1
|
Saraswat Ohri S, Myers SA, Rood B, Brown BL, Chilton PM, Slomnicki L, Liu Y, Wei GZ, Andres KR, Mohan D, Howard RM, Whittemore SR, Hetman M. Reduced White Matter Damage and Lower Neuroinflammatory Potential of Microglia and Macrophages in Hri/Eif2ak1 -/- Mice After Contusive Spinal Cord Injury. Glia 2025. [PMID: 39760211 DOI: 10.1002/glia.24669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Cellular stressors inhibit general protein synthesis while upregulating stress response transcripts and/or proteins. Phosphorylation of the translation factor eIF2α by one of the several stress-activated kinases is a trigger for such signaling, known as the integrated stress response (ISR). The ISR regulates cell survival and function under stress. Here, germline knockout mice were used to determine contributions by three major ISR kinases, HRI/EIF2AK1, GCN2/EIF2AK4, and PKR//EIF2AK2, to pathogenesis of moderate contusive spinal cord injury (SCI) at the thoracic T9 level. One-day post-injury (dpi), reduced levels of peIF2α were found in Hri-/- and Gcn2-/-, but not in Pkr-/- mice. In addition, Hri-/- mice showed attenuated expression of the downstream ISR transcripts, Atf4 or Chop. Such differential effects of SCI-activated ISR correlated with a strong or moderate enhancement of locomotor recovery in Hri-/- or Gcn2-/- mice, respectively. Hri-/- mice also showed reduced white matter loss, increased content of oligodendrocytes (OL) and attenuated neuroinflammation, including decreased lipid accumulation in microglia/macrophages. Cultured neonatal Hri-/- OLs showed lower ISR cytotoxicity. Moreover, cell autonomous reduction in neuroinflammatory potential was observed in microglia and bone marrow-derived macrophages derived from Hri-/- mice. These data identify HRI as a major positive regulator of SCI-associated secondary injury. In addition, targeting HRI may enable multimodal neuroprotection to enhance functional recovery after SCI.
Collapse
Affiliation(s)
- Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Scott A Myers
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Benjamin Rood
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry & Molecular Genetics, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Brandon L Brown
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Paula M Chilton
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Norton Neuroscience Institute, Louisville, Kentucky, USA
| | - Lukasz Slomnicki
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Yu Liu
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - George Z Wei
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Divya Mohan
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry & Molecular Genetics, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry & Molecular Genetics, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
2
|
Storkebaum E, Rosenblum K, Sonenberg N. Messenger RNA Translation Defects in Neurodegenerative Diseases. Reply. N Engl J Med 2023; 388:2110-2111. [PMID: 37256994 DOI: 10.1056/nejmc2304387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Affiliation(s)
- Erik Storkebaum
- Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| | | | | |
Collapse
|
3
|
Mitostasis, Calcium and Free Radicals in Health, Aging and Neurodegeneration. Biomolecules 2021; 11:biom11071012. [PMID: 34356637 PMCID: PMC8301949 DOI: 10.3390/biom11071012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondria play key roles in ATP supply, calcium homeostasis, redox balance control and apoptosis, which in neurons are fundamental for neurotransmission and to allow synaptic plasticity. Their functional integrity is maintained by mitostasis, a process that involves mitochondrial transport, anchoring, fusion and fission processes regulated by different signaling pathways but mainly by the peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). PGC-1α also favors Ca2+ homeostasis, reduces oxidative stress, modulates inflammatory processes and mobilizes mitochondria to where they are needed. To achieve their functions, mitochondria are tightly connected to the endoplasmic reticulum (ER) through specialized structures of the ER termed mitochondria-associated membranes (MAMs), which facilitate the communication between these two organelles mainly to aim Ca2+ buffering. Alterations in mitochondrial activity enhance reactive oxygen species (ROS) production, disturbing the physiological metabolism and causing cell damage. Furthermore, cytosolic Ca2+ overload results in an increase in mitochondrial Ca2+, resulting in mitochondrial dysfunction and the induction of mitochondrial permeability transition pore (mPTP) opening, leading to mitochondrial swelling and cell death through apoptosis as demonstrated in several neuropathologies. In summary, mitochondrial homeostasis is critical to maintain neuronal function; in fact, their regulation aims to improve neuronal viability and to protect against aging and neurodegenerative diseases.
Collapse
|
4
|
Yamaguchi N, Sawano T, Fukumoto K, Nakatani J, Inoue S, Doe N, Yanagisawa D, Tooyama I, Nakagomi T, Matsuyama T, Tanaka H. Voluntary running exercise after focal cerebral ischemia ameliorates dendritic spine loss and promotes functional recovery. Brain Res 2021; 1767:147542. [PMID: 34077764 DOI: 10.1016/j.brainres.2021.147542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 11/26/2022]
Abstract
Cerebral infarction causes motor, sensory, and cognitive impairments. Although rehabilitation enhances recovery of activities of daily living after cerebral infarction, its mechanism remains elusive due to the lack of reproducibility and low survival rate of brain ischemic model animals. Here, to investigate the relationship between rehabilitative intervention, motor function, and pathophysiological remodeling of the tissue in the ipsilateral hemisphere after cerebral infarction, we took advantage of a highly reproducible model of cerebral infarction using C.B-17/Icr-+/+Jcl mice. In this model, we confirmed that voluntary running exercise improved functional recovery after ischemia. Exercise did not alter the volume of infarction or survived cortex, or the number of NeuN-labeled cells in the peri-infarct cortex. In mice who did not exercise, the number of basal dendritic spines of layer 5 pyramidal cells decreased in the peri-infarct motor cortex, whereas in mice who exercised it remained at the normal level. The voluntary exercise intervention maintained basal dendritic spine density within the peri-infarct area, which may reflect an adaptive remodeling of the surviving neural circuitry that might contribute to promoting the recovery of activities of daily living.
Collapse
Affiliation(s)
- Natsumi Yamaguchi
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
| | - Toshinori Sawano
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan.
| | - Kae Fukumoto
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
| | - Jin Nakatani
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
| | - Shota Inoue
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
| | - Nobutaka Doe
- General Education Center, Hyogo University of Health Sciences, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Takayuki Nakagomi
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan; Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8501, Japan
| | - Hidekazu Tanaka
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan.
| |
Collapse
|
5
|
Ramos-Fernández E, Arrázola MS, Oliva CA, Arredondo SB, Varela-Nallar L, Inestrosa NC. Wnt5a promotes hippocampal postsynaptic development and GluN2B-induced expression via the eIF2α HRI kinase. Sci Rep 2021; 11:7395. [PMID: 33795747 PMCID: PMC8016897 DOI: 10.1038/s41598-021-86708-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Wnt signaling plays a key role in neurodevelopment and neuronal maturation. Specifically, Wnt5a stimulates postsynaptic assemblies, increases glutamatergic neurotransmission and, through calcium signaling, generates nitric oxide (NO). Trying to unveil the molecular pathway triggering these postsynaptic effects, we found that Wnt5a treatment induces a time-dependent increases in the length of the postsynaptic density (PSD), elicits novel synaptic contacts and facilitates F-actin flow both in in vitro and ex vivo models. These effects were partially abolished by the inhibition of the Heme-regulated eukaryotic initiation factor 2α (HRI) kinase, a kinase which phosphorylates the initiation translational factor eIF2α. When phosphorylated, eIF2α normally avoids the translation of proteins not needed during stress conditions, in order to avoid unnecessary energetic expenses. However, phosphorylated eIF2α promotes the translation of some proteins with more than one open reading frame in its 5′ untranslated region. One of these proteins targeted by Wnt-HRI-eIF2α mediated translation is the GluN2B subunit of the NMDA receptor. The identified increase in GluN2B expression correlated with increased NMDA receptor function. Considering that NMDA receptors are crucial for excitatory synaptic transmission, the molecular pathway described here contributes to the understanding of the fast and plastic translational mechanisms activated during learning and memory processes.
Collapse
Affiliation(s)
- Eva Ramos-Fernández
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile. .,École polytechnique fédérale de Lausanne, Lausanne, Switzerland.
| | - Macarena S Arrázola
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile.,Centro de Biología Integrativa, Universidad Mayor, Santiago, Chile
| | - Carolina A Oliva
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile
| | - Sebastián B Arredondo
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Lorena Varela-Nallar
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
6
|
Amyloid Beta-Peptide Increases BACE1 Translation through the Phosphorylation of the Eukaryotic Initiation Factor-2 α. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2739459. [PMID: 33014268 PMCID: PMC7525306 DOI: 10.1155/2020/2739459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/02/2022]
Abstract
Alzheimer's disease (AD) is tightly linked to oxidative stress since amyloid beta-peptide (Aβ) aggregates generate free radicals. Moreover, the aggregation of Aβ is increased by oxidative stress, and the neurotoxicity induced by the oligomers and fibrils is in part mediated by free radicals. Interestingly, it has been reported that oxidative stress can also induce BACE1 transcription and expression. BACE1 is the key enzyme in the cleavage of the amyloid precursor protein to produce Aβ, and the expression of this enzyme has been previously shown to be enhanced in the brains of Alzheimer's patients. Here, we have found that BACE1 expression is increased in the hippocampi from AD patients at both the early (Braak stage II) and late (Braak stage VI) stages of the disease as studied by immunohistochemistry and western blot. To address the role of Aβ and oxidative stress in the regulation of BACE1 expression, we have analyzed the effect of subtoxic concentrations of Aβ oligomers (0.25 μM) and H2O2 (10 mM) on a human neuroblastoma cell line. Firstly, our results show that Aβ oligomers and H2O2 induce an increase of BACE1 mRNA as we studied by qPCR. Regarding BACE1 translation, it is dependent on the phosphorylation of the eukaryotic initiation factor 2α (eIF2α), since BACE1 mRNA bears a 5′UTR that avoids its translation under basal conditions. BACE1 5′UTR contains four upstream initiating codons (uAUGs), and its translation is activated when eIF2α is phosphorylated. Consistently, we have obtained that Aβ oligomers and H2O2 increase the levels of BACE1 and p-eIF2α assayed by western blot and confocal microscopy. Our results suggest that Aβ oligomers increase BACE1 translation by phosphorylating eIF2α in a process that involves oxidative stress and conforms a pathophysiological loop, where the Aβ once aggregated favors its own production continuously by the increase in BACE1 expression as observed in AD patients.
Collapse
|
7
|
Bocai NI, Marcora MS, Belfiori-Carrasco LF, Morelli L, Castaño EM. Endoplasmic Reticulum Stress in Tauopathies: Contrasting Human Brain Pathology with Cellular and Animal Models. J Alzheimers Dis 2020; 68:439-458. [PMID: 30775999 DOI: 10.3233/jad-181021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The accumulation and spreading of protein tau in the human brain are major features of neurodegenerative disorders known as tauopathies. In addition to several subcellular abnormalities, tau aggregation within neurons seems capable of triggering endoplasmic reticulum (ER) stress and the consequent unfolded protein response (UPR). In metazoans, full activation of a complex ER-UPR network may restore proteostasis and ER function or, if stress cannot be solved, commit cells to apoptosis. Due to these alternative outcomes (survival or death), the pharmacological manipulation of ER-UPR has become the focus of potential therapies in many human diseases, including tauopathies. Here we update and analyze the experimental data from human brain, cellular, and animal models linking tau accumulation and ER-UPR. We further discuss mechanistic aspects and put the ER-UPR into perspective as a possible therapeutic target in this group of diseases.
Collapse
Affiliation(s)
- Nadia I Bocai
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María S Marcora
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lautaro F Belfiori-Carrasco
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Laura Morelli
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Eduardo M Castaño
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
8
|
Alvarez-Castelao B, Tom Dieck S, Fusco CM, Donlin-Asp P, Perez JD, Schuman EM. The switch-like expression of heme-regulated kinase 1 mediates neuronal proteostasis following proteasome inhibition. eLife 2020; 9:52714. [PMID: 32329716 PMCID: PMC7224698 DOI: 10.7554/elife.52714] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
We examined the feedback between the major protein degradation pathway, the ubiquitin-proteasome system (UPS), and protein synthesis in rat and mouse neurons. When protein degradation was inhibited, we observed a coordinate dramatic reduction in nascent protein synthesis in neuronal cell bodies and dendrites. The mechanism for translation inhibition involved the phosphorylation of eIF2α, surprisingly mediated by eIF2α kinase 1, or heme-regulated kinase inhibitor (HRI). Under basal conditions, neuronal expression of HRI is barely detectable. Following proteasome inhibition, HRI protein levels increase owing to stabilization of HRI and enhanced translation, likely via the increased availability of tRNAs for its rare codons. Once expressed, HRI is constitutively active in neurons because endogenous heme levels are so low; HRI activity results in eIF2α phosphorylation and the resulting inhibition of translation. These data demonstrate a novel role for neuronal HRI that senses and responds to compromised function of the proteasome to restore proteostasis.
Collapse
Affiliation(s)
| | | | - Claudia M Fusco
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Paul Donlin-Asp
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Julio D Perez
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| |
Collapse
|
9
|
Functions and dysfunctions of nitric oxide in brain. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1949-1967. [DOI: 10.1016/j.bbadis.2018.11.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 02/06/2023]
|
10
|
Guix FX, Sartório CL, Ill-Raga G. BACE1 Translation: At the Crossroads Between Alzheimer's Disease Neurodegeneration and Memory Consolidation. J Alzheimers Dis Rep 2019; 3:113-148. [PMID: 31259308 PMCID: PMC6597968 DOI: 10.3233/adr-180089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human life unfolds not only in time and space, but also in the recollection and interweaving of memories. Therefore, individual human identity depends fully on a proper access to the autobiographical memory. Such access is hindered under pathological conditions such as Alzheimer’s disease, which affects millions of people worldwide. Unfortunately, no effective cure exists to prevent this disorder, the impact of which will rise alarmingly within the next decades. While Alzheimer’s disease is largely considered to be the outcome of amyloid-β (Aβ) peptide accumulation in the brain, conceiving this complex disorder strictly as the result of Aβ-neurotoxicity is perhaps a too straight-line simplification. Instead, complementary to this view, the tableau of molecular disarrangements in the Alzheimer’s disease brain may be reflecting, at least in part, a loss of function phenotype in memory processing. Here we take BACE1 translation and degradation as a gateway to study molecular mechanisms putatively involved in the transition between memory and neurodegeneration. BACE1 participates in the excision of Aβ-peptide from its precursor holoprotein, but plays a role in synaptic plasticity too. Its translation is governed by eIF2α phosphorylation: a hub integrating cellular responses to stress, but also a critical switch in memory consolidation. Paralleling these dualities, the eIF2α-kinase HRI has been shown to be a nitric oxide-dependent physiological activator of hippocampal BACE1 translation. Finally, beholding BACE1 as a representative protease active in the CNS, we venture a new perspective on the cellular basis of memory, which may incorporate neurodegeneration in itself as a drift in memory consolidating systems.
Collapse
Affiliation(s)
- Francesc X Guix
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa-CSIC, Madrid, Spain
| | - Carmem L Sartório
- Division of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Gerard Ill-Raga
- Division of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
11
|
Maltsev AV, Bal NV, Balaban PM. LTP suppression by protein synthesis inhibitors is NO-dependent. Neuropharmacology 2018; 146:276-288. [PMID: 30540927 DOI: 10.1016/j.neuropharm.2018.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/19/2018] [Accepted: 12/08/2018] [Indexed: 01/25/2023]
Abstract
For several decades, the ability of protein synthesis inhibitors (PSI) to suppress the long-term potentiation (LTP) of hippocampal responses is known. It is considered that mechanisms of such impairment are related to a cessation of translation and a delayed depletion of the protein pool required for maintenance of synaptic plasticity. The present study demonstrates that cycloheximide or anisomycin applications reduce amplitudes of the field excitatory postsynaptic potentials as well as the presynaptically mediated form of plasticity, the paired-pulse facilitation after LTP induction in neurons of the CA1 area of hippocampus. We showed that nitric oxide signaling could be one of the pathways that cause the LTP decrease induced by cycloheximide or anisomycin. Inhibitor of the NO synthase, L-NNA or the NO scavenger, PTIO, rescued the late-phase LTP and restored the paired-pulse facilitation up to the control levels. For the first time we have directly measured the nitric oxide production induced by application of the translation blockers in hippocampal neurons using the NO-sensitive dye DAF-FM. Inhibitory analysis demonstrated that changes during protein synthesis blockade downstream the NO signaling cascade are cGMP-independent and apparently are implemented through degradation of target proteins. Prolonged application of the NO donor SNAP impaired the LTP maintenance in the same manner as PSI.
Collapse
Affiliation(s)
- Alexander V Maltsev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia
| | - Natalia V Bal
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia.
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia
| |
Collapse
|
12
|
Huang LE, Guo SH, Thitiseranee L, Yang Y, Zhou YF, Yao YX. N-methyl D-aspartate receptor subtype 2B antagonist, Ro 25-6981, attenuates neuropathic pain by inhibiting postsynaptic density 95 expression. Sci Rep 2018; 8:7848. [PMID: 29777135 PMCID: PMC5959906 DOI: 10.1038/s41598-018-26209-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
Postsynaptic density-95 (PSD-95) is a synaptic scaffolding protein that plays a crucial role in the development of neuropathic pain. However, the underlying mechanism remains unclear. To address the role of PSD-95 in N-methyl-D-aspartate receptor subtype 2B (NR2B) -mediated chronic pain, we investigated the relationship between PSD-95 activation and NR2B function in the spinal cord, by using a rat model of sciatic nerve chronic constriction injury (CCI). We demonstrate that the expression levels of total PSD-95 and cAMP response element binding protein (CREB), as well as phosphorylated NR2B, PSD-95, and CREB, in the spinal dorsal horn, and the interaction of NR2B with PSD-95 were increased in the CCI animals. Intrathecal injection of the selective NR2B antagonist Ro 25-6981 increased paw withdrawal latency, in a thermal pain assessment test. Moreover, repeated treatment with Ro 25-6981 markedly attenuated the thermal hypersensitivity, and inhibited the CCI-induced upregulation of PSD-95 in the spinal dorsal horn. Furthermore, intrathecal injection of the PSD-95 inhibitor strikingly reversed the thermal and mechanical hyperalgesia. Our results suggest that blocking of NR2B signaling in the spinal cord could be used as a therapeutic candidate for treating neuropathic pain.
Collapse
Affiliation(s)
- Ling-Er Huang
- First Affiliated Hospital, Zhejiang University School of Medicine, Department of Anesthesia, Hangzhou, 310003, P.R. China
| | - Shao-Hui Guo
- First Affiliated Hospital, Zhejiang University School of Medicine, Department of Anesthesia, Hangzhou, 310003, P.R. China
| | | | - Yan Yang
- Zhejiang University School of Medicine, Centre for Neuroscience, Hangzhou, 310016, P.R. China
| | - Yan-Feng Zhou
- First Affiliated Hospital, Zhejiang University School of Medicine, Department of Anesthesia, Hangzhou, 310003, P.R. China
| | - Yong-Xing Yao
- First Affiliated Hospital, Zhejiang University School of Medicine, Department of Anesthesia, Hangzhou, 310003, P.R. China.
| |
Collapse
|
13
|
Burwick N, Aktas BH. The eIF2-alpha kinase HRI: a potential target beyond the red blood cell. Expert Opin Ther Targets 2017; 21:1171-1177. [PMID: 29063813 DOI: 10.1080/14728222.2017.1397133] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The eIF2α kinase heme-regulated inhibitor (HRI) is one of four well-described kinases that phosphorylate eIF2α in response to various cell stressors, resulting in reduced ternary complex formation and attenuation of mRNA translation. Although HRI is well known for its role as a heme sensor in erythroid progenitors, pharmacologic activation of HRI has been demonstrated to have anti-cancer activity across a wide range of tumor sub-types. Here, the potential of HRI activators as novel cancer therapeutics is explored. Areas covered: We provide an introduction to eIF2 signaling pathways in general, and specifically review data on the eIF2α kinase HRI in erythroid and non-erythroid cells. We review aspects of targeting eIF2 signaling in cancer and highlight promising data using HRI activators against tumor cells. Expert opinion: Pharmacologic activation of HRI inhibits tumor growth as a single agent without appreciable toxicity in vivo. The ability of HRI activators to provide direct and sustained eIF2α phosphorylation without inducing oxidative stress or broad eIF2α kinase activation may be especially advantageous for tolerability. Combination therapy with established therapeutics may further augment anti-cancer activity to overcome disease resistance.
Collapse
Affiliation(s)
- Nicholas Burwick
- a Division of hematology , VA Puget Sound Health Care System , Seattle , WA , USA.,b Division of Hematology , University of Washington School of Medicine , Seattle WA , USA
| | - Bertal H Aktas
- c Department of Medicine , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| |
Collapse
|
14
|
Kinases of eIF2a Switch Translation of mRNA Subset during Neuronal Plasticity. Int J Mol Sci 2017; 18:ijms18102213. [PMID: 29065505 PMCID: PMC5666893 DOI: 10.3390/ijms18102213] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022] Open
Abstract
Compared to other types of cells, neurons express the largest number of diverse mRNAs, including neuron-specific ones. This mRNA diversity is required for neuronal function, memory storage, maintenance and retrieval. Regulation of translation in neurons is very complicated and involves various proteins. Some proteins, implementing translational control in other cell types, are used by neurons for synaptic plasticity. In this review, we discuss the neuron-specific activity of four kinases: protein kinase R (PKR), PKR-like endoplasmic reticulum kinase (PERK), general control nonderepressible 2 kinase (GCN2), and heme-reguated eIF2α kinase (HRI), the substrate for which is α-subunit of eukaryotic initiation factor 2 (eIF2α). Phosphorylation of eIF2α is necessary for the cell during stress conditions, such as lack of amino acids, energy stress or viral infection. We propose that, during memory formation, neurons use some mechanisms similar to those involved in the cellular stress. The four eIF2α kinases regulate translation of certain mRNAs containing upstream open reading frames (uORFs). These mRNAs encode proteins involved in the processes of long-term potentiation (LTP) or long-term depression (LTD). The review examines some neuronal proteins for which translation regulation by eIF2 was suggested and checked experimentally. Of such proteins, we pay close attention to protein kinase Mζ, which is involved in memory storage and regulated at the translational level.
Collapse
|