1
|
Anzalone M, Karam SA, Briting SRR, Petersen S, Thomsen MB, Babcock AA, Landau AM, Finsen B, Metaxas A. Serotonin-2B receptor (5-HT 2BR) expression and binding in the brain of APP swe/PS1 dE9 transgenic mice and in Alzheimer's disease brain tissue. Neurosci Lett 2025; 844:138013. [PMID: 39426582 DOI: 10.1016/j.neulet.2024.138013] [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: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Despite well-documented dysregulation in central serotonergic signaling in Alzheimer's disease (AD), knowledge about the potential involvement of the serotonin-2B receptor (5-HT2BR) subtype remains sparse. Here, we assessed the levels of 5-HT2BRs in brain tissue from APPswe/PS1dE9 transgenic (TG) mice, AD patients, and adult microglial cells. 5-HT2BR mRNA was measured by RT-qPCR in ageing TG and wild-type (WT) mice, in samples from the middle frontal gyrus of female, AD and control subjects, and in microglia from the cerebral cortex of WT mice. The density of 5-HT2BRs was measured by autoradiography using [3H]RS 127445. Both mouse and human brains had low levels of 5-HT2BR mRNA. In whole-brain mouse samples, mRNA expression was significantly lower in TG mice compared to WT at > 18 months of age. In the Aβ-plaque-burdened neocortex and hippocampus of old TG mice, however, levels of 5-HT2BR mRNA were two-fold higher over control, with similar elevations observed in the Aβ-plaque-burdened frontal cortex of human AD patients. 5-HT2BR mRNA expression varied widely in adult microglia and was higher compared to other cortical cell subtypes. In mice, specific [3H]RS-127445 binding in the cortex was first detected after 3 months of age. The density of 5-HT2BRs was low and overall reduced in TG, compared to WT mice. Binding was detectable but too low to be reliably quantified in the human cortex. Our results document Aβ-associated increases in 5-HT2BR mRNA expression and suggest reduced receptor binding in the context of AD. Studies investigating the functional involvement of microglial 5-HT2BRs in AD are considered relevant.
Collapse
Affiliation(s)
- Marco Anzalone
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Sarmad A Karam
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Sanne R R Briting
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Sussanne Petersen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Majken B Thomsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Alicia A Babcock
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark.
| | - Athanasios Metaxas
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| |
Collapse
|
2
|
Benita BA, Koss KM. Peptide discovery across the spectrum of neuroinflammation; microglia and astrocyte phenotypical targeting, mediation, and mechanistic understanding. Front Mol Neurosci 2024; 17:1443985. [PMID: 39634607 PMCID: PMC11616451 DOI: 10.3389/fnmol.2024.1443985] [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: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 12/07/2024] Open
Abstract
Uncontrolled and chronic inflammatory states in the Central Nervous System (CNS) are the hallmark of neurodegenerative pathology and every injury or stroke-related insult. The key mediators of these neuroinflammatory states are glial cells known as microglia, the resident immune cell at the core of the inflammatory event, and astroglia, which encapsulate inflammatory insults in proteoglycan-rich scar tissue. Since the majority of neuroinflammation is exclusively based on the responses of said glia, their phenotypes have been identified to be on an inflammatory spectrum encompassing developmental, homeostatic, and reparative behaviors as opposed to their ability to affect devastating cell death cascades and scar tissue formation. Recently, research groups have focused on peptide discovery to identify these phenotypes, find novel mechanisms, and mediate or re-engineer their actions. Peptides retain the diverse function of proteins but significantly reduce the activity dependence on delicate 3D structures. Several peptides targeting unique phenotypes of microglia and astroglia have been identified, along with several capable of mediating deleterious behaviors or promoting beneficial outcomes in the context of neuroinflammation. A comprehensive review of the peptides unique to microglia and astroglia will be provided along with their primary discovery methodologies, including top-down approaches using known biomolecules and naïve strategies using peptide and phage libraries.
Collapse
Affiliation(s)
| | - Kyle M. Koss
- Department of Surgery, University of Arizona, Tucson, AZ, United States
- Department of Neurobiology, University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
- Sealy Institute for Drug Discovery (SIDD), University of Texas Medical Branch (UTMB) at Galvestion, Galvestion, TX, United States
| |
Collapse
|
3
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [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: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
4
|
Früholz I, Meyer-Luehmann M. The intricate interplay between microglia and adult neurogenesis in Alzheimer's disease. Front Cell Neurosci 2024; 18:1456253. [PMID: 39360265 PMCID: PMC11445663 DOI: 10.3389/fncel.2024.1456253] [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: 06/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system, play a crucial role in regulating adult neurogenesis and contribute significantly to the pathogenesis of Alzheimer's disease (AD). Under physiological conditions, microglia support and modulate neurogenesis through the secretion of neurotrophic factors, phagocytosis of apoptotic cells, and synaptic pruning, thereby promoting the proliferation, differentiation, and survival of neural progenitor cells (NPCs). However, in AD, microglial function becomes dysregulated, leading to chronic neuroinflammation and impaired neurogenesis. This review explores the intricate interplay between microglia and adult neurogenesis in health and AD, synthesizing recent findings to provide a comprehensive overview of the current understanding of microglia-mediated regulation of adult neurogenesis. Furthermore, it highlights the potential of microglia-targeted therapies to modulate neurogenesis and offers insights into potential avenues for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Iris Früholz
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Zhang D, Rodríguez-Kirby LAR, Lin Y, Song M, Wang L, Wang L, Kanatani S, Jimenez-Beristain T, Dang Y, Zhong M, Kukanja P, Wang S, Chen XL, Gao F, Wang D, Xu H, Lou X, Liu Y, Chen J, Sestan N, Uhlén P, Kriegstein A, Zhao H, Castelo-Branco G, Fan R. Spatial dynamics of mammalian brain development and neuroinflammation by multimodal tri-omics mapping. RESEARCH SQUARE 2024:rs.3.rs-4814866. [PMID: 39184075 PMCID: PMC11343178 DOI: 10.21203/rs.3.rs-4814866/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
The ability to spatially map multiple layers of the omics information over different time points allows for exploring the mechanisms driving brain development, differentiation, arealization, and alterations in disease. Herein we developed and applied spatial tri-omic sequencing technologies, DBiT ARP-seq (spatial ATAC-RNA-Protein-seq) and DBiT CTRP-seq (spatial CUT&Tag-RNA-Protein-seq) together with multiplexed immunofluorescence imaging (CODEX) to map spatial dynamic remodeling in brain development and neuroinflammation. A spatiotemporal tri-omic atlas of the mouse brain was obtained at different stages from postnatal day P0 to P21, and compared to the regions of interest in the human developing brains. Specifically, in the cortical area, we discovered temporal persistence and spatial spreading of chromatin accessibility for the layer-defining transcription factors. In corpus callosum, we observed dynamic chromatin priming of myelin genes across the subregions. Together, it suggests a role for layer specific projection neurons to coordinate axonogenesis and myelination. We further mapped the brain of a lysolecithin (LPC) neuroinflammation mouse model and observed common molecular programs in development and neuroinflammation. Microglia, exhibiting both conserved and distinct programs for inflammation and resolution, are transiently activated not only at the core of the LPC lesion, but also at distal locations presumably through neuronal circuitry. Thus, this work unveiled common and differential mechanisms in brain development and neuroinflammation, resulting in a valuable data resource to investigate brain development, function and disease.
Collapse
Affiliation(s)
- Di Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- These authors contributed equally
| | - Leslie A Rubio Rodríguez-Kirby
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- These authors contributed equally
| | - Yingxin Lin
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Mengyi Song
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Li Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Lijun Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Shigeaki Kanatani
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tony Jimenez-Beristain
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yonglong Dang
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mei Zhong
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Shaohui Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Xinyi Lisa Chen
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Fu Gao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Dejiang Wang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Hang Xu
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xing Lou
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Yang Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Jinmiao Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Per Uhlén
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Arnold Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
6
|
Zhang D, Rubio Rodríguez-Kirby LA, Lin Y, Song M, Wang L, Wang L, Kanatani S, Jimenez-Beristain T, Dang Y, Zhong M, Kukanja P, Wang S, Chen XL, Gao F, Wang D, Xu H, Lou X, Liu Y, Chen J, Sestan N, Uhlén P, Kriegstein A, Zhao H, Castelo-Branco G, Fan R. Spatial dynamics of mammalian brain development and neuroinflammation by multimodal tri-omics mapping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605493. [PMID: 39091821 PMCID: PMC11291146 DOI: 10.1101/2024.07.28.605493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The ability to spatially map multiple layers of the omics information over different time points allows for exploring the mechanisms driving brain development, differentiation, arealization, and alterations in disease. Herein we developed and applied spatial tri-omic sequencing technologies, DBiT ARP-seq (spatial ATAC-RNA-Protein-seq) and DBiT CTRP-seq (spatial CUT&Tag-RNA-Protein-seq) together with multiplexed immunofluorescence imaging (CODEX) to map spatial dynamic remodeling in brain development and neuroinflammation. A spatiotemporal tri-omic atlas of the mouse brain was obtained at different stages from postnatal day P0 to P21, and compared to the regions of interest in the human developing brains. Specifically, in the cortical area, we discovered temporal persistence and spatial spreading of chromatin accessibility for the layer-defining transcription factors. In corpus callosum, we observed dynamic chromatin priming of myelin genes across the subregions. Together, it suggests a role for layer specific projection neurons to coordinate axonogenesis and myelination. We further mapped the brain of a lysolecithin (LPC) neuroinflammation mouse model and observed common molecular programs in development and neuroinflammation. Microglia, exhibiting both conserved and distinct programs for inflammation and resolution, are transiently activated not only at the core of the LPC lesion, but also at distal locations presumably through neuronal circuitry. Thus, this work unveiled common and differential mechanisms in brain development and neuroinflammation, resulting in a valuable data resource to investigate brain development, function and disease.
Collapse
Affiliation(s)
- Di Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- These authors contributed equally
| | - Leslie A Rubio Rodríguez-Kirby
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- These authors contributed equally
| | - Yingxin Lin
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Mengyi Song
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Li Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Lijun Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Shigeaki Kanatani
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tony Jimenez-Beristain
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Yonglong Dang
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mei Zhong
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Shaohui Wang
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Xinyi Lisa Chen
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Fu Gao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Dejiang Wang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Hang Xu
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xing Lou
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Yang Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Jinmiao Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Per Uhlén
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Arnold Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco; San Francisco, CA 94143, USA
- Department of Neurology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
7
|
Guerra-Cantera S, Frago LM, Espinoza-Chavarria Y, Collado-Pérez R, Jiménez-Hernaiz M, Torrecilla-Parra M, Barrios V, Belsham DD, Laursen LS, Oxvig C, Argente J, Chowen JA. Palmitic Acid Modulation of the Insulin-Like Growth Factor System in Hypothalamic Astrocytes and Neurons. Neuroendocrinology 2024; 114:958-974. [PMID: 39043147 DOI: 10.1159/000540442] [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: 11/24/2023] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Insulin-like growth factor (IGF)1 and IGF2 have neuroprotective effects, but less is known regarding how other members of the IGF system, including IGF binding proteins (IGFBPs) and the regulatory proteinase pappalysin-1 (PAPP-A) and its endogenous inhibitor stanniocalcin-2 (STC2) participate in this process. Here, we analyzed whether these members of the IGF system are modified in neurons and astrocytes in response to palmitic acid (PA), a fatty acid that induces cell stress when increased centrally. METHODS Primary hypothalamic astrocyte cultures from male and female PND2 rats and the pro-opiomelanocortin (POMC) neuronal cell line, mHypoA-POMC/GFP-2, were treated with PA, IGF1 or both. To analyze the role of STC2 in astrocytes, siRNA assays were employed. RESULTS In astrocytes of both sexes, PA rapidly increased cell stress factors followed by increased Pappa and Stc2 mRNA levels and then a decrease in Igf1, Igf2, and Igfbp2 expression and cell number. Exogenous IGF1 did not revert these effects. In mHypoA-POMC/GFP-2 neurons, PA reduced cell number and Pomc and Igf1 mRNA levels, and increased Igfbp2 and Stc2, again with no effect of exogenous IGF1. PA increased STC2 expression, but no effects of decreasing its levels by interference assays or exogenous STC2 treatment in astrocytes were found. CONCLUSIONS The response of the IGF system to PA was cell and sex specific, but no protective effects of the IGFs were found. However, the modifications in hypothalamic PAPP-A and STC2 indicate that further studies are required to determine their role in the response to fatty acids and possibly in metabolic control.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Yesenia Espinoza-Chavarria
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Torrecilla-Parra
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Lisbeth S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
8
|
Hou L, Ma J, Feng X, Chen J, Dong BH, Xiao L, Zhang X, Guo B. Caffeic acid and diabetic neuropathy: Investigating protective effects and insulin-like growth factor 1 (IGF-1)-related antioxidative and anti-inflammatory mechanisms in mice. Heliyon 2024; 10:e32623. [PMID: 38975173 PMCID: PMC11225750 DOI: 10.1016/j.heliyon.2024.e32623] [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: 10/09/2023] [Revised: 05/15/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Diabetic neuropathy (DN) represents a common and debilitating complication of diabetes, affecting a significant proportion of patients. Despite available treatments focusing on symptom management, there remains an unmet need for therapies that address the underlying pathophysiology. In pursuit of novel interventions, this study evaluated the therapeutic effects of caffeic acid-a natural phenolic compound prevalent in various foods-on diabetic neuropathy using a mouse model, particularly examining its interaction with the Insulin-like Growth Factor 1 (IGF-1) signaling pathway. Caffeic acid was administered orally at two dosages (5 mg/kg and 10 mg/kg), and a comprehensive set of outcomes including fasting blood glucose levels, body weight, sensory behavior, spinal cord oxidative stress markers, inflammatory cytokines, and components of the IGF-1 signaling cascade were assessed. Additionally, to determine the specific contribution of IGF-1 signaling to the observed benefits, IGF1R inhibitor Picropodophyllin (PPP) was co-administered with caffeic acid. Our results demonstrated that caffeic acid, at both dosages, effectively reduced hyperglycemia and alleviated sensory behavioral deficits in diabetic mice. This was accompanied by a marked decrease in oxidative stress markers and an increase in antioxidant enzyme activities within the spinal cord. Significantly lowered microglial activation and inflammatory cytokine expression highlighted the potent antioxidative and anti-inflammatory effects of caffeic acid. Moreover, increases in both serum and spinal levels of IGF-1, along with elevated phosphorylated IGF1R, implicated the IGF-1 signaling pathway as a mediator of caffeic acid's neuroprotective actions. The partial reversal of caffeic acid's benefits by PPP substantiated the pivotal engagement of IGF-1 signaling in mediating its effects. Our findings delineate the capability of caffeic acid to mitigate DN symptoms, particularly through reducing spinal oxidative stress and inflammation, and pinpoint the integral role of IGF-1 signaling in these protective mechanisms. The insights gleaned from this study not only position caffeic acid as a promising dietary adjunct for managing diabetic neuropathy but also highlight the therapeutic potential of targeting spinal IGF-1 signaling as part of a strategic treatment approach.
Collapse
Affiliation(s)
- Leina Hou
- Department of Anesthesiology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Jiaqi Ma
- Department of Radiology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Xugang Feng
- Department of General Surgery, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Jing Chen
- Department of Medical Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Bu-huai Dong
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| | - Li Xiao
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| | - Xi Zhang
- Department of Pediatric Neurology, Northwest Women and Children's Hospital, Xi'an, 710049, China
| | - Bin Guo
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| |
Collapse
|
9
|
Muzio L, Perego J. CNS Resident Innate Immune Cells: Guardians of CNS Homeostasis. Int J Mol Sci 2024; 25:4865. [PMID: 38732082 PMCID: PMC11084235 DOI: 10.3390/ijms25094865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Although the CNS has been considered for a long time an immune-privileged organ, it is now well known that both the parenchyma and non-parenchymal tissue (meninges, perivascular space, and choroid plexus) are richly populated in resident immune cells. The advent of more powerful tools for multiplex immunophenotyping, such as single-cell RNA sequencing technique and upscale multiparametric flow and mass spectrometry, helped in discriminating between resident and infiltrating cells and, above all, the different spectrum of phenotypes distinguishing border-associated macrophages. Here, we focus our attention on resident innate immune players and their primary role in both CNS homeostasis and pathological neuroinflammation and neurodegeneration, two key interconnected aspects of the immunopathology of multiple sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Lab, IRCCS San Raffaele Scientific Institute, Institute of Experimental Neurology, 20133 Milan, Italy;
| | | |
Collapse
|
10
|
Lewitt MS, Boyd GW. Role of the Insulin-like Growth Factor System in Neurodegenerative Disease. Int J Mol Sci 2024; 25:4512. [PMID: 38674097 PMCID: PMC11049992 DOI: 10.3390/ijms25084512] [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: 03/27/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
The insulin-like growth factor (IGF) system has paracrine and endocrine roles in the central nervous system. There is evidence that IGF signalling pathways have roles in the pathophysiology of neurodegenerative disease. This review focusses on Alzheimer's disease and Parkinson's disease, the two most common neurodegenerative disorders that are increasing in prevalence globally in relation to the aging population and the increasing prevalence of obesity and type 2 diabetes. Rodent models used in the study of the molecular pathways involved in neurodegeneration are described. However, currently, no animal model fully replicates these diseases. Mice with triple mutations in APP, PSEN and MAPT show promise as models for the testing of novel Alzheimer's therapies. While a causal relationship is not proven, the fact that age, obesity and T2D are risk factors in both strengthens the case for the involvement of the IGF system in these disorders. The IGF system is an attractive target for new approaches to management; however, there are gaps in our understanding that first need to be addressed. These include a focus beyond IGF-I on other members of the IGF system, including IGF-II, IGF-binding proteins and the type 2 IGF receptor.
Collapse
Affiliation(s)
- Moira S. Lewitt
- School of Health and Life Sciences, University of the West of Scotland, Paisley PA1 2BE, UK
| | - Gary W. Boyd
- School of Health and Life Sciences, University of the West of Scotland, Hamilton G72 0LH, UK;
| |
Collapse
|
11
|
Chen F, Lu K, Bai N, Hao Y, Wang H, Zhao X, Yue F. Oral administration of ellagic acid mitigates perioperative neurocognitive disorders, hippocampal oxidative stress, and neuroinflammation in aged mice by restoring IGF-1 signaling. Sci Rep 2024; 14:2509. [PMID: 38291199 PMCID: PMC10827749 DOI: 10.1038/s41598-024-53127-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024] Open
Abstract
This study investigates the potential of ellagic acid (EA), a phytochemical with antioxidant and anti-inflammatory properties, in managing perioperative neurocognitive disorders (PND). PND, which represents a spectrum of cognitive impairments often faced by elderly patients, is principally linked to surgical and anesthesia procedures, and heavily impacted by oxidative stress in the hippocampus and microglia-induced neuroinflammation. Employing an aged mice model subjected to abdominal surgery, we delve into EA's ability to counteract postoperative oxidative stress and cerebral inflammation by engaging the Insulin-like growth factor-1 (IGF-1) pathway. Our findings revealed that administering EA orally notably alleviated post-surgical cognitive decline in older mice, a fact that was manifested in improved performance during maze tests. This enhancement in the behavioral performance of the EA-treated mice corresponded with the rejuvenation of IGF-1 signaling, a decrease in oxidative stress markers in the hippocampus (like MDA and carbonylated protein), and an increase in the activity of antioxidant enzymes such as SOD and CAT. Alongside these, we observed a decrease in microglia-driven neuroinflammation in the hippocampus, thus underscoring the antioxidant and anti-inflammatory roles of EA. Interestingly, when EA was given in conjunction with an IGF1R inhibitor, these benefits were annulled, accentuating the pivotal role that the IGF-1 pathway plays in the neuroprotective potential of EA. Hence, EA could serve as a potent candidate for safeguarding against PND in older patients by curbing oxidative stress and neuroinflammation through the activation of the IGF-1 pathway.
Collapse
Affiliation(s)
- Fang Chen
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Kai Lu
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Ning Bai
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Yabo Hao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Hui Wang
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Xinrong Zhao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Fang Yue
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
12
|
Alassaf M, Rajan A. Diet-induced glial insulin resistance impairs the clearance of neuronal debris in Drosophila brain. PLoS Biol 2023; 21:e3002359. [PMID: 37934726 PMCID: PMC10629620 DOI: 10.1371/journal.pbio.3002359] [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: 03/09/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023] Open
Abstract
Obesity significantly increases the risk of developing neurodegenerative disorders, yet the precise mechanisms underlying this connection remain unclear. Defects in glial phagocytic function are a key feature of neurodegenerative disorders, as delayed clearance of neuronal debris can result in inflammation, neuronal death, and poor nervous system recovery. Mounting evidence indicates that glial function can affect feeding behavior, weight, and systemic metabolism, suggesting that diet may play a role in regulating glial function. While it is appreciated that glial cells are insulin sensitive, whether obesogenic diets can induce glial insulin resistance and thereby impair glial phagocytic function remains unknown. Here, using a Drosophila model, we show that a chronic obesogenic diet induces glial insulin resistance and impairs the clearance of neuronal debris. Specifically, obesogenic diet exposure down-regulates the basal and injury-induced expression of the glia-associated phagocytic receptor, Draper. Constitutive activation of systemic insulin release from Drosophila insulin-producing cells (IPCs) mimics the effect of diet-induced obesity on glial Draper expression. In contrast, genetically attenuating systemic insulin release from the IPCs rescues diet-induced glial insulin resistance and Draper expression. Significantly, we show that genetically stimulating phosphoinositide 3-kinase (Pi3k), a downstream effector of insulin receptor (IR) signaling, rescues high-sugar diet (HSD)-induced glial defects. Hence, we establish that obesogenic diets impair glial phagocytic function and delays the clearance of neuronal debris.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, Washington, United States of America
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, Washington, United States of America
| |
Collapse
|
13
|
Guerra-Cantera S, Frago LM, Jiménez-Hernaiz M, Collado-Pérez R, Canelles S, Ros P, García-Piqueras J, Pérez-Nadador I, Barrios V, Argente J, Chowen JA. The metabolic effects of resumption of a high fat diet after weight loss are sex dependent in mice. Sci Rep 2023; 13:13227. [PMID: 37580448 PMCID: PMC10425431 DOI: 10.1038/s41598-023-40514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023] Open
Abstract
Dietary restriction is a frequent strategy for weight loss, but adherence is difficult and returning to poor dietary habits can result in more weight gain than that previously lost. How weight loss due to unrestricted intake of a healthy diet affects the response to resumption of poor dietary habits is less studied. Moreover, whether this response differs between the sexes and if the insulin-like growth factor (IGF) system, sex dependent and involved in metabolic control, participates is unknown. Mice received rodent chow (6% Kcal from fat) or a high-fat diet (HFD, 62% Kcal from fat) for 4 months, chow for 3 months plus 1 month of HFD, or HFD for 2 months, chow for 1 month then HFD for 1 month. Males and females gained weight on HFD and lost weight when returned to chow at different rates (p < 0.001), but weight gain after resumption of HFD intake was not affected by previous weight loss in either sex. Glucose metabolism was more affected by HFD, as well as the re-exposure to HFD after weight loss, in males. This was associated with increases in hypothalamic mRNA levels of IGF2 (p < 0.01) and IGF binding protein (IGFBP) 2 (p < 0.05), factors involved in glucose metabolism, again only in males. Likewise, IGF2 increased IGFBP2 mRNA levels only in hypothalamic astrocytes from males (p < 0.05). In conclusion, the metabolic responses to dietary changes were less severe and more delayed in females and the IGF system might be involved in some of the sex specific observations.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Department of Endocrinology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge García-Piqueras
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Iris Pérez-Nadador
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain.
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
| |
Collapse
|
14
|
Tu X, Jain A, Parra Bueno P, Decker H, Liu X, Yasuda R. Local autocrine plasticity signaling in single dendritic spines by insulin-like growth factors. SCIENCE ADVANCES 2023; 9:eadg0666. [PMID: 37531435 PMCID: PMC10396292 DOI: 10.1126/sciadv.adg0666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
The insulin superfamily of peptides is essential for homeostasis as well as neuronal plasticity, learning, and memory. Here, we show that insulin-like growth factors 1 and 2 (IGF1 and IGF2) are differentially expressed in hippocampal neurons and released in an activity-dependent manner. Using a new fluorescence resonance energy transfer sensor for IGF1 receptor (IGF1R) with two-photon fluorescence lifetime imaging, we find that the release of IGF1 triggers rapid local autocrine IGF1R activation on the same spine and more than several micrometers along the stimulated dendrite, regulating the plasticity of the activated spine in CA1 pyramidal neurons. In CA3 neurons, IGF2, instead of IGF1, is responsible for IGF1R autocrine activation and synaptic plasticity. Thus, our study demonstrates the cell type-specific roles of IGF1 and IGF2 in hippocampal plasticity and a plasticity mechanism mediated by the synthesis and autocrine signaling of IGF peptides in pyramidal neurons.
Collapse
Affiliation(s)
- Xun Tu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA
| | - Anant Jain
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Paula Parra Bueno
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Helena Decker
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Xiaodan Liu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| |
Collapse
|
15
|
Xie M, Gu S, Hong Y, Liu Y, Rong X, Lu W, Liu H, Algradi AM, Naseem A, Shu Z, Wang Q. Study on the mechanism of Coptis chinensis Franch. And its main active components in treating Alzheimer's disease based on SCFAs using Orbitrap Fusion Lumos Tribrid MS. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116392. [PMID: 37028611 DOI: 10.1016/j.jep.2023.116392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Coptis chinensis Franch. (CCF), as an extensively used traditional Chinese medicine, has therapeutic effects on Alzheimer's disease (AD), but its mechanism of action has not yet been elucidated. AIM OF THE STUDY This study aims to reveal the mechanism of action of CCF via the gut-brain axis, and provide a new strategy for the clinical treatment of AD. MATERIALS AND METHODS APPswe/PS1ΔE9 mice were used as AD models, and were given CCF extract by intragastric administration. Barnes maze was used to test the therapeutic effect of CCF on the treatment of AD. To reveal the mechanism of action of CCF in the treatment of AD, Vanquish Flex UHPLC-orbitrap fusion lumos mass was chosen to detect endogenous differential metabolite; MetaboAnalyst 5.0 was applied to derive relevant metabolic pathways; similarly, to explore the effects of CCF on the gut-brain axis, Vanquish Flex UPLC-Orbitrap fusion lumos mass was utilized to detect the changes in the content of SCFAs in AD mice after CCF administration; the prototype components and metabolites in CCF were identified by UPLC/ESI/qTOF-MS, then their effects on Bifidobacterium breve were explored. RESULTS CCF shortened the latency time of AD mice, improved the target quadrant ratio of AD mice, and made the maze roadmap simpler of AD mice; CCF regulated fifteen potential metabolites of AD mice, interestingly, ILA (indole-3-lactic acid) in SCFAs (short-chain fatty acids) was also included; CCF acted on histidine and phenylalanine metabolic pathways of AD mice; CCF increased the contents of acetic acid and ILA in AD mice; magnoflorine, jatrorrhizine, coptisine, groenlandicine, thalifendine, palmatine, berberine, epiberberine, hydroxylated jatrorrhizine, and 3-methoxydemethyleneberberine in CCF were detected in fecal samples of AD mice; magnoflorine, palmatrubine, 13-methylberberine, berberine, coptisine, and palmatine promoted the growth of Bifidobacterium breve. CONCLUSIONS we have demonstrated that CCF acts on the gut-brain axis by regulating SCFAs to treat AD.
Collapse
Affiliation(s)
- Minzhen Xie
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Siqi Gu
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Hong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yan Liu
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xiaohui Rong
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Wanying Lu
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Heng Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Adnan Mohammed Algradi
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Anam Naseem
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - ZunPeng Shu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Qi Wang
- Department of Medicinal Chemistry and Natural Medicinal Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
16
|
Sullivan M, Fernandez-Aranda F, Camacho-Barcia L, Harkin A, Macrì S, Mora-Maltas B, Jiménez-Murcia S, O'Leary A, Ottomana AM, Presta M, Slattery D, Scholtz S, Glennon JC. Insulin and Disorders of Behavioural Flexibility. Neurosci Biobehav Rev 2023; 150:105169. [PMID: 37059405 DOI: 10.1016/j.neubiorev.2023.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Behavioural inflexibility is a symptom of neuropsychiatric and neurodegenerative disorders such as Obsessive-Compulsive Disorder, Autism Spectrum Disorder and Alzheimer's Disease, encompassing the maintenance of a behaviour even when no longer appropriate. Recent evidence suggests that insulin signalling has roles apart from its regulation of peripheral metabolism and mediates behaviourally-relevant central nervous system (CNS) functions including behavioural flexibility. Indeed, insulin resistance is reported to generate anxious, perseverative phenotypes in animal models, with the Type 2 diabetes medication metformin proving to be beneficial for disorders including Alzheimer's Disease. Structural and functional neuroimaging studies of Type 2 diabetes patients have highlighted aberrant connectivity in regions governing salience detection, attention, inhibition and memory. As currently available therapeutic strategies feature high rates of resistance, there is an urgent need to better understand the complex aetiology of behaviour and develop improved therapeutics. In this review, we explore the circuitry underlying behavioural flexibility, changes in Type 2 diabetes, the role of insulin in CNS outcomes and mechanisms of insulin involvement across disorders of behavioural inflexibility.
Collapse
Affiliation(s)
- Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Fernando Fernandez-Aranda
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Lucía Camacho-Barcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrew Harkin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Bernat Mora-Maltas
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Aet O'Leary
- University Hospital Frankfurt, Frankfurt, Germany
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Neuroscience Unit, Department of Medicine, University of Parma, 43100 Parma, Italy
| | - Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | - Jeffrey C Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
17
|
Alassaf M, Rajan A. Diet-Induced Glial Insulin Resistance Impairs The Clearance Of Neuronal Debris. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531940. [PMID: 36945507 PMCID: PMC10028983 DOI: 10.1101/2023.03.09.531940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Obesity significantly increases the risk of developing neurodegenerative disorders, yet the precise mechanisms underlying this connection remain unclear. Defects in glial phagocytic function are a key feature of neurodegenerative disorders, as delayed clearance of neuronal debris can result in inflammation, neuronal death, and poor nervous system recovery. Mounting evidence indicates that glial function can affect feeding behavior, weight, and systemic metabolism, suggesting that diet may play a role in regulating glial function. While it is appreciated that glial cells are insulin sensitive, whether obesogenic diets can induce glial insulin resistance and thereby impair glial phagocytic function remains unknown. Here, using a Drosophila model, we show that a chronic obesogenic diet induces glial insulin resistance and impairs the clearance of neuronal debris. Specifically, obesogenic diet exposure downregulates the basal and injury-induced expression of the glia-associated phagocytic receptor, Draper. Constitutive activation of systemic insulin release from Drosophila Insulin-producing cells (IPCs) mimics the effect of diet-induced obesity on glial draper expression. In contrast, genetically attenuating systemic insulin release from the IPCs rescues diet-induced glial insulin resistance and draper expression. Significantly, we show that genetically stimulating Phosphoinositide 3-kinase (PI3K), a downstream effector of Insulin receptor signaling, rescues HSD-induced glial defects. Hence, we establish that obesogenic diets impair glial phagocytic function and delays the clearance of neuronal debris.
Collapse
|
18
|
Wang Q, Zheng J, Pettersson S, Reynolds R, Tan EK. The link between neuroinflammation and the neurovascular unit in synucleinopathies. SCIENCE ADVANCES 2023; 9:eabq1141. [PMID: 36791205 PMCID: PMC9931221 DOI: 10.1126/sciadv.abq1141] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 01/19/2023] [Indexed: 05/28/2023]
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glial cells, and neurons. As a fundamental functional module in the central nervous system, the NVU maintains homeostasis in the microenvironment and the integrity of the blood-brain barrier. Disruption of the NVU and interactions among its components are involved in the pathophysiology of synucleinopathies, which are characterized by the pathological accumulation of α-synuclein. Neuroinflammation contributes to the pathophysiology of synucleinopathies, including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. This review aims to summarize the neuroinflammatory response of glial cells and vascular cells in the NVU. We also review neuroinflammation in the context of the cross-talk between glial cells and vascular cells, between glial cells and pericytes, and between microglia and astroglia. Last, we discuss how α-synuclein affects neuroinflammation and how neuroinflammation influences the aggregation and spread of α-synuclein and analyze different properties of α-synuclein in synucleinopathies.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Karolinska Institutet, Department of Odontology, 171 77 Solna, Sweden
- Faculty of Medical Sciences, Sunway University, Subang Jaya, 47500 Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
19
|
An Early Enriched Experience Drives an Activated Microglial Profile at Site of Corrective Neuroplasticity in Ten-m3 Knock-Out Mice. eNeuro 2023; 10:ENEURO.0162-22.2022. [PMID: 36635245 PMCID: PMC9831145 DOI: 10.1523/eneuro.0162-22.2022] [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: 04/20/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 12/15/2022] Open
Abstract
Environmental enrichment (EE) is beneficial for brain development and function, but our understanding of its capacity to drive circuit repair, the underlying mechanisms, and how this might vary with age remains limited. Ten-m3 knock-out (KO) mice exhibit a dramatic and stereotyped mistargeting of ipsilateral retinal inputs to the thalamus, resulting in visual deficits. We have recently shown a previously unexpected capacity for EE during early postnatal life (from birth for six weeks) to drive the partial elimination of miswired axonal projections, along with a recovery of visually mediated behavior, but the timeline of this repair was unclear. Here, we reveal that with just 3.5 weeks of EE from birth, Ten-m3 KOs exhibit a partial behavioral rescue, accompanied by pruning of the most profoundly miswired retinogeniculate terminals. Analysis suggests that the pruning is underway at this time point, providing an ideal opportunity to probe potential mechanisms. With the shorter EE-period, we found a localized increase in microglial density and activation profile within the identified geniculate region where corrective pruning was observed. No comparable response to EE was found in age-matched wild-type (WT) mice. These findings identify microglia as a potential mechanistic link through which EE drives the elimination of miswired neural circuits during early postnatal development. Activity driven, atypical recruitment of microglia to prune aberrant connectivity and restore function may have important therapeutic implications for neurodevelopmental disorders such as autistic spectrum disorder.
Collapse
|
20
|
Dabin R, Wei C, Liang S, Ke C, Zhihan W, Ping Z. Astrocytic IGF-1 and IGF-1R Orchestrate Mitophagy in Traumatic Brain Injury via Exosomal miR-let-7e. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3504279. [PMID: 36062186 PMCID: PMC9433209 DOI: 10.1155/2022/3504279] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/16/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022]
Abstract
Defective brain hormonal signaling and autophagy have been associated with neurodegeneration after brain insults, characterized by neuronal loss and cognitive dysfunction. However, few studies have linked them in the context of brain injury. Insulin-like growth factor-1 (IGF-1) is an important hormone that contributes to growth, cell proliferation, and autophagy and is also expressed in the brain. Here, we assessed the clinical data from TBI patients and performed both in vitro and in vivo experiments with proteomic and gene-chip analysis to assess the functions of IGF-1 in mitophagy following TBI. We show that reduced plasma IGF-1 is correlated with cognition in TBI patients. Overexpression of astrocytic IGF-1 improves cognitive dysfunction and mitophagy in TBI mice. Mechanically, proteomics data show that the IGF-1-related NF-κB pathway transcriptionally regulates decapping mRNA2 (Dcp2) and miR-let-7, together with IGF-1R to orchestrate mitophagy in TBI. Finally, we demonstrate that brain injury induces impaired mitophagy at the chronic stage and that IGF-1 treatment could facilitate the mitophagy markers via exosomal miR-let-7e. By showing that IGF-1 is an important mediator of the beneficial effect of the neural-endocrine network in TBI models, our findings place IGF-1/IGF-1R as a potential target capable of noncoding RNAs and opposing mitophagy failure and cognitive impairment in TBI.
Collapse
Affiliation(s)
- Ren Dabin
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Chen Wei
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Shu Liang
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai, China
| | - Cao Ke
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Wang Zhihan
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Zheng Ping
- Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| |
Collapse
|
21
|
Chen P, Wang W, Liu R, Lyu J, Zhang L, Li B, Qiu B, Tian A, Jiang W, Ying H, Jing R, Wang Q, Zhu K, Bai R, Zeng L, Duan S, Liu C. Olfactory sensory experience regulates gliomagenesis via neuronal IGF1. Nature 2022; 606:550-556. [PMID: 35545672 DOI: 10.1038/s41586-022-04719-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/01/2022] [Indexed: 01/03/2023]
Abstract
Animals constantly receive various sensory stimuli, such as odours, sounds, light and touch, from the surrounding environment. These sensory inputs are essential for animals to search for food and avoid predators, but they also affect their physiological status, and may cause diseases such as cancer. Malignant gliomas-the most lethal form of brain tumour1-are known to intimately communicate with neurons at the cellular level2,3. However, it remains unclear whether external sensory stimuli can directly affect the development of malignant glioma under normal living conditions. Here we show that olfaction can directly regulate gliomagenesis. In an autochthonous mouse model that recapitulates adult gliomagenesis4-6 originating in oligodendrocyte precursor cells (OPCs), gliomas preferentially emerge in the olfactory bulb-the first relay of brain olfactory circuitry. Manipulating the activity of olfactory receptor neurons (ORNs) affects the development of glioma. Mechanistically, olfaction excites mitral and tufted (M/T) cells, which receive sensory information from ORNs and release insulin-like growth factor 1 (IGF1) in an activity-dependent manner. Specific knockout of Igf1 in M/T cells suppresses gliomagenesis. In addition, knocking out the IGF1 receptor in pre-cancerous mutant OPCs abolishes the ORN-activity-dependent mitogenic effects. Our findings establish a link between sensory experience and gliomagenesis through their corresponding sensory neuronal circuits.
Collapse
Affiliation(s)
- Pengxiang Chen
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China.,Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Wei Wang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Rui Liu
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Jiahui Lyu
- Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, P.R. China
| | - Lei Zhang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China.,Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Baizhou Li
- Department of Pathology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Biying Qiu
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China.,Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Anhao Tian
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Wenhong Jiang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China.,Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Honggang Ying
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China
| | - Rui Jing
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China.,Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Qianqian Wang
- Laboratory Animal Center of Zhejiang University, Hangzhou, P.R. China
| | - Keqing Zhu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Ruiliang Bai
- Department of Physical Medicine and Rehabilitation of The Affiliated Sir Run Shumen Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Linghui Zeng
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, P.R. China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China.,Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, P.R. China.,The Institute of Brain and Cognitive Sciences, Zhejiang University City College, Hangzhou, P.R. China.,Chuanqi Research and Development Center of Zhejiang University, Hangzhou, P.R. China
| | - Chong Liu
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, P.R. China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, P.R. China. .,The Institute of Brain and Cognitive Sciences, Zhejiang University City College, Hangzhou, P.R. China. .,Chuanqi Research and Development Center of Zhejiang University, Hangzhou, P.R. China.
| |
Collapse
|
22
|
Sivasaravanaparan M, Olesen LØ, Severino M, von Linstow MCU, Lambertsen KL, Gramsbergen JB, Hasselstrøm J, Metaxas A, Wiborg O, Finsen B. Efficacy of Chronic Paroxetine Treatment in Mitigating Amyloid Pathology and Microgliosis in APPSWE/PS1ΔE9 Transgenic Mice. J Alzheimers Dis 2022; 87:685-699. [PMID: 35342093 DOI: 10.3233/jad-220019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Modulation of serotonergic signaling by treatment with selective serotonin reuptake inhibitors (SSRIs) has been suggested to mitigate amyloid-β (Aβ) pathology in Alzheimer's disease, in addition to exerting an anti-depressant action. OBJECTIVE To investigate the efficacy of chronic treatment with the SSRI paroxetine, in mitigating Aβ pathology and Aβ plaque-induced microgliosis in the hippocampus of 18-month-old APPswe/PS1 ΔE9 mice. METHODS Plaque-bearing APPswe/PS1 ΔE9 and wildtype mice were treated with paroxetine per os at a dose of 5 mg/kg/day, from 9 to 18 months of age. The per os treatment was monitored by recording of the body weights and serum paroxetine concentrations, and by assessment of the serotonin transporter occupancy by [3H]DASB-binding in wildtype mice. Additionally, 5,7-dihydroxytryptamine was administered to 9-month-old APPswe/PS1 ΔE9 mice, to examine the effect of serotonin depletion on Aβ pathology. Aβ pathology was evaluated by Aβ plaque load estimation and the Aβ 42/Aβ 40 ratio by ELISA. RESULTS Paroxetine treatment led to > 80% serotonin transporter occupancy. The treatment increased the body weight of wildtype mice, but not of APPswe/PS1 ΔE9 mice. The treatment had no effect on the Aβ plaque load (p = 0.39), the number and size of plaques, or the Aβ plaque-induced increases in microglial numbers in the dentate gyrus. Three months of serotonin depletion did not significantly impact the Aβ plaque load or Aβ 42/Aβ 40 ratio in APPswe/PS1 ΔE9 mice at 12 months. CONCLUSION Our results show that chronic treatment with the SSRI paroxetine does not mitigate Aβ pathology and Aβ plaque-induced microgliosis in the hippocampus of APPswe/PS1 ΔE9 mice.
Collapse
Affiliation(s)
- Mithula Sivasaravanaparan
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Denmark
| | | | - Maurizio Severino
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Denmark
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, Denmark
| | - Jan Bert Gramsbergen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Denmark
| | | | - Athanasios Metaxas
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Denmark.,Department of Life Sciences, School of Science, European University Cyprus, Nicosia, Cyprus
| | - Ove Wiborg
- Department of Clinical Medicine, Aarhus University Hospital, Denmark.,Department of Health Science and Technology, Aalborg University, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Denmark.,Department of Clinical Research, BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, University of Southern Denmark, Denmark
| |
Collapse
|
23
|
von Linstow CU, Waider J, Bergh MSS, Anzalone M, Madsen C, Nicolau AB, Wirenfeldt M, Lesch KP, Finsen B. The Combined Effects of Amyloidosis and Serotonin Deficiency by Tryptophan Hydroxylase-2 Knockout Impacts Viability of the APP/PS1 Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2021; 85:1283-1300. [DOI: 10.3233/jad-210581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: A decline of brain serotonin (5-HT) is held responsible for the changes in mood that can be observed in Alzheimer’s disease (AD). However, 5-HT’ergic signaling is also suggested to reduce the production of pathogenic amyloid-4β (Aβ). Objective: To investigate the effect of targeted inactivation of tryptophan hydroxylase-2 (Tph2), which is essential for neuronal 5-HT synthesis, on amyloidosis in amyloid precursor protein (APP)swe/presenilin 1 (PS1) ΔE9 transgenic mice. Methods: Triple-transgenic (3xTg) APP/PS1 mice with partial (+/-) or complete Tph2 knockout (–/–) were allowed to survive until 6 months old with APP/PS1, Tph2–/–, and wildtype mice. Survival and weight were recorded. Levels of Aβ 42/40/38, soluble APPα (sAβPPα) and sAβPPβ, and cytokines were analyzed by mesoscale, neurotransmitters by mass spectrometry, and gene expression by quantitative PCR. Tph2, microglia, and Aβ were visualized histologically. Results: Tph2 inactivation in APP/PS1 mice significantly reduced viability, without impacting soluble and insoluble Aβ 42 and Aβ 40 in neocortex and hippocampus, and with only mild changes of soluble Aβ 42/Aβ 40. However, sAβPPα and sAβPPβ in hippocampus and Aβ 38 and Aβ 40 in cerebrospinal fluid were reduced. 3xTg–/–mice were devoid of Tph2 immunopositive fibers and 5-HT. Cytokines were unaffected by genotype, as were neocortical TNF, HTR2a and HTR2b mRNA levels in Tph2–/– mice. Microglia clustered around Aβ plaques regardless of genotype. Conclusion: The results suggest that Tph2 inactivation influences AβPP processing, at least in the hippocampus, although levels of Aβ are unchanged. The reduced viability of 3xTg–/–mice could indicate that 5-HT protects against the seizures that can impact the viability of APP/PS1 mice.
Collapse
Affiliation(s)
- Christian Ulrich von Linstow
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Würzburg, Germany
| | - Marianne Skov-Skov Bergh
- Section for Drug Abuse Research, Department of Forensic Sciences, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Marco Anzalone
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Cecilie Madsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Aina Battle Nicolau
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Martin Wirenfeldt
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
- Department of Pathology, Institute of Clinical Science, Odense University Hospital, Denmark
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- BRIDGE - Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
24
|
Shinjyo N, Kita K. Infection and Immunometabolism in the Central Nervous System: A Possible Mechanistic Link Between Metabolic Imbalance and Dementia. Front Cell Neurosci 2021; 15:765217. [PMID: 34795562 PMCID: PMC8592913 DOI: 10.3389/fncel.2021.765217] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndromes are frequently associated with dementia, suggesting that the dysregulation of energy metabolism can increase the risk of neurodegeneration and cognitive impairment. In addition, growing evidence suggests the link between infections and brain disorders, including Alzheimer's disease. The immune system and energy metabolism are in an intricate relationship. Infection triggers immune responses, which are accompanied by imbalance in cellular and organismal energy metabolism, while metabolic disorders can lead to immune dysregulation and higher infection susceptibility. In the brain, the activities of brain-resident immune cells, including microglia, are associated with their metabolic signatures, which may be affected by central nervous system (CNS) infection. Conversely, metabolic dysregulation can compromise innate immunity in the brain, leading to enhanced CNS infection susceptibility. Thus, infection and metabolic imbalance can be intertwined to each other in the etiology of brain disorders, including dementia. Insulin and leptin play pivotal roles in the regulation of immunometabolism in the CNS and periphery, and dysfunction of these signaling pathways are associated with cognitive impairment. Meanwhile, infectious complications are often comorbid with diabetes and obesity, which are characterized by insulin resistance and leptin signaling deficiency. Examples include human immunodeficiency virus (HIV) infection and periodontal disease caused by an oral pathogen Porphyromonas gingivalis. This review explores potential interactions between infectious agents and insulin and leptin signaling pathways, and discuss possible mechanisms underlying the relationship between infection, metabolic dysregulation, and brain disorders, particularly focusing on the roles of insulin and leptin.
Collapse
Affiliation(s)
- Noriko Shinjyo
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan.,Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan.,Department of Host-Defense Biochemistry, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
25
|
Vinuesa A, Pomilio C, Gregosa A, Bentivegna M, Presa J, Bellotto M, Saravia F, Beauquis J. Inflammation and Insulin Resistance as Risk Factors and Potential Therapeutic Targets for Alzheimer's Disease. Front Neurosci 2021; 15:653651. [PMID: 33967682 PMCID: PMC8102834 DOI: 10.3389/fnins.2021.653651] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Overnutrition and modern diets containing high proportions of saturated fat are among the major factors contributing to a low-grade state of inflammation, hyperglycemia and dyslipidemia. In the last decades, the global rise of type 2 diabetes and obesity prevalence has elicited a great interest in understanding how changes in metabolic function lead to an increased risk for premature brain aging and the development of neurodegenerative disorders such as Alzheimer's disease (AD). Cognitive impairment and decreased neurogenic capacity could be a consequence of metabolic disturbances. In these scenarios, the interplay between inflammation and insulin resistance could represent a potential therapeutic target to prevent or ameliorate neurodegeneration and cognitive impairment. The present review aims to provide an update on the impact of metabolic stress pathways on AD with a focus on inflammation and insulin resistance as risk factors and therapeutic targets.
Collapse
Affiliation(s)
- Angeles Vinuesa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Pomilio
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Amal Gregosa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melisa Bentivegna
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jessica Presa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina Bellotto
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Beauquis
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
26
|
Eshraghi M, Adlimoghaddam A, Mahmoodzadeh A, Sharifzad F, Yasavoli-Sharahi H, Lorzadeh S, Albensi BC, Ghavami S. Alzheimer's Disease Pathogenesis: Role of Autophagy and Mitophagy Focusing in Microglia. Int J Mol Sci 2021; 22:3330. [PMID: 33805142 PMCID: PMC8036323 DOI: 10.3390/ijms22073330] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/14/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurological disorder, and currently, there is no cure for it. Several pathologic alterations have been described in the brain of AD patients, but the ultimate causative mechanisms of AD are still elusive. The classic hallmarks of AD, including amyloid plaques (Aβ) and tau tangles (tau), are the most studied features of AD. Unfortunately, all the efforts targeting these pathologies have failed to show the desired efficacy in AD patients so far. Neuroinflammation and impaired autophagy are two other main known pathologies in AD. It has been reported that these pathologies exist in AD brain long before the emergence of any clinical manifestation of AD. Microglia are the main inflammatory cells in the brain and are considered by many researchers as the next hope for finding a viable therapeutic target in AD. Interestingly, it appears that the autophagy and mitophagy are also changed in these cells in AD. Inside the cells, autophagy and inflammation interact in a bidirectional manner. In the current review, we briefly discussed an overview on autophagy and mitophagy in AD and then provided a comprehensive discussion on the role of these pathways in microglia and their involvement in AD pathogenesis.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA;
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Aida Adlimoghaddam
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; (A.A.); (B.C.A.)
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Farzaneh Sharifzad
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (F.S.); (H.Y.-S.)
| | - Hamed Yasavoli-Sharahi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (F.S.); (H.Y.-S.)
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Benedict C. Albensi
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; (A.A.); (B.C.A.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine, Katowice School of Technology, 40-555 Katowice, Poland
| |
Collapse
|
27
|
Mygind L, Bergh MSS, Tejsi V, Vaitheeswaran R, Lambertsen KL, Finsen B, Metaxas A. Tumor Necrosis Factor (TNF) Is Required for Spatial Learning and Memory in Male Mice under Physiological, but Not Immune-Challenged Conditions. Cells 2021; 10:608. [PMID: 33803476 PMCID: PMC8002217 DOI: 10.3390/cells10030608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/23/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence demonstrates that inflammatory cytokines-such as tumor necrosis factor (TNF)-are produced at low levels in the brain under physiological conditions and may be crucial for synaptic plasticity, neurogenesis, learning and memory. Here, we examined the effects of developmental TNF deletion on spatial learning and memory using 11-13-month-old TNF knockout (KO) and C57BL6/J wild-type (WT) mice. The animals were tested in the Barnes maze (BM) arena under baseline conditions and 48 h following an injection of the endotoxin lipopolysaccharide (LPS), which was administered at a dose of 0.5 mg/kg. Vehicle-treated KO mice were impaired compared to WT mice during the acquisition and memory-probing phases of the BM test. No behavioral differences were observed between WT and TNF-KO mice after LPS treatment. Moreover, there were no differences in the hippocampal content of glutamate and noradrenaline between groups. The effects of TNF deletion on spatial learning and memory were observed in male, but not female mice, which were not different compared to WT mice under baseline conditions. These results indicate that TNF is required for spatial learning and memory in male mice under physiological, non-inflammatory conditions, however not following the administration of LPS. Inflammatory signalling can thereby modulate spatial cognition in male subjects, highlighting the importance of sex- and probably age-stratified analysis when examining the role of TNF in the brain.
Collapse
Affiliation(s)
- Leda Mygind
- Institute of Molecular Medicine, Department of Neurobiology, University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense C, Denmark; (L.M.); (V.T.); (R.V.); (K.L.L.)
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, DK-5000 Odense C, Denmark
| | - Marianne Skov-Skov Bergh
- Department of Forensic Sciences, Division of Laboratory Medicine, Oslo University Hospital, Loviseberggata, 60456 Oslo, Norway;
| | - Vivien Tejsi
- Institute of Molecular Medicine, Department of Neurobiology, University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense C, Denmark; (L.M.); (V.T.); (R.V.); (K.L.L.)
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, DK-5000 Odense C, Denmark
| | - Ramanan Vaitheeswaran
- Institute of Molecular Medicine, Department of Neurobiology, University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense C, Denmark; (L.M.); (V.T.); (R.V.); (K.L.L.)
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, DK-5000 Odense C, Denmark
| | - Kate L. Lambertsen
- Institute of Molecular Medicine, Department of Neurobiology, University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense C, Denmark; (L.M.); (V.T.); (R.V.); (K.L.L.)
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, DK-5000 Odense C, Denmark
- Department of Neurology, Odense University Hospital, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark
| | - Bente Finsen
- Institute of Molecular Medicine, Department of Neurobiology, University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense C, Denmark; (L.M.); (V.T.); (R.V.); (K.L.L.)
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, DK-5000 Odense C, Denmark
| | - Athanasios Metaxas
- Institute of Molecular Medicine, Department of Neurobiology, University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense C, Denmark; (L.M.); (V.T.); (R.V.); (K.L.L.)
- BRIDGE—Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsløws Vej 19, DK-5000 Odense C, Denmark
- School of Science, Department of Life Sciences, European University Cyprus, 6 Diogenis Str., Nicosia 1516, Cyprus
| |
Collapse
|
28
|
Hermann DM, Gunzer M. Modulating Microglial Cells for Promoting Brain Recovery and Repair. Front Cell Neurosci 2021; 14:627987. [PMID: 33505251 PMCID: PMC7829249 DOI: 10.3389/fncel.2020.627987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/07/2020] [Indexed: 11/22/2022] Open
Abstract
Representing the brain’s innate immune cells that interact vividly with blood-derived immune cells and brain parenchymal cells, microglia set the stage for successful brain remodeling and repair in the aftermath of brain damage. With the development of pharmacological colony-stimulating factor-1 receptor inhibitors, which allow inhibiting or depleting microglial cells, and of transgenic mice, allowing the inducible depletion of microglial cells, experimental tools have become available for studying roles of microglia in neurodegenerative and neurorestorative processes. These models open fundamental insights into roles of microglia in controlling synaptic plasticity in the healthy and the injured brain. Acting as a switch from injury to repair, microglial cells might open opportunities for promoting neurological recovery in human patients upon brain injury.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Matthias Gunzer
- Institute of Experimental Immunology and Imaging, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| |
Collapse
|
29
|
Role of Microglia in Modulating Adult Neurogenesis in Health and Neurodegeneration. Int J Mol Sci 2020; 21:ijms21186875. [PMID: 32961703 PMCID: PMC7555074 DOI: 10.3390/ijms21186875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
Microglia are the resident immune cells of the brain, constituting the powerhouse of brain innate immunity. They originate from hematopoietic precursors that infiltrate the developing brain during different stages of embryogenesis, acquiring a phenotype characterized by the presence of dense ramifications. Microglial cells play key roles in maintaining brain homeostasis and regulating brain immune responses. They continuously scan and sense the brain environment to detect any occurring changes. Upon detection of a signal related to physiological or pathological processes, the cells are activated and transform to an amoeboid-like phenotype, mounting adequate responses that range from phagocytosis to secretion of inflammatory and trophic factors. The overwhelming evidence suggests that microglia are crucially implicated in influencing neuronal proliferation and differentiation, as well as synaptic connections, and thereby cognitive and behavioral functions. Here, we review the role of microglia in adult neurogenesis under physiological conditions, and how this role is affected in neurodegenerative diseases.
Collapse
|
30
|
Gray SC, Kinghorn KJ, Woodling NS. Shifting equilibriums in Alzheimer's disease: the complex roles of microglia in neuroinflammation, neuronal survival and neurogenesis. Neural Regen Res 2020; 15:1208-1219. [PMID: 31960800 PMCID: PMC7047786 DOI: 10.4103/1673-5374.272571] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/02/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease is the leading cause of dementia. Its increased prevalence in developed countries, due to the sharp rise in ageing populations, presents one of the costliest challenges to modern medicine. In order to find disease-modifying therapies to confront this challenge, a more complete understanding of the pathogenesis of Alzheimer's disease is necessary. Recent studies have revealed increasing evidence for the roles played by microglia, the resident innate immune system cells of the brain. Reflecting the well-established roles of microglia in reacting to pathogens and inflammatory stimuli, there is now a growing literature describing both protective and detrimental effects for individual cytokines and chemokines produced by microglia in Alzheimer's disease. A smaller but increasing number of studies have also addressed the divergent roles played by microglial neurotrophic and neurogenic factors, and how their perturbation may play a key role in the pathogenesis of Alzheimer's disease. Here we review recent findings on the roles played by microglia in neuroinflammation, neuronal survival and neurogenesis in Alzheimer's disease. In each case, landmark studies have provided evidence for the divergent ways in which microglia can either promote neuronal function and survival, or perturb neuronal function, leading to cell death. In many cases, the secreted molecules of microglia can lead to divergent effects depending on the magnitude and context of microglial activation. This suggests that microglial functions must be maintained in a fine equilibrium, in order to support healthy neuronal function, and that the cellular microenvironment in the Alzheimer's disease brain disrupts this fine balance, leading to neurodegeneration. Thus, an understanding of microglial homeostasis, both in health and across the trajectory of the disease state, will improve our understanding of the pathogenic mechanisms underlying Alzheimer's disease, and will hopefully lead to the development of microglial-based therapeutic strategies to restore equilibrium in the Alzheimer's disease brain.
Collapse
Affiliation(s)
- Sophie C. Gray
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J. Kinghorn
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Nathaniel S. Woodling
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
| |
Collapse
|
31
|
Benmamar-Badel A, Owens T, Wlodarczyk A. Protective Microglial Subset in Development, Aging, and Disease: Lessons From Transcriptomic Studies. Front Immunol 2020; 11:430. [PMID: 32318054 PMCID: PMC7147523 DOI: 10.3389/fimmu.2020.00430] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/25/2020] [Indexed: 12/25/2022] Open
Abstract
Microglial heterogeneity has been the topic of much discussion in the scientific community. Elucidation of their plasticity and adaptability to disease states triggered early efforts to characterize microglial subsets. Over time, their phenotypes, and later on their homeostatic signature, were revealed, through the use of increasingly advanced transcriptomic techniques. Recently, an increasing number of these "microglial signatures" have been reported in various homeostatic and disease contexts. Remarkably, many of these states show similar overlapping microglial gene expression patterns, both in homeostasis and in disease or injury. In this review, we integrate information from these studies, and we propose a unique subset, for which we introduce a core signature, based on our own research and reports from the literature. We describe that this subset is found in development and in normal aging as well as in diverse diseases. We discuss the functions of this subset as well as how it is induced.
Collapse
Affiliation(s)
- Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
- Department of Neurology, Slagelse Hospital, Institute of Regional Health Research, Slagelse, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| |
Collapse
|