in mice further revealed that Kdm7a knockdown in hippocampus repress neuron activity, which leading to impairment of emotion and memory. Collectively, the study reveals that KDM7A affects neuron functions by regulating IEGs, which may provide new clues for understanding epigenetic mechanisms in neurological disorders.
Collapse
Affiliation(s)
- Yifan Wang
- Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesFudan University200032130 Dong An RoadShanghaiChina
| | - Qin Hong
- Shengli Clinical Medical College of Fujian Medical University, Center for Experimental Research in Clinical MedicineFujian Provincial Hospital134 East StreetFuzhou350001China
| | - Yueyue Xia
- Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesFudan University200032130 Dong An RoadShanghaiChina
| | - Zhao Zhang
- Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesFudan University200032130 Dong An RoadShanghaiChina
| | - Bo Wen
- Key Laboratory of Metabolism and Molecular Medicine of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesFudan University200032130 Dong An RoadShanghaiChina
| |
Collapse
|
10
|
Siwakoti B, Lien TS, Lin YY, Pethaperumal S, Hung SC, Sun DS, Cheng CF, Chang HH. The Role of Activating Transcription Factor 3 in Metformin's Alleviation of Gastrointestinal Injury Induced by Restraint Stress in Mice. Int J Mol Sci 2023; 24:10995. [PMID: 37446172 DOI: 10.3390/ijms241310995] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Metformin is one of the most commonly used drugs for type 2 diabetes mellitus. In addition to its anti-diabetic property, evidence suggests more potential applications for metformin, such as antiaging, cellular protection, and anti-inflammation. Studies have reported that metformin activates pathways with anti-inflammatory effects, enhances the integrity of gut epithelial tight junctions, and promotes a healthy gut microbiome. These actions contribute to the protective effect of metformin against gastrointestinal (GI) tract injury. However, whether metformin plays a protective role in psychological-stress-associated GI tract injury remains elusive. We aim to elucidate the potential protective effect of metformin on the GI system and develop an effective intervention strategy to counteract GI injury induced by acute psychological stress. By monitoring the levels of GI-nonabsorbable Evans blue dye in the bloodstream, we assessed the progression of GI injury in live mice. Our findings demonstrate that the administration of metformin effectively mitigated GI leakage caused by psychological stress. The GI protective effect of metformin is more potent when used on wild-type mice than on activating-transcription-factor 3 (ATF3)-deficient (ATF3-/-) mice. As such, metformin-mediated rescue was conducted in an ATF3-dependent manner. In addition, metformin-mediated protection is associated with the induction of stress-induced GI mRNA expressions of the stress-induced genes ATF3 and AMP-activated protein kinase. Furthermore, metformin treatment-mediated protection of CD326+ GI epithelial cells against stress-induced apoptotic cell death was observed in wild-type but not in ATF3-/- mice. These results suggest that metformin plays a protective role in stress-induced GI injury and that ATF3 is an essential regulator for metformin-mediated rescue of stress-induced GI tract injury.
Collapse
Affiliation(s)
- Bijaya Siwakoti
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 97004, Taiwan
| | - Te-Sheng Lien
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 97004, Taiwan
| | - You-Yen Lin
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 97004, Taiwan
| | - Subhashree Pethaperumal
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 97004, Taiwan
| | - Shih-Che Hung
- Institute of Medical Sciences, Tzu-Chi University, Hualien 97004, Taiwan
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 97004, Taiwan
- Institute of Medical Sciences, Tzu-Chi University, Hualien 97004, Taiwan
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 23142, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 97004, Taiwan
- Institute of Medical Sciences, Tzu-Chi University, Hualien 97004, Taiwan
| |
Collapse
|
11
|
Srakočić S, Gorup D, Kutlić D, Petrović A, Tarabykin V, Gajović S. Reactivation of corticogenesis-related transcriptional factors BCL11B and SATB2 after ischemic lesion of the adult mouse brain. Sci Rep 2023; 13:8539. [PMID: 37237015 DOI: 10.1038/s41598-023-35515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The aim of this study was to characterize expression of corticogenesis-related transcription factors BCL11B and SATB2 after brain ischemic lesion in the adult mice, and to analyze their correlation to the subsequent brain recovery. Ischemic brain lesion was induced by transient middle cerebral artery occlusion followed by reperfusion, and the animals with ischemic lesion were compared to the sham controls. Progression of the brain damage and subsequent recovery was longitudinally monitored structurally, by magnetic resonance imaging, and functionally, by neurological deficit assessment. Seven days after the ischemic injury the brains were isolated and analyzed by immunohistochemistry. The results showed higher expression in the brain of both, BCL11B and SATB2 in the animals with ischemic lesion compared to the sham controls. The co-expression of both markers, BCL11B and SATB2, increased in the ischemic brains, as well as the co-expression of BCL11B with the beneficial transcriptional factor ATF3 but not its co-expression with detrimental HDAC2. BCL11B was mainly implicated in the ipsilateral and SATB2 in the contralateral brain hemisphere, and their level in these regions correlated with the functional recovery rate. The results indicate that the reactivation of corticogenesis-related transcription factors BCL11B and SATB2 is beneficial after brain ischemic lesion.
Collapse
Affiliation(s)
- Sanja Srakočić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Dunja Gorup
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
- Universität Zürich, Universitätspital Zürich, Zürich, Switzerland
| | - Dominik Kutlić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Ante Petrović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, Berlin, Germany
- Institute of Neuroscience, University of Nizhny Novgorod, Pr. Gagarina 24, Nizhny Novgorod, Russia
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, 10000, Zagreb, Croatia.
| |
Collapse
|
12
|
Activating Transcription Factor 3 Diminishes Ischemic Cerebral Infarct and Behavioral Deficit by Downregulating Carboxyl-Terminal Modulator Protein. Int J Mol Sci 2023; 24:ijms24032306. [PMID: 36768628 PMCID: PMC9917101 DOI: 10.3390/ijms24032306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Activating transcription factor 3 (ATF3) is a stress-induced transcription factor and a familiar neuronal marker for nerve injury. This factor has been shown to protect neurons from hypoxic insult in vitro by suppressing carboxyl-terminal modulator protein (CTMP) transcription, and indirectly activating the anti-apoptotic Akt/PKB cascade. Despite prior studies in vitro, whether this neuroprotective pathway also exists in the brain in vivo after ischemic insult remains to be determined. In the present study, we showed a rapid and marked induction of ATF3 mRNA throughout ischemia-reperfusion in a middle cerebral artery (MCA) occlusion model. Although the level of CTMP mRNA was quickly induced upon ischemia, its level showed only a mild increase after reperfusion. With the gain-of-function approach, both pre- and post-ischemic administration of Ad-ATF3 ameliorated brain infarct and neurological deficits. Whereas, with the loss-of-function approach, ATF3 knockout (KO) mice showed bigger infarct and worse functional outcome after ischemia. In addition, these congenital defects were rescued upon reintroducing ATF3 to the brain of KO mice. ATF3 overexpression led to a lower level of CTMP and a higher level of p-Akt(473) in the ischemic brain. On the contrary, ATF3 KO resulted in upregulation of CTMP and downregulation of p-Akt(473) instead. Furthermore, post-ischemic CTMP siRNA knockdown led to smaller infarct and better behaviors. CTMP siRNA knockdown increased the level of p-Akt(473), but did not alter the ATF3 level in the ischemic brain, upholding the ATF3→CTMP signal cascade. In summary, our proof-of-principle experiments support the existence of neuroprotective ATF3→CTMP signal cascade regulating the ischemic brain. Furthermore, these results suggest the therapeutic potential for both ATF3 overexpression and CTMP knockdown for stroke treatment.
Collapse
|
13
|
Fröhlich A, Olde Heuvel F, Rehman R, Krishnamurthy SS, Li S, Li Z, Bayer D, Conquest A, Hagenston AM, Ludolph A, Huber-Lang M, Boeckers T, Knöll B, Morganti-Kossmann MC, Bading H, Roselli F. Neuronal nuclear calcium signaling suppression of microglial reactivity is mediated by osteoprotegerin after traumatic brain injury. J Neuroinflammation 2022; 19:279. [PMCID: PMC9675197 DOI: 10.1186/s12974-022-02634-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Background Traumatic brain injury (TBI) is characterized by massive changes in neuronal excitation, from acute excitotoxicity to chronic hyper- or hypoexcitability. Nuclear calcium signaling pathways are involved in translating changes in synaptic inputs and neuronal activity into discrete transcriptional programs which not only affect neuronal survival and synaptic integrity, but also the crosstalk between neurons and glial cells. Here, we report the effects of blunting neuronal nuclear calcium signals in the context of TBI. Methods We used AAV vectors to express the genetically encoded and nuclear-targeted calcium buffer parvalbumin (PV.NLS.mCherry) or the calcium/calmodulin buffer CaMBP4.mCherry in neurons only. Upon TBI, the extent of neuroinflammation, neuronal death and synaptic loss were assessed by immunohistochemistry and targeted transcriptome analysis. Modulation of the overall level of neuronal activity was achieved by PSAM/PSEM chemogenetics targeted to parvalbumin interneurons. The functional impact of neuronal nuclear calcium buffering in TBI was assessed by quantification of spontaneous whisking. Results Buffering neuronal nuclear calcium unexpectedly resulted in a massive and long-lasting increase in the recruitment of reactive microglia to the injury site, which was characterized by a disease-associated and phagocytic phenotype. This effect was accompanied by a substantial surge in synaptic loss and significantly reduced whisking activity. Transcriptome analysis revealed a complex effect of TBI in the context of neuronal nuclear calcium buffering, with upregulation of complement factors, chemokines and interferon-response genes, as well as the downregulation of synaptic genes and epigenetic regulators compared to control conditions. Notably, nuclear calcium buffering led to a substantial loss in neuronal osteoprotegerin (OPG), whereas stimulation of neuronal firing induced OPG expression. Viral re-expression of OPG resulted in decreased microglial recruitment and synaptic loss. OPG upregulation was also observed in the CSF of human TBI patients, underscoring its translational value. Conclusion Neuronal nuclear calcium signals regulate the degree of microglial recruitment and reactivity upon TBI via, among others, osteoprotegerin signals. Our findings support a model whereby neuronal activity altered after TBI exerts a powerful impact on the neuroinflammatory cascade, which in turn contributes to the overall loss of synapses and functional impairment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02634-4.
Collapse
Affiliation(s)
- Albrecht Fröhlich
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany
| | - Florian Olde Heuvel
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany
| | - Rida Rehman
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany
| | - Sruthi Sankari Krishnamurthy
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany ,CEMMA (Cellular and Molecular Mechanisms in Aging) Research Training Group, Ulm, Germany
| | - Shun Li
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany
| | - Zhenghui Li
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany ,Dept. of Neurosurgery, Kaifeng Central Hospital, Kaifeng, China
| | - David Bayer
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany ,CEMMA (Cellular and Molecular Mechanisms in Aging) Research Training Group, Ulm, Germany
| | - Alison Conquest
- grid.1623.60000 0004 0432 511XNational Trauma Research Institute and Department of Neurosurgery, The Alfred Hospital, Melbourne, Australia
| | - Anna M. Hagenston
- grid.7700.00000 0001 2190 4373Interdisciplinary Center for Neurosciences, Department of Neurobiology, Heidelberg University, Heidelberg, Germany
| | - Albert Ludolph
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Markus Huber-Lang
- grid.6582.90000 0004 1936 9748Institute for Clinical and Experimental Trauma Immunology, Ulm University, Ulm, Germany
| | - Tobias Boeckers
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany ,grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Bernd Knöll
- grid.6582.90000 0004 1936 9748Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Maria Cristina Morganti-Kossmann
- grid.1623.60000 0004 0432 511XNational Trauma Research Institute and Department of Neurosurgery, The Alfred Hospital, Melbourne, Australia ,grid.134563.60000 0001 2168 186XDepartment of Child Health, Barrow Neurological Institute at Phoenix Children’s Hospital, University of Arizona College of Medicine, Phoenix, Phoenix, AZ USA
| | - Hilmar Bading
- grid.7700.00000 0001 2190 4373Interdisciplinary Center for Neurosciences, Department of Neurobiology, Heidelberg University, Heidelberg, Germany
| | - Francesco Roselli
- grid.6582.90000 0004 1936 9748Dept. of Neurology, Ulm University, Ulm, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany ,Present Address: Center for Biomedical Research, Helmholtzstrasse 8, 89081 Ulm, Germany
| |
Collapse
|
14
|
Targeting NMDA Receptors in Emotional Disorders: Their Role in Neuroprotection. Brain Sci 2022; 12:brainsci12101329. [PMID: 36291261 PMCID: PMC9599159 DOI: 10.3390/brainsci12101329] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 12/03/2022] Open
Abstract
Excitatory glutamatergic neurotransmission mediated through N-methyl-D-Aspartate (NMDA) receptors (NMDARs) is essential for synaptic plasticity and neuronal survival. While under pathological states, abnormal NMDAR activation is involved in the occurrence and development of psychiatric disorders, which suggests a directional modulation of NMDAR activity that contributes to the remission and treatment of psychiatric disorders. This review thus focuses on the involvement of NMDARs in the pathophysiological processes of psychiatric mood disorders and analyzes the neuroprotective mechanisms of NMDARs. Firstly, we introduce NMDAR-mediated neural signaling pathways in brain function and mood regulation as well as the pathophysiological mechanisms of NMDARs in emotion-related mental disorders such as anxiety and depression. Then, we provide an in-depth summary of current NMDAR modulators that have the potential to be developed into clinical drugs and their pharmacological research achievements in the treatment of anxiety and depression. Based on these findings, drug-targeting for NMDARs might open up novel territory for the development of therapeutic agents for refractory anxiety and depression.
Collapse
|
15
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
16
|
Amin D, Kuwajima T. Differential Retinal Ganglion Cell Vulnerability, A Critical Clue for the Identification of Neuroprotective Genes in Glaucoma. FRONTIERS IN OPHTHALMOLOGY 2022; 2:905352. [PMID: 38983528 PMCID: PMC11182220 DOI: 10.3389/fopht.2022.905352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/05/2022] [Indexed: 07/11/2024]
Abstract
Retinal ganglion cells (RGCs) are the neurons in the retina which directly project to the brain and transmit visual information along the optic nerve. Glaucoma, one of the leading causes of blindness, is characterized by elevated intraocular pressure (IOP) and degeneration of the optic nerve, which is followed by RGC death. Currently, there are no clinical therapeutic drugs or molecular interventions that prevent RGC death outside of IOP reduction. In order to overcome these major barriers, an increased number of studies have utilized the following combined analytical methods: well-established rodent models of glaucoma including optic nerve injury models and transcriptomic gene expression profiling, resulting in the successful identification of molecules and signaling pathways relevant to RGC protection. In this review, we present a comprehensive overview of pathological features in a variety of animal models of glaucoma and top differentially expressed genes (DEGs) depending on disease progression, RGC subtypes, retinal regions or animal species. By comparing top DEGs among those different transcriptome profiles, we discuss whether commonly listed DEGs could be defined as potential novel therapeutic targets in glaucoma, which will facilitate development of future therapeutic neuroprotective strategies for treatments of human patients in glaucoma.
Collapse
Affiliation(s)
- Dwarkesh Amin
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Takaaki Kuwajima
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Petrović A, Ban J, Ivaničić M, Tomljanović I, Mladinic M. The Role of ATF3 in Neuronal Differentiation and Development of Neuronal Networks in Opossum Postnatal Cortical Cultures. Int J Mol Sci 2022; 23:ijms23094964. [PMID: 35563354 PMCID: PMC9100162 DOI: 10.3390/ijms23094964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022] Open
Abstract
Activating transcription factor 3 (ATF3), a member of the ATF/cAMP response element-binding (CREB) family, is upregulated by various intracellular and extracellular signals such as injury and signals related to cell proliferation. ATF3 also belongs to the regeneration-associated genes (RAG) group of transcription factors. RAG and ATF/CREB transcription factors that play an important role in embryonic neuronal development and PNS regeneration may also be involved in postnatal neuronal differentiation and development, as well as in the regeneration of the injured CNS. Here we investigated the effect of ATF3 in differentiation, neural outgrowth, network formation, and regeneration after injury using postnatal dissociated cortical neurons derived from neonatal opossums (Monodelphis domestica). Our results show that RAG and ATF genes are differentially expressed in early differentiated neurons versus undifferentiated neurospheres and that many members of those families, ATF3 in particular, are upregulated in cortical cultures obtained from younger animals that have the ability to fully functionally regenerate spinal cord after injury. In addition, we observed different intracellular localization of ATF3 that shifts from nuclear (in neuronal progenitors) to cytoplasmic (in more mature neurons) during neuronal differentiation. The ATF3 inhibition, pharmacological or by specific antibody, reduced the neurite outgrowth and differentiation and caused increased cell death in early differentiating cortical neuronal cultures, suggesting the importance of ATF3 in the CNS development of neonatal opossums. Finally, we investigated the regeneration capacity of primary cortical cultures after mechanical injury using the scratch assay. Remarkably, neonatal opossum-derived cultures retain their capacity to regenerate for up to 1 month in vitro. Inhibition of ATF3 correlates with reduced neurite outgrowth and regeneration after injury. These results indicate that ATF3, and possibly other members of RAG and ATF/CREB family of transcription factors, have an important role both during cortical postnatal development and in response after injury.
Collapse
|
18
|
Ma N, Li G, Fu X. Protective role of activating transcription factor 3 against neuronal damage in rats with cerebral ischemia. Brain Behav 2022; 12:e2522. [PMID: 35263513 PMCID: PMC9014992 DOI: 10.1002/brb3.2522] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/17/2021] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The participation of activating transcription factor 3 (ATF3) in transient middle cerebral artery occlusion and reperfusion injury has been reported. However, the precise mechanism of ATF3 in cerebral ischemia is little known so far. Thus, the study examines the mechanism of action underlying the protective role of ATF3 following middle cerebral artery occlusion (MCAO) in rats. METHODS AND RESULTS The MCAO rats exhibited reduced body weight and motor ability, while increased neurological deficits and brain infarct volume. Gene ontology (GO) enrichment and KEGG pathway analyses revealed that differentially expressed genes were mainly enriched in the TLR4/NF-κB signaling. Moreover, ATF3 was the most differentially expressed gene in brain tissues of MCAO rats versus sham-operated rats, which could bind to CCL2. ATF3 was reduced in MCAO rats, and ATF3 inhibited CCL2 expression to mediate the TLR4/NF-κB signaling. Functionally, ATF3 inhibited neuronal apoptosis, microglia activation, and pro-inflammatory cytokine production to alleviate brain injury in rats. By contrast, CCL2 was overexpressed in neurons and microglia, and CCL2 mitigated the effects of ATF3 to exacerbate brain injury in rats. CONCLUSION Our findings suggested that ATF3 repressed neuronal apoptosis and microglia activation caused by cerebral ischemia via targeting CCL2 and mediating the TLR4/NF-κB signaling.
Collapse
Affiliation(s)
- Na Ma
- Department of Neurology, Caoxian People's Hospital, Heze, P. R. China
| | - Gaixia Li
- Women and Children's Hospital, Qingdao University, Qingdao, P. R. China
| | - Xiuxin Fu
- Department of Neurology, Weifang People's Hospital Affiliated to Weifang Medical College, Weifang, P. R. China
| |
Collapse
|
19
|
Wang T, Xu J, Xu Y, Xiao J, Bi N, Gu X, Wang HL. Gut microbiota shapes social dominance through modulating HDAC2 in the medial prefrontal cortex. Cell Rep 2022; 38:110478. [PMID: 35263606 DOI: 10.1016/j.celrep.2022.110478] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/29/2021] [Accepted: 02/08/2022] [Indexed: 12/17/2022] Open
Abstract
Social dominance is a ubiquitous phenomenon among social animals, including humans. To date, individual attributes leading to dominance (after a contest) remain largely elusive. Here, we report that socially dominant rats can be distinguished from subordinates based on their intestinal microbiota. When dysbiosis is induced, rats are predisposed to a subordinate state, while dysbiotic rats reclaim social dominance following microbiota transplantation. Winning hosts are characterized by core microbes, a majority of which are associated with butyrate production, and the sole colonization of Clostridium butyricum is sufficient to restore dominance. Regarding molecular aspects, a histone deacetylase, HDAC2, is responsive to microbial status and mediates competition outcome; however, this occurs only in a restricted population of cells in the medial prefrontal cortex (mPFC). Furthermore, HDAC2 acts by modulating synaptic activity in mPFC. Together, these findings uncover a link between commensals and host dominance, providing insight into the gut-brain mechanisms underlying dominance determination.
Collapse
Affiliation(s)
- Tian Wang
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Jinchun Xu
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Yi Xu
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei 230009, China.
| | - Jie Xiao
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Nanxi Bi
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Xiaozhen Gu
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China
| | - Hui-Li Wang
- School of Food and Bioengineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei 230009, China.
| |
Collapse
|
20
|
McMahon DB, Kuek LE, Johnson ME, Johnson PO, Horn RL, Carey RM, Adappa ND, Palmer JN, Lee RJ. The bitter end: T2R bitter receptor agonists elevate nuclear calcium and induce apoptosis in non-ciliated airway epithelial cells. Cell Calcium 2022; 101:102499. [PMID: 34839223 PMCID: PMC8752513 DOI: 10.1016/j.ceca.2021.102499] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/21/2021] [Accepted: 10/31/2021] [Indexed: 01/03/2023]
Abstract
Bitter taste receptors (T2Rs) localize to airway motile cilia and initiate innate immune responses in retaliation to bacterial quorum sensing molecules. Activation of cilia T2Rs leads to calcium-driven NO production that increases cilia beating and directly kills bacteria. Several diseases, including chronic rhinosinusitis, COPD, and cystic fibrosis, are characterized by loss of motile cilia and/or squamous metaplasia. To understand T2R function within the altered landscape of airway disease, we studied T2Rs in non-ciliated airway cell lines and primary cells. Several T2Rs localize to the nucleus in de-differentiated cells that typically localize to cilia in differentiated cells. As cilia and nuclear import utilize shared proteins, some T2Rs may target to the nucleus in the absence of motile cilia. T2R agonists selectively elevated nuclear and mitochondrial calcium through a G-protein-coupled receptor phospholipase C mechanism. Additionally, T2R agonists decreased nuclear cAMP, increased nitric oxide, and increased cGMP, consistent with T2R signaling. Furthermore, exposure to T2R agonists led to nuclear calcium-induced mitochondrial depolarization and caspase activation. T2R agonists induced apoptosis in primary bronchial and nasal cells differentiated at air-liquid interface but then induced to a squamous phenotype by apical submersion. Air-exposed well-differentiated cells did not die. This may be a last-resort defense against bacterial infection. However, it may also increase susceptibility of de-differentiated or remodeled epithelia to damage by bacterial metabolites. Moreover, the T2R-activated apoptosis pathway occurs in airway cancer cells. T2Rs may thus contribute to microbiome-tumor cell crosstalk in airway cancers. Targeting T2Rs may be useful for activating cancer cell apoptosis while sparing surrounding tissue.
Collapse
Affiliation(s)
- Derek B. McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Correspondence: Derek B. McMahon, PhD or Robert J. Lee, PhD, Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA, 215-573-9766, (D.B.M.) or (R.J.L)
| | - Li Eon Kuek
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Madeline E. Johnson
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paige O. Johnson
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rachel L.J. Horn
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ryan M. Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nithin D. Adappa
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - James N. Palmer
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Robert J. Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Correspondence: Derek B. McMahon, PhD or Robert J. Lee, PhD, Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA, 215-573-9766, (D.B.M.) or (R.J.L)
| |
Collapse
|
21
|
Yang T, Guo R, Ofengeim D, Hwang JY, Zukin RS, Chen J, Zhang F. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal Death. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
22
|
Safaralizade M, Fuderer R, Grosse R, Zhao B. Measuring nuclear calcium and actin assembly in living cells. J Biochem 2021; 169:287-294. [PMID: 33479753 DOI: 10.1093/jb/mvab002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/29/2020] [Indexed: 11/12/2022] Open
Abstract
Nuclear calcium signalling has emerged as a critical mechanism regulating processes like chromatin organization and gene expression. Recently, we have shown that nuclear calcium elevation triggers rapid and transient actin filament assembly inside the nucleus. Here, we constructed and employed a nuclear-specific calcium sensor based upon the new generation of genetically encoded probes jGCaMP7f. By fusing a nuclear localization signal to jGCaMP7f, we achieved highly efficient nuclear-specific targeting. Comparing the jGCaMP7f-NLS probe with the previous GCaMP6f-NLS calcium sensor showed clearly that jGCaMP7f-NLS is more sensitive and reverses significantly quicker thereby reflecting rapid nuclear calcium transients in a closely physiological manner. We further confirm that nuclear calcium transients precede nuclear actin polymerization by several seconds. Our data show that calcium-triggered nuclear actin assembly in fibroblasts is independent of the actin nucleating Arp2/3 complex. Together, jGCaMP7f-NLS represents an easy to use, reliable and highly sensitive nuclear calcium sensor that allows to tightly interrogate real-time, spatiotemporal calcium signalling and calcium-elicited effects in the nucleus of living cells.
Collapse
Affiliation(s)
| | | | - Robert Grosse
- Institute of Pharmacology, Faculty of Medicine.,Centre for Integrative Biological Signaling Studies, University of Freiburg, Albertstraße 25, D-79104 Freiburg, Germany
| | - Bing Zhao
- Institute of Pharmacology, Faculty of Medicine.,Centre for Integrative Biological Signaling Studies, University of Freiburg, Albertstraße 25, D-79104 Freiburg, Germany
| |
Collapse
|
23
|
Effects of datumetine on hippocampal NMDAR activity. Toxicol Rep 2021; 8:1131-1142. [PMID: 34150523 PMCID: PMC8190477 DOI: 10.1016/j.toxrep.2021.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/16/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022] Open
Abstract
The usage (abuse) of Datura metel is becoming increasingly worrisome among the Nigerian populace especially among the youth considering its side effects such as hallucination. This work was designed to identify the phytochemicals in datura plant that potentially interact with NMDAR as it affects the electrical and memory activities of the brain. Ligand-protein interaction was assessed using autodock vina to identify phytochemicals that can interact with NMDAR. Datumetine was found to have the best interaction fit with NMDAR at both allosteric and orthosteric binding sites. Furthermore, using electrophysiological, behavioural and western blotting techniques, it was observed that the administration of datumetine positively modulates the NMDAR current by prolonging burst duration and interspike interval, induces seizures in C57BL/6 mice. Acute exposure leads to memory deficit on NOR and Y-maze test while immunoblotting results showed increased expression of GluN1 and CamKIIα while pCamKIIα-T286, CREB and BDNF were downregulated. The results showed that the memory deficit seen in datura intoxication is possibly the effects of datumetine on NMDAR.
Collapse
|
24
|
Simats A, Ramiro L, García-Berrocoso T, Briansó F, Gonzalo R, Martín L, Sabé A, Gill N, Penalba A, Colomé N, Sánchez A, Canals F, Bustamante A, Rosell A, Montaner J. A Mouse Brain-based Multi-omics Integrative Approach Reveals Potential Blood Biomarkers for Ischemic Stroke. Mol Cell Proteomics 2020; 19:1921-1936. [PMID: 32868372 PMCID: PMC7710142 DOI: 10.1074/mcp.ra120.002283] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke remains a leading cause of death and disability worldwide. Despite continuous advances, the identification of key molecular signatures in the hyper-acute phase of ischemic stroke is still a primary interest for translational research on stroke diagnosis, prognosis, and treatment. Data integration from high-throughput -omics techniques has become crucial to unraveling key interactions among different molecular elements in complex biological contexts, such as ischemic stroke. Thus, we used advanced data integration methods for a multi-level joint analysis of transcriptomics and proteomics data sets obtained from mouse brains at 2 h after cerebral ischemia. By modeling net-like correlation structures, we identified an integrated network of genes and proteins that are differentially expressed at a very early stage after stroke. We validated 10 of these deregulated elements in acute stroke, and changes in their expression pattern over time after cerebral ischemia were described. Of these, CLDN20, GADD45G, RGS2, BAG5, and CTNND2 were next evaluated as blood biomarkers of cerebral ischemia in mice and human blood samples, which were obtained from stroke patients and patients presenting stroke-mimicking conditions. Our findings indicate that CTNND2 levels in blood might potentially be useful for distinguishing ischemic strokes from stroke-mimicking conditions in the hyper-acute phase of the disease. Furthermore, circulating GADD45G content within the first 6 h after stroke could also play a key role in predicting poor outcomes in stroke patients. For the first time, we have used an integrative biostatistical approach to elucidate key molecules in the initial stages of stroke pathophysiology and highlight new notable molecules that might be further considered as blood biomarkers of ischemic stroke.
Collapse
Affiliation(s)
- Alba Simats
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Ramiro
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ferran Briansó
- Bioinformatics and Biostatistics Unit, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Genetics, Microbiology and Statistics Dept., Universitat de Barcelona, Barcelona, Spain
| | - Ricardo Gonzalo
- Bioinformatics and Biostatistics Unit, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luna Martín
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Sabé
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Natalia Gill
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Penalba
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nuria Colomé
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sánchez
- Bioinformatics and Biostatistics Unit, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Genetics, Microbiology and Statistics Dept., Universitat de Barcelona, Barcelona, Spain
| | - Francesc Canals
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
25
|
Vallese F, Catoni C, Cieri D, Barazzuol L, Ramirez O, Calore V, Bonora M, Giamogante F, Pinton P, Brini M, Calì T. An expanded palette of improved SPLICS reporters detects multiple organelle contacts in vitro and in vivo. Nat Commun 2020; 11:6069. [PMID: 33247103 PMCID: PMC7699637 DOI: 10.1038/s41467-020-19892-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Membrane contact sites between virtually any known organelle have been documented and, in the last decades, their study received momentum due to their importance for fundamental activities of the cell and for the subtle comprehension of many human diseases. The lack of tools to finely image inter-organelle proximity hindered our understanding on how these subcellular communication hubs mediate and regulate cell homeostasis. We develop an improved and expanded palette of split-GFP-based contact site sensors (SPLICS) for the detection of single and multiple organelle contact sites within a scalable distance range. We demonstrate their flexibility under physiological conditions and in living organisms.
Collapse
Affiliation(s)
- Francesca Vallese
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Domenico Cieri
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lucia Barazzuol
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Omar Ramirez
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Valentina Calore
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Flavia Giamogante
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of General Pathology, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Marisa Brini
- Department of Biology, University of Padova, Padova, Italy.
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, Padova, Italy. .,Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
| |
Collapse
|
26
|
Yan J, Bengtson CP, Buchthal B, Hagenston AM, Bading H. Coupling of NMDA receptors and TRPM4 guides discovery of unconventional neuroprotectants. Science 2020; 370:370/6513/eaay3302. [PMID: 33033186 DOI: 10.1126/science.aay3302] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 05/05/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
Excitotoxicity induced by NMDA receptors (NMDARs) is thought to be intimately linked to high intracellular calcium load. Unexpectedly, NMDAR-mediated toxicity can be eliminated without affecting NMDAR-induced calcium signals. Instead, excitotoxicity requires physical coupling of NMDARs to TRPM4. This interaction is mediated by intracellular domains located in the near-membrane portions of the receptors. Structure-based computational drug screening using the interaction interface of TRPM4 in complex with NMDARs identified small molecules that spare NMDAR-induced calcium signaling but disrupt the NMDAR/TRPM4 complex. These interaction interface inhibitors strongly reduce NMDA-triggered toxicity and mitochondrial dysfunction, abolish cyclic adenosine monophosphate-responsive element-binding protein (CREB) shutoff, boost gene induction, and reduce neuronal loss in mouse models of stroke and retinal degeneration. Recombinant or small-molecule NMDAR/TRPM4 interface inhibitors may mitigate currently untreatable human neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Yan
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Bettina Buchthal
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
27
|
Chen WJ, Lai YJ, Lee JL, Wu ST, Hsu YJ. CREB/ATF3 signaling mediates indoxyl sulfate-induced vascular smooth muscle cell proliferation and neointimal formation in uremia. Atherosclerosis 2020; 315:43-54. [PMID: 33227547 DOI: 10.1016/j.atherosclerosis.2020.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/15/2020] [Accepted: 11/06/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Uremic patients are characterized by an increased risk of atherosclerotic cardiovascular diseases. Vascular smooth muscle cell (VSMC) proliferation contributes to neointimal formation, a main pathological feature in atherosclerosis. Activation of CREB/ATF3 signaling is pivotal in VSMC proliferation, yet its role in uremic atherosclerosis is unknown. This study aimed to explore whether CREB/ATF3 signaling is involved in the molecular mechanism underlying neointimal formation in uremia. METHODS AND RESULTS Treatment of VSMCs with uremic toxin (indoxyl sulfate [IS]) activated cAMP/CREB/ATF3/cyclin D signaling, which was reflected by increased VSMC proliferation. Blocking cAMP/PKA/CREB/ATF3 signaling attenuated the promoting effect of IS on cyclin D1 expression and VSMC proliferation. Loss-of-function and time-dependent experiments showed that ATF3 lies downstream of the CREB signaling. Mutational analysis of cyclin D1 promoter along with chromatin immunoprecipitation assays showed that CREB/ATF3 signaling participated in IS-induced cyclin D transcription. In vivo, phosphorylated CREB (an active form of CREB) and ATF3 were prominently upregulated in the neointima of experimental uremic rats, the atherosclerotic plaques of uremic ApoE-/- mice, and the iliac arteries of uremic patients. Notably, the use of lentivirus to knock down ATF3 in the neointima of balloon-injured arteries could suppress the effect of uremia in vivo, including neointimal formation and cyclin D expression. CONCLUSIONS In this study, we demonstrated that CREB/ATF3-related signaling may be involved in IS-induced VSMC proliferation and the pathogenesis of neointimal formation during uremia.
Collapse
Affiliation(s)
- Wei-Jan Chen
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Ying-Ju Lai
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Jia-Lin Lee
- Institute of Molecular and Cellular Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Sheng-Tang Wu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
28
|
Mauceri D, Buchthal B, Hemstedt TJ, Weiss U, Klein CD, Bading H. Nasally delivered VEGFD mimetics mitigate stroke-induced dendrite loss and brain damage. Proc Natl Acad Sci U S A 2020; 117:8616-8623. [PMID: 32229571 PMCID: PMC7165430 DOI: 10.1073/pnas.2001563117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In the adult brain, vascular endothelial growth factor D (VEGFD) is required for structural integrity of dendrites and cognitive abilities. Alterations of dendritic architectures are hallmarks of many neurologic disorders, including stroke-induced damage caused by toxic extrasynaptic NMDA receptor (eNMDAR) signaling. Here we show that stimulation of eNMDARs causes a rapid shutoff of VEGFD expression, leading to a dramatic loss of dendritic structures. Using the mouse middle cerebral artery occlusion (MCAO) stroke model, we have established the therapeutic potential of recombinant mouse VEGFD delivered intraventricularly to preserve dendritic architecture, reduce stroke-induced brain damage, and facilitate functional recovery. An easy-to-use therapeutic intervention for stroke was developed that uses a new class of VEGFD-derived peptide mimetics and postinjury nose-to-brain delivery.
Collapse
Affiliation(s)
- Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld (INF) 366, 69120 Heidelberg, Germany
| | - Bettina Buchthal
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld (INF) 366, 69120 Heidelberg, Germany
| | - Thekla J Hemstedt
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld (INF) 366, 69120 Heidelberg, Germany
| | - Ursula Weiss
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld (INF) 366, 69120 Heidelberg, Germany
| | - Christian D Klein
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, INF 364, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Im Neuenheimer Feld (INF) 366, 69120 Heidelberg, Germany;
| |
Collapse
|
29
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Piccirillo S, Magi S, Castaldo P, Preziuso A, Lariccia V, Amoroso S. NCX and EAAT transporters in ischemia: At the crossroad between glutamate metabolism and cell survival. Cell Calcium 2020; 86:102160. [PMID: 31962228 DOI: 10.1016/j.ceca.2020.102160] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 01/29/2023]
Abstract
Energy metabolism impairment is a central event in the pathophysiology of ischemia. The limited availability of glucose and oxygen strongly affects mitochondrial activity, thus leading to ATP depletion. In this setting, the switch to alternative energy sources could ameliorate cells survival by enhancing ATP production, thus representing an attractive strategy for ischemic treatment. In this regard, some studies have recently re-evaluated the metabolic role of glutamate and its potential to promote cell survival under pathological conditions. In the present review, we discuss the ability of glutamate to exert an "energizing role" in cardiac and neuronal models of hypoxia/reoxygenation (H/R) injury, focusing on the Na+/Ca2+ exchanger (NCX) and the Na+-dependent excitatory amino acid transporters (EAATs) as key players in this metabolic pathway.
Collapse
Affiliation(s)
- Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy.
| | - Pasqualina Castaldo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126, Ancona, Italy
| |
Collapse
|
31
|
Bas-Orth C, Koch M, Lau D, Buchthal B, Bading H. A microRNA signature of toxic extrasynaptic N-methyl-D-aspartate (NMDA) receptor signaling. Mol Brain 2020; 13:3. [PMID: 31924235 PMCID: PMC6954508 DOI: 10.1186/s13041-020-0546-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/05/2020] [Indexed: 12/27/2022] Open
Abstract
The cellular consequences of N-Methyl-D-Aspartate receptor (NMDAR) stimulation depend on the receptors' subcellular localization. Synaptic NMDARs promote plasticity and survival whereas extrasynaptic NMDARs mediate excitotoxicity and contribute to cell death in neurodegenerative diseases. The mechanisms that couple activation of extrasynaptic NMDARs to cell death remain incompletely understood. We here show that activation of extrasynaptic NMDARs by bath application of NMDA or L-glutamate leads to the upregulation of a group of 19 microRNAs in cultured mouse hippocampal neurons. In contrast, none of these microRNAs is induced upon stimulation of synaptic activity. Increased microRNA expression depends on the pri-miRNA processing enzyme Drosha, but not on de novo gene transcription. These findings suggest that toxic NMDAR signaling involves changes in the expression levels of particular microRNAs.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany. .,Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany.
| | - Mirja Koch
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - David Lau
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Bettina Buchthal
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| |
Collapse
|
32
|
Djemil S, Chen X, Zhang Z, Lee J, Rauf M, Pak DTS, Dzakpasu R. Activation of nicotinic acetylcholine receptors induces potentiation and synchronization within in vitro hippocampal networks. J Neurochem 2019; 153:468-484. [PMID: 31821553 DOI: 10.1111/jnc.14938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are known to play a role in cognitive functions of the hippocampus, such as memory consolidation. Given that they conduct Ca2+ and are capable of regulating the release of glutamate and γ-aminobutyric acid (GABA) within the hippocampus, thereby shifting the excitatory-inhibitory ratio, we hypothesized that the activation of nAChRs will result in the potentiation of hippocampal networks and alter synchronization. We used nicotine as a tool to investigate the impact of activation of nAChRs on neuronal network dynamics in primary embryonic rat hippocampal cultures prepared from timed-pregnant Sprague-Dawley rats. We perturbed cultured hippocampal networks with increasing concentrations of bath-applied nicotine and performed network extracellular recordings of action potentials using a microelectrode array. We found that nicotine modulated network dynamics in a concentration-dependent manner; it enhanced firing of action potentials as well as facilitated bursting activity. In addition, we used pharmacological agents to determine the contributions of discrete nAChR subtypes to the observed network dynamics. We found that β4-containing nAChRs are necessary for the observed increases in spiking, bursting, and synchrony, while the activation of α7 nAChRs augments nicotine-mediated network potentiation but is not necessary for its manifestation. We also observed that antagonists of N-methyl-D-aspartate receptors (NMDARs) and group I metabotropic glutamate receptors (mGluRs) partially blocked the effects of nicotine. Furthermore, nicotine exposure promoted autophosphorylation of Ca2+ /calmodulin-dependent kinase II (CaMKII) and serine 831 phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit GluA1. These results suggest that nicotinic receptors induce potentiation and synchronization of hippocampal networks and glutamatergic synaptic transmission. Findings from this work highlight the impact of cholinergic signaling in generating network-wide potentiation in the form of enhanced spiking and bursting dynamics that coincide with molecular correlates of memory such as increased phosphorylation of CaMKII and GluA1. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Ziyue Zhang
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Jisoo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mikael Rauf
- Department of Human Science, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Rhonda Dzakpasu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Department of Physics, Georgetown University, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
33
|
Schlüter A, Aksan B, Diem R, Fairless R, Mauceri D. VEGFD Protects Retinal Ganglion Cells and, consequently, Capillaries against Excitotoxic Injury. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:281-299. [PMID: 32055648 PMCID: PMC7005343 DOI: 10.1016/j.omtm.2019.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/19/2019] [Indexed: 02/08/2023]
Abstract
In the central nervous system, neurons and the vasculature influence each other. While it is well described that a functional vascular system is trophic to neurons and that vascular damage contributes to neurodegeneration, the opposite scenario in which neural damage might impact the microvasculature is less defined. In this study, using an in vivo excitotoxic approach in adult mice as a tool to cause specific damage to retinal ganglion cells, we detected subsequent damage to endothelial cells in retinal capillaries. Furthermore, we detected decreased expression of vascular endothelial growth factor D (VEGFD) in retinal ganglion cells. In vivo VEGFD supplementation via neuronal-specific viral-mediated expression or acute intravitreal delivery of the mature protein preserved the structural and functional integrity of retinal ganglion cells against excitotoxicity and, additionally, spared endothelial cells from degeneration. Viral-mediated suppression of expression of the VEGFD-binding receptor VEGFR3 in retinal ganglion cells revealed that VEGFD exerts its protective capacity directly on retinal ganglion cells, while protection of endothelial cells is the result of upheld neuronal integrity. These findings suggest that VEGFD supplementation might be a novel, clinically applicable approach for neuronal and vascular protection.
Collapse
Affiliation(s)
- Annabelle Schlüter
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Bahar Aksan
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Im Neuenheimer Feld 368, 69120 Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Im Neuenheimer Feld 368, 69120 Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Coppola T, Beraud-Dufour S, Lebrun P, Blondeau N. Bridging the Gap Between Diabetes and Stroke in Search of High Clinical Relevance Therapeutic Targets. Neuromolecular Med 2019; 21:432-444. [PMID: 31489567 DOI: 10.1007/s12017-019-08563-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022]
Abstract
Diabetes affects more than 425 million people worldwide, a scale approaching pandemic proportion. Diabetes represents a major risk factor for stroke, and therefore is actively addressed for stroke prevention. However, how diabetes affects stroke severity has not yet been extensively considered, which is surprising given the evident but understudied common mechanistic features of both pathologies. The increase in number of diabetic people, incidence of stroke in the presence of this specific risk factor, and the exacerbation of ischemic brain damage in diabetic conditions (at least in animal models) warrants the need to integrate this comorbidity in preclinical studies of brain ischemia to develop novel therapeutic approaches. Therefore, a better understanding of the commonalties involved in the course of both diseases would offer the promise of discovering novel neuroprotective pathways that would be more appropriated to clinical scenarios. In this article, we will review the relevant mechanisms that have been identified as common traits of both pathologies and that could be, to our knowledge, potential targets in both pathologies.
Collapse
Affiliation(s)
- Thierry Coppola
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France.
| | - Sophie Beraud-Dufour
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France
| | - Patricia Lebrun
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France
| | - Nicolas Blondeau
- Université Côte d'Azur, CNRS, IPMC, 660 route des Lucioles, 06560, Valbonne, France.
| |
Collapse
|
35
|
Monaco S, Jahraus B, Samstag Y, Bading H. Conditions of limited calcium influx (CLCI) inhibits IL2 induction and favors expression of anergy-related genes in TCR/CD3 and CD28 costimulated primary human T cells. Mol Immunol 2019; 114:81-87. [PMID: 31344552 DOI: 10.1016/j.molimm.2019.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/31/2022]
Abstract
Calcium is a key regulator of the T cell immune response. Depending on the spatial properties (nucleus versus cytoplasm) of the calcium signals generated after CD3xCD28 stimulation, primary human T cells either mount a productive immune response or develop tolerance. Nuclear calcium acts as a genomic decision maker: during T cell activation, it drives expression of genes associated with a productive immune response while in its absence, stimulated T cells acquire an anergy-like gene profile. Selective inhibition of nuclear calcium signaling in stimulated T cells blocks the productive immune response and directs the cells towards an anergy-like state. Here we show that the two transcriptional programs that include, respectively, the 'activation gene', interleukin 2 (IL2) and 'anergy-related genes', EGR2, EGR3, and CREM have different requirements for transmembrane calcium flux. By either lowering extracellular calcium concentrations with EGTA or using low concentrations of the ORAI blockers, BTP2 or RO2959, we reduced transmembrane calcium flux in human primary T cells stimulated with CD3xCD28. These 'conditions of limited calcium influx' (CLCI) blocked CD3xCD28-induced IL2 expression but only moderately affected induction of the anergy-related genes EGR2, EGR3, and CREM. We observed no difference in NFAT2 nuclear translocation after CD3xCD28 stimulation between normal conditions and CLCI. These results indicate that CLCI favors expression of anergy-related genes in activated human T cells. CLCI may be used to develop novel means for pro-tolerance immunosuppressive treatments.
Collapse
Affiliation(s)
- Sara Monaco
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany.
| | - Beate Jahraus
- Department of Immunology, Heidelberg University, 69120, Heidelberg, Germany.
| | - Yvonne Samstag
- Department of Immunology, Heidelberg University, 69120, Heidelberg, Germany.
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany.
| |
Collapse
|
36
|
Cai M, Zhu Y, Li Z, Josephs-Spaulding J, Zhou Y, Hu Y, Chen H, Liu Y, He W, Zhang J. Profiling the Gene Expression and DNA Methylation in the Mouse Brain after Ischemic Preconditioning. Neuroscience 2019; 406:249-261. [DOI: 10.1016/j.neuroscience.2019.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
|
37
|
Zhu J, Wan Y, Xu H, Wu Y, Hu B, Jin H. The role of endogenous tissue-type plasminogen activator in neuronal survival after ischemic stroke: friend or foe? Cell Mol Life Sci 2019; 76:1489-1506. [PMID: 30656378 PMCID: PMC11105644 DOI: 10.1007/s00018-019-03005-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 12/29/2022]
Abstract
Endogenous protease tissue-type plasminogen activator (tPA) has highly efficient fibrinolytic activity and its recombinant variants alteplase and tenecteplase are established as highly effective thrombolytic drugs for ischemic stroke. Endogenous tPA is constituted of five functional domains through which it interacts with a variety of substrates, binding proteins and receptors, thus having enzymatic and cytokine-like effects to act on all cell types of the brain. In the past 2 decades, numerous studies have explored the clinical relevance of endogenous tPA in neurological diseases, especially in ischemic stroke. tPA is released from many cells within the brain parenchyma exposed to ischemia conditions in vitro and in vivo, which is believed to control neuronal fate. Some studies proved that tPA could induce blood-brain barrier disruption, neural excitotoxicity and inflammation, while others indicated that tPA also has anti-excitotoxic, neurotrophic and anti-apoptotic effects on neurons. Therefore, more work is needed to elucidate how tPA mediates such opposing functions that may amplify tPA from a therapeutic means into a key therapeutic target in endogenous neuroprotection after stroke. In this review, we summarize the biological characteristics and pleiotropic functions of tPA in the brain. Then we focus on possible hypotheses about why and how endogenous tPA mediates ischemic neuronal death and survival. Finally, we analyze how endogenous tPA affects neuron fate in ischemic stroke in a comprehensive view.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yan Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hexiang Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yulang Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
38
|
Ernst AS, Böhler LI, Hagenston AM, Hoffmann A, Heiland S, Sticht C, Bendszus M, Hecker M, Bading H, Marti HH, Korff T, Kunze R. EphB2-dependent signaling promotes neuronal excitotoxicity and inflammation in the acute phase of ischemic stroke. Acta Neuropathol Commun 2019; 7:15. [PMID: 30722785 PMCID: PMC6362601 DOI: 10.1186/s40478-019-0669-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Local cerebral hypoperfusion causes ischemic stroke while driving multiple cell-specific responses including inflammation, glutamate-induced neurotoxicity mediated via NMDAR, edema formation and angiogenesis. Despite the relevance of these pathophysiological mechanisms for disease progression and outcome, molecular determinants controlling the onset of these processes are only partially understood. In this context, our study intended to investigate the functional role of EphB2, a receptor tyrosine kinase that is crucial for synapse function and binds to membrane-associated ephrin-B ligands. Cerebral ischemia was induced in Ephb2−/− mice by transient middle cerebral artery occlusion followed by different times (6, 12, 24 and 48 h) of reperfusion. Histological, neurofunctional and transcriptome analyses indicated an increase in EphB2 phosphorylation under these conditions and attenuated progression of stroke in Ephb2−/− mice. Moreover, while infiltration of microglia/macrophages and astrocytes into the peri-infarct region was not altered, expression of the pro-inflammatory mediators MCP-1 and IL-6 was decreased in these mice. In vitro analyses indicated that binding of EphB2 to astrocytic ephrin-B ligands stimulates NF-κB-mediated cytokine expression via the MAPK pathway. Further magnetic resonance imaging of the Ephb2−/− ischemic brain revealed a lower level of cytotoxic edema formation within 6 h upon onset of reperfusion. On the mechanistic level, absence of neuronal EphB2 decreased the mitochondrial Ca2+ load upon specific activation of NMDAR but not during synaptic activity. Furthermore, neuron-specific loss of ephrin-B2 reduced the extent of cerebral tissue damage in the acute phase of ischemic stroke. Collectively, EphB2 may promote the immediate response to an ischemia-reperfusion event in the central nervous system by (i) pro-inflammatory activation of astrocytes via ephrin-B-dependent signaling and (ii) amplification of NMDA-evoked neuronal excitotoxicity.
Collapse
|
39
|
Balu DT, Coyle JT. Altered CREB Binding to Activity-Dependent Genes in Serine Racemase Deficient Mice, a Mouse Model of Schizophrenia. ACS Chem Neurosci 2018; 9:2205-2209. [PMID: 29172439 DOI: 10.1021/acschemneuro.7b00404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
cAMP-response-element-binding protein (CREB) is a transcription factor ubiquitously expressed in the brain that regulates neuroplasticity by modulating gene expression. The influx of calcium through N-methyl-d-aspartate receptors (NMDARs) is a well-defined mechanism that leads to the increased expression of CREB-dependent genes, including brain derived neurotrophic factor (BDNF), microRNA-132, and activity-regulated cytoskeleton-associated protein (Arc). These molecules are implicated in the pathophysiology of schizophrenia. We previously demonstrated that serine racemase knockout (SR-/-) mice, which exhibit NMDAR hypofunction due to a lack of the forebrain NMDAR co-agonist d-serine, also have reduced expression of CREB-dependent genes in the hippocampus. Using chromatin immunoprecipitation, we show here that, in SR-/- mice, there is less CREB bound to the promoter regions of BDNF, microRNA-132, and Arc. These data suggest that NMDAR hypofunction in SR-/- mice leads to reduced CREB binding on known activity-dependent genes, in turn contributing to their reduced expression.
Collapse
Affiliation(s)
- Darrick T. Balu
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts 02115, United States
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, Massachusetts 02478, United States
| | - Joseph T. Coyle
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, Massachusetts 02478, United States
| |
Collapse
|
40
|
Post-injury Nose-to-Brain Delivery of Activin A and SerpinB2 Reduces Brain Damage in a Mouse Stroke Model. Mol Ther 2018; 26:2357-2365. [PMID: 30093305 DOI: 10.1016/j.ymthe.2018.07.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/14/2018] [Accepted: 07/19/2018] [Indexed: 01/01/2023] Open
Abstract
Synaptic NMDA receptors activating nuclear calcium-driven adaptogenomics control a potent body-own neuroprotective mechanism, referred to as acquired neuroprotection. Viral vector-mediated gene transfer in conjunction with stereotactic surgery has previously demonstrated the proficiency of several nuclear calcium-regulated genes to protect in vivo against brain damage caused by toxic extrasynaptic NMDA receptor signaling following seizures or stroke. Here we used noninvasive nose-to-brain administration of Activin A and SerpinB2, two secreted nuclear calcium-regulated neuroprotectants, for post-injury treatment of brain damage following middle cerebral artery occlusion (MCAO) in C57BL/6N mice. The observed reduction of the infarct volume was comparable to the protection obtained by intracerebroventricular injection of recombinant Activin A or SerpinB2 or by stereotactic delivery 3 weeks prior to the injury of a recombinant adeno-associated virus containing an expression cassette for the potent neuroprotective transcription factor Npas4. These results establish post-injury, nose-to-brain delivery of Activin A and SerpinB2 as effective and possibly clinically applicable treatments of acute and chronic neurodegenerative conditions.
Collapse
|
41
|
Chandrasekar A, Olde Heuvel F, Wepler M, Rehman R, Palmer A, Catanese A, Linkus B, Ludolph A, Boeckers T, Huber-Lang M, Radermacher P, Roselli F. The Neuroprotective Effect of Ethanol Intoxication in Traumatic Brain Injury Is Associated with the Suppression of ErbB Signaling in Parvalbumin-Positive Interneurons. J Neurotrauma 2018; 35:2718-2735. [PMID: 29774782 DOI: 10.1089/neu.2017.5270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ethanol intoxication (EI) is a frequent comorbidity of traumatic brain injury (TBI), but the impact of EI on TBI pathogenic cascades and prognosis is unclear. Although clinical evidence suggests that EI may have neuroprotective effects, experimental support is, to date, inconclusive. We aimed at elucidating the impact of EI on TBI-associated neurological deficits, signaling pathways, and pathogenic cascades in order to identify new modifiers of TBI pathophysiology. We have shown that ethanol administration (5 g/kg) before trauma enhances behavioral recovery in a weight-drop TBI model. Neuronal survival in the injured somatosensory cortex was also enhanced by EI. We have used phospho-receptor tyrosine kinase (RTK) arrays to screen the impact of ethanol on TBI-induced activation of RTK in somatosensory cortex, identifying ErbB2/ErbB3 among the RTKs activated by TBI and suppressed by ethanol. Phosphorylation of ErbB2/3/4 RTKs were upregulated in vGlut2+ excitatory synapses in the injured cortex, including excitatory synapses located on parvalbumin (PV)-positive interneurons. Administration of selective ErbB inhibitors was able to recapitulate, to a significant extent, the neuroprotective effects of ethanol both in sensorimotor performance and structural integrity. Further, suppression of PV interneurons in somatosensory cortex before TBI, by engineered receptors with orthogonal pharmacology, could mimic the beneficial effects of ErbB inhibitors. Thus, we have shown that EI interferes with TBI-induced pathogenic cascades at multiple levels, with one prominent pathway, involving ErbB-dependent modulation of PV interneurons.
Collapse
Affiliation(s)
| | | | - Martin Wepler
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Rida Rehman
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Annette Palmer
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Alberto Catanese
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Birgit Linkus
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Albert Ludolph
- 1 Department of Neurology, Ulm University , Ulm, Germany
| | - Tobias Boeckers
- 4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| | - Markus Huber-Lang
- 3 Institute of Clinical and Experimental Trauma-Immunology, Ulm University , Ulm, Germany
| | - Peter Radermacher
- 2 Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University , Ulm, Germany
| | - Francesco Roselli
- 1 Department of Neurology, Ulm University , Ulm, Germany .,4 Department of Anatomy and Cell Biology, Ulm University , Ulm, Germany
| |
Collapse
|
42
|
Breit M, Queisser G. What Is Required for Neuronal Calcium Waves? A Numerical Parameter Study. JOURNAL OF MATHEMATICAL NEUROSCIENCE 2018; 8:9. [PMID: 30006849 PMCID: PMC6045568 DOI: 10.1186/s13408-018-0064-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/15/2018] [Indexed: 06/03/2023]
Abstract
Neuronal calcium signals propagating by simple diffusion and reaction with mobile and stationary buffers are limited to cellular microdomains. The distance intracellular calcium signals can travel may be significantly increased by means of calcium-induced calcium release from internal calcium stores, notably the endoplasmic reticulum. The organelle, which can be thought of as a cell-within-a-cell, is able to sequester large amounts of cytosolic calcium ions via SERCA pumps and selectively release them into the cytosol through ryanodine receptor channels leading to the formation of calcium waves. In this study, we set out to investigate the basic properties of such dendritic calcium waves and how they depend on the three parameters dendrite radius, ER radius and ryanodine receptor density in the endoplasmic membrane. We demonstrate that there are stable and abortive regimes for calcium waves, depending on the above morphological and physiological parameters. In stable regimes, calcium waves can travel across long dendritic distances, similar to electrical action potentials. We further observe that abortive regimes exist, which could be relevant for spike-timing dependent plasticity, as travel distances and wave velocities vary with changing intracellular architecture. For some of these regimes, analytic functions could be derived that fit the simulation data. In parameter spaces, that are non-trivially influenced by the three-dimensional calcium concentration profile, we were not able to derive such a functional description, demonstrating the mathematical requirement to model and simulate biochemical signaling in three-dimensional space.
Collapse
Affiliation(s)
- Markus Breit
- G-CSC, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
43
|
Chandrasekar A, Heuvel FO, Tar L, Hagenston AM, Palmer A, Linkus B, Ludolph AC, Huber-Lang M, Boeckers T, Bading H, Roselli F. Parvalbumin Interneurons Shape Neuronal Vulnerability in Blunt TBI. Cereb Cortex 2018; 29:2701-2715. [DOI: 10.1093/cercor/bhy139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 04/20/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
| | | | - Lilla Tar
- Department of Neurology, Ulm University, Ulm-DE, Germany
| | - Anna M Hagenston
- Department of Neurobiology—IZN, Heidelberg University, Heidelberg-DE, Germany
| | - Annette Palmer
- Department of Orthopedic trauma, Hand, Plastic and Reconstruction Surgery, Institute of Clinical and Experimental Trauma Immunology, Ulm University, Ulm-DE, Germany
| | - Birgit Linkus
- Department of Neurology, Ulm University, Ulm-DE, Germany
| | | | - Markus Huber-Lang
- Department of Orthopedic trauma, Hand, Plastic and Reconstruction Surgery, Institute of Clinical and Experimental Trauma Immunology, Ulm University, Ulm-DE, Germany
| | - Tobias Boeckers
- Department of Anatomy and Cell Biology, Ulm University, Ulm-DE, Germany
| | - Hilmar Bading
- Department of Neurobiology—IZN, Heidelberg University, Heidelberg-DE, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm-DE, Germany
- Department of Orthopedic trauma, Hand, Plastic and Reconstruction Surgery, Institute of Clinical and Experimental Trauma Immunology, Ulm University, Ulm-DE, Germany
- Neurozentrum—Ulm University, Ulm-DE, Germany
| |
Collapse
|
44
|
Kramer NJ, Haney MS, Morgens DW, Jovičić A, Couthouis J, Li A, Ousey J, Ma R, Bieri G, Tsui CK, Shi Y, Hertz NT, Tessier-Lavigne M, Ichida JK, Bassik MC, Gitler AD. CRISPR-Cas9 screens in human cells and primary neurons identify modifiers of C9ORF72 dipeptide-repeat-protein toxicity. Nat Genet 2018; 50:603-612. [PMID: 29507424 PMCID: PMC5893388 DOI: 10.1038/s41588-018-0070-7] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022]
Abstract
Hexanucleotide repeat expansions in the C9orf72 gene are the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (c9FTD/ALS). The nucleotide repeat expansions are translated into dipeptide repeat (DPR) proteins, which are aggregation-prone and may contribute to neurodegeneration. We used the CRISPR-Cas9 system to perform genome-wide gene knockout screens for suppressors and enhancers of C9orf72 DPR toxicity in human cells. We validated hits by performing secondary CRISPR-Cas9 screens in primary mouse neurons. We uncovered potent modifiers of DPR toxicity whose gene products function in nucleocytoplasmic transport, the endoplasmic reticulum (ER), proteasome, RNA processing pathways, and in chromatin modification. One modifier, TMX2, modulated the ER-stress signature elicited by C9orf72 DPRs in neurons, and improved survival of human induced motor neurons from C9orf72 ALS patients. Together, this work demonstrates the promise of CRISPR-Cas9 screens to define mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicholas J Kramer
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael S Haney
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - David W Morgens
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ana Jovičić
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - James Ousey
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosanna Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gregor Bieri
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - C Kimberly Tsui
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yingxiao Shi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | | | | | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
45
|
Pai CS, Sharma PK, Huang HT, Loganathan S, Lin H, Hsu YL, Phasuk S, Liu IY. The Activating Transcription Factor 3 ( Atf3) Homozygous Knockout Mice Exhibit Enhanced Conditioned Fear and Down Regulation of Hippocampal GELSOLIN. Front Mol Neurosci 2018. [PMID: 29515366 PMCID: PMC5826182 DOI: 10.3389/fnmol.2018.00037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The genetic and molecular basis underlying fear memory formation is a key theme in anxiety disorder research. Because activating transcription factor 3 (ATF3) is induced under stress conditions and is highly expressed in the hippocampus, we hypothesize that ATF3 plays a role in fear memory formation. We used fear conditioning and various other paradigms to test Atf3 knockout mice and study the role of ATF3 in processing fear memory. The results demonstrated that the lack of ATF3 specifically enhanced the expression of fear memory, which was indicated by a higher incidence of the freeze response after fear conditioning, whereas the occurrence of spatial memory including Morris Water Maze and radial arm maze remained unchanged. The enhanced freezing behavior and normal spatial memory of the Atf3 knockout mice resembles the fear response and numbing symptoms often exhibited by patients affected with posttraumatic stress disorder. Additionally, we determined that after fear conditioning, dendritic spine density was increased, and expression of Gelsolin, the gene encoding a severing protein for actin polymerization, was down-regulated in the bilateral hippocampi of the Atf3 knockout mice. Taken together, our results suggest that ATF3 may suppress fear memory formation in mice directly or indirectly through mechanisms involving modulation of actin polymerization.
Collapse
Affiliation(s)
- Chia-Sheng Pai
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Pranao K Sharma
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Hsien-Ting Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | | | - Heng Lin
- Department of Physiology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Luan Hsu
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Sarayut Phasuk
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Department of Physiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ingrid Y Liu
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
46
|
Chandrasekar A, Aksan B, Heuvel FO, Förstner P, Sinske D, Rehman R, Palmer A, Ludolph A, Huber-Lang M, Böckers T, Mauceri D, Knöll B, Roselli F. Neuroprotective effect of acute ethanol intoxication in TBI is associated to the hierarchical modulation of early transcriptional responses. Exp Neurol 2018; 302:34-45. [PMID: 29306704 DOI: 10.1016/j.expneurol.2017.12.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/08/2017] [Accepted: 12/30/2017] [Indexed: 01/01/2023]
Abstract
Ethanol intoxication is a risk factor for traumatic brain injury (TBI) but clinical evidence suggests that it may actually improve the prognosis of intoxicated TBI patients. We have employed a closed, weight-drop TBI model of different severity (2cm or 3cm falling height), preceded (-30min) or followed (+20min) by ethanol administration (5g/Kg). This protocol allows us to study the interaction of binge ethanol intoxication in TBI, monitoring behavioral changes, histological responses and the transcriptional regulation of a series of activity-regulated genes (immediate early genes, IEGs). We demonstrate that ethanol pretreatment before moderate TBI (2cm) significantly reduces neurological impairment and accelerates recovery. In addition, better preservation of neuronal numbers and cFos+cells was observed 7days after TBI. At transcriptional level, ethanol reduced the upregulation of a subset of IEGs encoding for transcription factors such as Atf3, c-Fos, FosB, Egr1, Egr3 and Npas4 but did not affect the upregulation of others (e.g. Gadd45b and Gadd45c). While a subset of IEGs encoding for effector proteins (such as Bdnf, InhbA and Dusp5) were downregulated by ethanol, others (such as Il-6) were unaffected. Notably, the majority of genes were sensitive to ethanol only when administered before TBI and not afterwards (the exceptions being c-Fos, Egr1 and Dusp5). Furthermore, while severe TBI (3cm) induced a qualitatively similar (but quantitatively larger) transcriptional response to moderate TBI, it was no longer sensitive to ethanol pretreatment. Thus, we have shown that a subset of the TBI-induced transcriptional responses were sensitive to ethanol intoxication at the instance of trauma (ultimately resulting in beneficial outcomes) and that the effect of ethanol was restricted to a certain time window (pre TBI treatment) and to TBI severity (moderate). This information could be critical for the translational value of ethanol in TBI and for the design of clinical studies aimed at disentangling the role of ethanol intoxication in TBI.
Collapse
Affiliation(s)
| | - Bahar Aksan
- Dept. of Neurobiology, IZN, University of Heidelberg, Germany
| | | | - Philip Förstner
- Institute of Physiological Chemistry, Ulm University, Germany
| | - Daniela Sinske
- Institute of Physiological Chemistry, Ulm University, Germany
| | | | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, Germany
| | - Tobias Böckers
- Dept. of Anatomy and Cell Biology, Ulm University, Germany
| | - Daniela Mauceri
- Dept. of Neurobiology, IZN, University of Heidelberg, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, Ulm University, Germany
| | | |
Collapse
|
47
|
Depletion of D3 dopamine receptor affects methamphetamine-induced expression patterns of Pde4b and Atf3. Neurosci Lett 2017; 665:54-60. [PMID: 29175029 DOI: 10.1016/j.neulet.2017.11.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 02/04/2023]
Abstract
The role of the D3 dopamine receptor (D3R) and the specific molecular mechanisms underlying the regulation of D3R via the cAMP signaling pathway in methamphetamine (METH) addiction are still unclear. Here, we measured changes in Pde4b and Atf3 in the cAMP signaling pathway of dopaminergic system components, including the nucleus accumbens (NAc), caudate putamen (CPu) and hippocampus (Hip), in D3R knockout mice(D3R-/-) 1h and 24h after METH-induced behavioral sensitization. We found that knocking out D3R attenuated METH-induced behavioral sensitization, and Pde4b and Atf3 exhibited different expression patterns in brain regions in response to METH. Knocking out D3R suppressed the METH-induced increase in Pde4b in the Hip of mice 24h after the final METH injection and augmented the METH-induced increase in Atf3 in the CPu of mice 1h after the final METH injection. Our study suggests that D3R knockout controls METH-induced behavioral sensitization via regulation of Pde4b and Atf3 in different brain regions. Furthermore, the responses of Pde4b and Atf3 to METH exposure depend on the specific region of the brain involved.
Collapse
|
48
|
Gey M, Wanner R, Schilling C, Pedro MT, Sinske D, Knöll B. Atf3 mutant mice show reduced axon regeneration and impaired regeneration-associated gene induction after peripheral nerve injury. Open Biol 2017; 6:rsob.160091. [PMID: 27581653 PMCID: PMC5008009 DOI: 10.1098/rsob.160091] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/01/2016] [Indexed: 12/27/2022] Open
Abstract
Axon injury in the peripheral nervous system (PNS) induces a regeneration-associated gene (RAG) response. Atf3 (activating transcription factor 3) is such a RAG and ATF3's transcriptional activity might induce ‘effector’ RAGs (e.g. small proline rich protein 1a (Sprr1a), Galanin (Gal), growth-associated protein 43 (Gap43)) facilitating peripheral axon regeneration. We provide a first analysis of Atf3 mouse mutants in peripheral nerve regeneration. In Atf3 mutant mice, facial nerve regeneration and neurite outgrowth of adult ATF3-deficient primary dorsal root ganglia neurons was decreased. Using genome-wide transcriptomics, we identified a neuropeptide-encoding RAG cluster (vasoactive intestinal peptide (Vip), Ngf, Grp, Gal, Pacap) regulated by ATF3. Exogenous administration of neuropeptides enhanced neurite growth of Atf3 mutant mice suggesting that these molecules might be effector RAGs of ATF3's pro-regenerative function. In addition to the induction of growth-promoting molecules, we present data that ATF3 suppresses growth-inhibiting molecules such as chemokine (C-C motif) ligand 2. In summary, we show a pro-regenerative ATF3 function during PNS nerve regeneration involving transcriptional activation of a neuropeptide-encoding RAG cluster. ATF3 is a general injury-inducible factor, therefore ATF3-mediated mechanisms identified herein might apply to other cell and injury types.
Collapse
Affiliation(s)
- Manuel Gey
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Renate Wanner
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Corinna Schilling
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Maria T Pedro
- Department of Neurosurgery, Bezirkskrankenhaus Günzburg, Ulm University, 89081 Ulm, Germany
| | - Daniela Sinske
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
49
|
Tran NQV, Nguyen AN, Takabe K, Yamagata Z, Miyake K. Pre-treatment with amitriptyline causes epigenetic up-regulation of neuroprotection-associated genes and has anti-apoptotic effects in mouse neuronal cells. Neurotoxicol Teratol 2017; 62:1-12. [PMID: 28511916 DOI: 10.1016/j.ntt.2017.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/09/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Antidepressants, such as imipramine and fluoxetine, are known to alter gene expression patterns by inducing changes in the epigenetic status of neuronal cells. There is also some evidence for the anti-apoptotic effect of various groups of antidepressants; however, this effect is complicated and cell-type dependent. Antidepressants of the tricyclic group, in particular amitriptyline, have been suggested to be beneficial in the treatment of neurodegenerative disorders. We examined whether amitriptyline exerts an anti-apoptotic effect via epigenetic mechanisms. Using DNA microarray, we analyzed global gene expression in mouse primary cultured neocortical neurons after treatment with amitriptyline and imipramine. The neuroprotection-associated genes, activating transcription factor 3 (Atf3) and heme oxygenase 1 (Hmox1), were up-regulated at both mRNA and protein levels by treatment with amitriptyline. Quantitative chromatin immunoprecipitation assay revealed that amitriptyline increased enrichments of trimethylation of histone H3 lysine 4 in the promoter regions of Atf3 and Hmox1 and acetylation of histone H3 lysine 9 in the promoter regions of Atf3, which indicate an active epigenetic status. Amitriptyline pre-treatment attenuated 1-methyl-4-phenylpyridinium ion (MPP+)- or amyloid β peptide 1-42 (Aβ1-42)-induced neuronal cell death and inhibited the activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). We found that Atf3 and Hmox1 were also up-regulated after Aβ1-42 treatment, and were further increased when pre-treated with amitriptyline. Interestingly, the highest up-regulation of Atf3 and Hmox1, at least at mRNA level, was observed after co-treatment with Aβ1-42 and amitriptyline, together with the loss of the neuroprotective effect. These findings suggest preconditioning and neuroprotective effects of amitriptyline; however, further investigations are needed for clarifying the contribution of epigenetic up-regulation of Atf3 and Hmox1 genes.
Collapse
Affiliation(s)
- Nguyen Quoc Vuong Tran
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - An Nghia Nguyen
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kyoko Takabe
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Zentaro Yamagata
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kunio Miyake
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
50
|
Bas-Orth C, Tan YW, Lau D, Bading H. Synaptic Activity Drives a Genomic Program That Promotes a Neuronal Warburg Effect. J Biol Chem 2017; 292:5183-5194. [PMID: 28196867 DOI: 10.1074/jbc.m116.761106] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/01/2017] [Indexed: 12/17/2022] Open
Abstract
Synaptic activity drives changes in gene expression to promote long lasting adaptations of neuronal structure and function. One example of such an adaptive response is the buildup of acquired neuroprotection, a synaptic activity- and gene transcription-mediated increase in the resistance of neurons against harmful conditions. A hallmark of acquired neuroprotection is the stabilization of mitochondrial structure and function. We therefore re-examined previously identified sets of synaptic activity-regulated genes to identify genes that are directly linked to mitochondrial function. In mouse and rat primary hippocampal cultures, synaptic activity caused an up-regulation of glycolytic genes and a concomitant down-regulation of genes required for oxidative phosphorylation, mitochondrial biogenesis, and maintenance. Changes in metabolic gene expression were induced by action potential bursting, but not by glutamate bath application activating extrasynaptic NMDA receptors. The specific and coordinate pattern of gene expression changes suggested that synaptic activity promotes a shift of neuronal energy metabolism from oxidative phosphorylation toward aerobic glycolysis, also known as the Warburg effect. The ability of neurons to up-regulate glycolysis has, however, been debated. We therefore used FACS sorting to show that, in mixed neuron glia co-cultures, activity-dependent regulation of metabolic gene expression occurred in neurons. Changes in gene expression were accompanied by changes in the phosphorylation-dependent regulation of the key metabolic enzyme, pyruvate dehydrogenase. Finally, increased synaptic activity caused an increase in the ratio of l-lactate production to oxygen consumption in primary hippocampal cultures. Based on these data we suggest the existence of a synaptic activity-mediated neuronal Warburg effect that may promote mitochondrial homeostasis and neuroprotection.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Yan-Wei Tan
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - David Lau
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- From the Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|