1
|
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, Edison P, Ramirez A, Cruchaga C, Lambert JC, Laza AR, Sanchez-Mut JV, Fischer A, Castro-Gomez S, Stein TD, Kleineidam L, Wagner M, Neher JJ, Cunningham C, Singhrao SK, Prinz M, Glass CK, Schlachetzki JCM, Butovsky O, Kleemann K, De Jaeger PL, Scheiblich H, Brown GC, Landreth G, Moutinho M, Grutzendler J, Gomez-Nicola D, McManus RM, Andreasson K, Ising C, Karabag D, Baker DJ, Liddelow SA, Verkhratsky A, Tansey M, Monsonego A, Aigner L, Dorothée G, Nave KA, Simons M, Constantin G, Rosenzweig N, Pascual A, Petzold GC, Kipnis J, Venegas C, Colonna M, Walter J, Tenner AJ, O'Banion MK, Steinert JR, Feinstein DL, Sastre M, Bhaskar K, Hong S, Schafer DP, Golde T, Ransohoff RM, Morgan D, Breitner J, Mancuso R, Riechers SP. Neuroinflammation in Alzheimer disease. Nat Rev Immunol 2024:10.1038/s41577-024-01104-7. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/20/2025]
Abstract
Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jeroen Hoozemanns
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charlotte Teunissen
- Department of Laboratory Medicine, VUMC Amsterdam, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Imperial College London, London, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Agustin Ruiz Laza
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jose Vicente Sanchez-Mut
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Alicante, Spain
| | - Andre Fischer
- Clinic for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Disease (DZNE), Göttingen, Germany
| | - Sergio Castro-Gomez
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Michael Wagner
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center Munich, Biochemistry, Medical Faculty, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Sim K Singhrao
- Brain and Behaviour Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip L De Jaeger
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Hannah Scheiblich
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Guy C Brown
- Deparment of Biochemistry, University of Cambridge, Cambridge, UK
| | - Gary Landreth
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Miguel Moutinho
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Ising
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Karabag
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Darren J Baker
- Department of Paediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York City, NY, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Malu Tansey
- College of Medicine, University of Florida, Gainsville, FL, USA
| | - Alon Monsonego
- Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Hôpital Saint-Antoine, Paris, France
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University of Bonn, Bonn, Germany
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto Biosanitario de Granada (ibs.Granada), Granada, Spain
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jochen Walter
- Center of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Douglas L Feinstein
- Department of NeuroAnesthesia, University of Illinois at Chicago, Chicago, IL, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kiran Bhaskar
- Department of Molecular Genetics & Microbiology and Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Todd Golde
- Department of Pharmacology and Chemical Biology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sean-Patrick Riechers
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
| |
Collapse
|
2
|
Ji E, Zhang Y, Li Z, Wei L, Wu Z, Li Y, Yu X, Song TJ. The Chemokine CCL2 Promotes Excitatory Synaptic Transmission in Hippocampal Neurons via GluA1 Subunit Trafficking. Neurosci Bull 2024; 40:1649-1666. [PMID: 38954270 PMCID: PMC11607194 DOI: 10.1007/s12264-024-01236-9] [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: 08/10/2023] [Accepted: 03/08/2024] [Indexed: 07/04/2024] Open
Abstract
The CC chemokine ligand 2 (CCL2, also known as MCP-1) and its cognate receptor CCR2 have well-characterized roles in chemotaxis. CCL2 has been previously shown to promote excitatory synaptic transmission and neuronal excitability. However, the detailed molecular mechanism underlying this process remains largely unclear. In cultured hippocampal neurons, CCL2 application rapidly upregulated surface expression of GluA1, in a CCR2-dependent manner, assayed using SEP-GluA1 live imaging, surface GluA1 antibody staining, and electrophysiology. Using pharmacology and reporter assays, we further showed that CCL2 upregulated surface GluA1 expression primarily via Gαq- and CaMKII-dependent signaling. Consistently, using i.p. injection of lipopolysaccharide to induce neuroinflammation, we found upregulated phosphorylation of S831 and S845 sites on AMPA receptor subunit GluA1 in the hippocampus, an effect blocked in Ccr2-/- mice. Together, these results provide a mechanism through which CCL2, and other secreted molecules that signal through G-protein coupled receptors, can directly regulate synaptic transmission.
Collapse
Affiliation(s)
- En Ji
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yuanyuan Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhiqiang Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Lai Wei
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xiang Yu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| | - Tian-Jia Song
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
- Shandong Provincial Key Medical and Health Laboratory of Psychiatric Genetics of Shandong Mental Health Center, Shandong University, Jinan, 250014, China.
| |
Collapse
|
3
|
Woods C, Wang G, Milner TA, Glass MJ. Tumor necrosis factor alpha induces NOX2-dependent reactive oxygen species production in hypothalamic paraventricular nucleus neurons following angiotensin II infusion. Neurochem Int 2024; 179:105825. [PMID: 39097233 DOI: 10.1016/j.neuint.2024.105825] [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: 12/21/2023] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
There is evidence that tumor necrosis factor alpha (TNFα) influences autonomic processes coordinated within the hypothalamic paraventricular nucleus (PVN), however, the signaling mechanisms subserving TNFα's actions in this brain area are unclear. In non-neuronal cell types, TNFα has been shown to play an important role in canonical NADPH oxidase (NOX2)-mediated production of reactive oxygen species (ROS), molecules also known to be critically involved in hypertension. However, little is known about the role of TNFα in NOX2-dependent ROS production in the PVN within the context of hypertension. Using dual labeling immunoelectron microscopy and dihydroethidium (DHE) microfluorography, we provide structural and functional evidence for interactions between TNFα and NOX2 in the PVN. The TNFα type 1 receptor (TNFR1), the major mediator of TNFα signaling in the PVN, was commonly co-localized with the catalytic gp91phox subunit of NOX2 in postsynaptic sites of PVN neurons. Additionally, there was an increase in dual labeled dendritic profiles following fourteen-day slow-pressor angiotensin II (AngII) infusion. Using DHE microfluorography, it was also shown that TNFα application resulted in a NOX2-dependent increase in ROS in isolated PVN neurons projecting to the spinal cord. Further, TNFα-mediated ROS production was heightened after AngII infusion. The finding that TNFR1 and gp91phox are positioned for rapid interactions, particularly in PVN-spinal cord projection neurons, provides a molecular substrate by which inflammatory signaling and oxidative stress may jointly contribute to AngII hypertension.
Collapse
Affiliation(s)
- Clara Woods
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA; Harold and Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Ave, New York, NY, 10065, USA
| | - Michael J Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
4
|
Crews FT, Coleman LG, Macht VA, Vetreno RP. Alcohol, HMGB1, and Innate Immune Signaling in the Brain. Alcohol Res 2024; 44:04. [PMID: 39135668 PMCID: PMC11318841 DOI: 10.35946/arcr.v44.1.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Binge drinking (i.e., consuming enough alcohol to achieve a blood ethanol concentration of 80 mg/dL, approximately 4-5 drinks within 2 hours), particularly in early adolescence, can promote progressive increases in alcohol drinking and alcohol-related problems that develop into compulsive use in the chronic relapsing disease, alcohol use disorder (AUD). Over the past decade, neuroimmune signaling has been discovered to contribute to alcohol-induced changes in drinking, mood, and neurodegeneration. This review presents a mechanistic hypothesis supporting high mobility group box protein 1 (HMGB1) and Toll-like receptor (TLR) signaling as key elements of alcohol-induced neuroimmune signaling across glia and neurons, which shifts gene transcription and synapses, altering neuronal networks that contribute to the development of AUD. This hypothesis may help guide further research on prevention and treatment. SEARCH METHODS The authors used the search terms "HMGB1 protein," "alcohol," and "brain" across PubMed, Scopus, and Embase to find articles published between 1991 and 2023. SEARCH RESULTS The database search found 54 references in PubMed, 47 in Scopus, and 105 in Embase. A total of about 100 articles were included. DISCUSSION AND CONCLUSIONS In the brain, immune signaling molecules play a role in normal development that differs from their functions in inflammation and the immune response, although cellular receptors and signaling are shared. In adults, pro-inflammatory signals have emerged as contributing to brain adaptation in stress, depression, AUD, and neurodegenerative diseases. HMGB1, a cytokine-like signaling protein released from activated cells, including neurons, is hypothesized to activate pro-inflammatory signals through TLRs that contribute to adaptations to binge and chronic heavy drinking. HMGB1 alone and in heteromers with other molecules activates TLRs and other immune receptors that spread signaling across neurons and glia. Both blood and brain levels of HMGB1 increase with ethanol exposure. In rats, an adolescent intermittent ethanol (AIE) binge drinking model persistently increases brain HMGB1 and its receptors; alters microglia, forebrain cholinergic neurons, and neuronal networks; and increases alcohol drinking and anxiety while disrupting cognition. Studies of human postmortem AUD brain have found elevated levels of HMGB1 and TLRs. These signals reduce cholinergic neurons, whereas microglia, the brain's immune cells, are activated by binge drinking. Microglia regulate synapses through complement proteins that can change networks affected by AIE that increase drinking, contributing to risks for AUD. Anti-inflammatory drugs, exercise, cholinesterase inhibitors, and histone deacetylase epigenetic inhibitors prevent and reverse the AIE-induced pathology. Further, HMGB1 antagonists and other anti-inflammatory treatments may provide new therapies for alcohol misuse and AUD. Collectively, these findings suggest that restoring the innate immune signaling balance is central to recovering from alcohol-related pathology.
Collapse
Affiliation(s)
- Fulton T. Crews
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Leon G. Coleman
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Victoria A. Macht
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
5
|
Hosseini Z, Beheshti F, Hosseini Kakhki FS, Hosseini M, Anaeigoudari A. Sodium nitroprusside restored lipopolysaccharide-induced learning and memory impairment in male rats via attenuating inflammation and oxidative stress. Physiol Rep 2024; 12:e16053. [PMID: 38806440 PMCID: PMC11133007 DOI: 10.14814/phy2.16053] [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: 03/04/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Inflammation and oxidative stress upset memory. We explored influence of sodium nitroprusside (SNP) on memory deficits resulted from lipopolysaccharide (LPS).Groups include control, LPS, LPS + SNP 1 mg/kg, LPS + SNP 2 mg/kg, and LPS + SNP 3 mg/kg. Morris water maze and passive avoidance tests and biochemical measurements were carried out.In Morris water maze, LPS prolonged time and distance for finding the platform. In probe trial, it diminished time spent and traveled distance in the target zone. Injection of 2 and 3 mg/kg of SNP overturned the effect of LPS. In passive avoidance task, LPS postponed entrance into darkroom and reduced time spent in light room and incremented time spent in darkroom in 3, 24, and 72 h after electrical shock. All three doses of SNP restored the effects of LPS. Biochemical experiments confirmed that LPS elevated interleukin-6 and malondialdehyde concentration and declined total thiol content and superoxide dismutase and catalase activity in the hippocampus and cortex tissues. SNP particularly at a 3 mg/kg dose ameliorated LPS effects on these parameters.SNP attenuated memory disabilities resulting from LPS through modifying inflammation and boosting antioxidant defense.
Collapse
Affiliation(s)
- Zeinab Hosseini
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| | - Farimah Beheshti
- Neuroscience Research CenterTorbat Heydariyeh University of Medical SciencesTorbat HeydariyehIran
- Department of Physiology, School of Paramedical SciencesTorbat Heydariyeh University of Medical SciencesTorbat HeydariyehIran
| | | | - Mahmoud Hosseini
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
- Psychiatry and Behavioral Sciences Research CenterMashhad University of Medical SciencesMashhadIran
| | - Akbar Anaeigoudari
- Department of Physiology, School of MedicineJiroft University of Medical SciencesJiroftIran
| |
Collapse
|
6
|
Ansari MA, Tripathi T, Venkidasamy B, Monziani A, Rajakumar G, Alomary MN, Alyahya SA, Onimus O, D'souza N, Barkat MA, Al-Suhaimi EA, Samynathan R, Thiruvengadam M. Multifunctional Nanocarriers for Alzheimer's Disease: Befriending the Barriers. Mol Neurobiol 2024; 61:3042-3089. [PMID: 37966683 DOI: 10.1007/s12035-023-03730-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Neurodegenerative diseases (NDDs) have been increasing in incidence in recent years and are now widespread worldwide. Neuronal death is defined as the progressive loss of neuronal structure or function which is closely associated with NDDs and represents the intrinsic features of such disorders. Amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer's, Parkinson's, and Huntington's diseases (AD, PD, and HD, respectively) are considered neurodegenerative diseases that affect a large number of people worldwide. Despite the testing of various drugs, there is currently no available therapy that can remedy or effectively slow the progression of these diseases. Nanomedicine has the potential to revolutionize drug delivery for the management of NDDs. The use of nanoparticles (NPs) has recently been developed to improve drug delivery efficiency and is currently subjected to extensive studies. Nanoengineered particles, known as nanodrugs, can cross the blood-brain barrier while also being less invasive compared to the most treatment strategies in use. Polymeric, magnetic, carbonic, and inorganic NPs are examples of NPs that have been developed to improve drug delivery efficiency. Primary research studies using NPs to cure AD are promising, but thorough research is needed to introduce these approaches to clinical use. In the present review, we discussed the role of metal-based NPs, polymeric nanogels, nanocarrier systems such as liposomes, solid lipid NPs, polymeric NPs, exosomes, quantum dots, dendrimers, polymersomes, carbon nanotubes, and nanofibers and surfactant-based systems for the therapy of neurodegenerative diseases. In addition, we highlighted nanoformulations such as N-butyl cyanoacrylate, poly(butyl cyanoacrylate), D-penicillamine, citrate-coated peptide, magnetic iron oxide, chitosan (CS), lipoprotein, ceria, silica, metallic nanoparticles, cholinesterase inhibitors, an acetylcholinesterase inhibitors, metal chelators, anti-amyloid, protein, and peptide-loaded NPs for the treatment of AD.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Takshashila Tripathi
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Alan Monziani
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Govindasamy Rajakumar
- Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Sami A Alyahya
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Oriane Onimus
- Faculty of Basic and Biomedical Sciences, University of Paris, Paris, France
| | - Naomi D'souza
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Research Consultation Department, Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Ramkumar Samynathan
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
7
|
Kadian M, Saini N, Khera A, Kumar A. Neuroprotective mechanism of trans,trans-Farnesol in an ICV-STZ-induced rat model of Alzheimer's pathology. Inflammopharmacology 2024; 32:1545-1573. [PMID: 38308793 DOI: 10.1007/s10787-023-01413-8] [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: 04/02/2023] [Accepted: 12/13/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a prominent cause of dementia, resulting in neurodegeneration and memory impairment. This condition imposes a considerable public health burden on both patients and their families due to the patients' functional impairments as well as the psychological and financial constraints. It has been well demonstrated that its aetiology involves proteinopathy, mitochondriopathies, and enhanced reactive oxygen species (ROS) generation, which are some of the key features of AD brains that further result in oxidative stress, excitotoxicity, autophagy, and mitochondrial dysfunction. OBJECTIVE The current investigation was created with the aim of elucidating the neurological defence mechanism of trans,trans-Farnesol (TF) against intracerebroventricular-streptozotocin (ICV-STZ)-induced Alzheimer-like symptoms and related pathologies in rodents. MATERIALS AND METHODS The current investigation involved male SD rats receiving TF (25-100 mg/kg, per oral) consecutively for 21 days in ICV-STZ-treated animals. An in silico study was carried out to explore the possible interaction between TF and NADH dehydrogenase and succinate dehydrogenase. Further, various behavioural (Morris water maze and novel object recognition test), biochemical (oxidants and anti-oxidant markers), activities of mitochondrial enzyme complexes and acetylcholinesterase (AChE), pro-inflammatory (tumor necrosis factor-alpha; TNF-α) levels, and histopathological studies were evaluated in specific brain regions. RESULTS Rats administered ICV-STZ followed by treatment with TF (25, 50, and 100 mg/kg) for 21 days had significantly better mental performance (reduced escape latency to access platform, extended time spent in target quadrant, and improved differential index) in the Morris water maze test and new object recognition test models when compared to control (ICV-STZ)-treated groups. Further, TF treatment significantly restored redox proportion, anti-oxidant levels, regained mitochondrial capacities, attenuated altered AChE action, levels of TNF-α, and histopathological alterations in certain brain regions in comparison with control. In in silico analysis, TF caused greater interaction with NADH dehydrogenase and succinate dehydrogenase. CONCLUSION The current work demonstrates the neuroprotective ability of TF in an experimental model with AD-like pathologies. The study further suggests that the neuroprotective impacts of TF may be related to its effects on TNF-α levels, oxidative stress pathways, and mitochondrial complex capabilities.
Collapse
Affiliation(s)
- Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Neetu Saini
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Alka Khera
- Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
8
|
Pinto MJ, Ragozzino D, Bessis A, Audinat E. Microglial Modulation of Synaptic Maturation, Activity, and Plasticity. ADVANCES IN NEUROBIOLOGY 2024; 37:209-219. [PMID: 39207694 DOI: 10.1007/978-3-031-55529-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, which are the resident immune cells of the CNS, also have important functions in physiological conditions. In this chapter, we review the experimental evidence that microglia modulate neuronal and synaptic activity during normal development and in adults. We show that microglia can regulate the maturation and function of both inhibitory and excitatory synapses that can be stimulated or repressed. We further review the fact that these regulations occur in various brain regions, through soluble and membrane molecules, directly or through other cell partners. This review emphasizes the fact that microglia are genuine and highly context-dependent and thus adaptable regulators of neuronal activity.
Collapse
Affiliation(s)
- Maria Joana Pinto
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
- Center for Neuroscience and Cell Biology (CNC), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - Alain Bessis
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Etienne Audinat
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
9
|
Dvorak NM, Domingo ND, Tapia CM, Wadsworth PA, Marosi M, Avchalumov Y, Fongsaran C, Koff L, Di Re J, Sampson CM, Baumgartner TJ, Wang P, Villarreal PP, Solomon OD, Stutz SJ, Aditi, Porter J, Gbedande K, Prideaux B, Green TA, Seeley EH, Samir P, Dineley KT, Vargas G, Zhou J, Cisneros I, Stephens R, Laezza F. TNFR1 signaling converging on FGF14 controls neuronal hyperactivity and sickness behavior in experimental cerebral malaria. J Neuroinflammation 2023; 20:306. [PMID: 38115011 PMCID: PMC10729485 DOI: 10.1186/s12974-023-02992-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Excess tumor necrosis factor (TNF) is implicated in the pathogenesis of hyperinflammatory experimental cerebral malaria (eCM), including gliosis, increased levels of fibrin(ogen) in the brain, behavioral changes, and mortality. However, the role of TNF in eCM within the brain parenchyma, particularly directly on neurons, remains underdefined. Here, we investigate electrophysiological consequences of eCM on neuronal excitability and cell signaling mechanisms that contribute to observed phenotypes. METHODS The split-luciferase complementation assay (LCA) was used to investigate cell signaling mechanisms downstream of tumor necrosis factor receptor 1 (TNFR1) that could contribute to changes in neuronal excitability in eCM. Whole-cell patch-clamp electrophysiology was performed in brain slices from eCM mice to elucidate consequences of infection on CA1 pyramidal neuron excitability and cell signaling mechanisms that contribute to observed phenotypes. Involvement of identified signaling molecules in mediating behavioral changes and sickness behavior observed in eCM were investigated in vivo using genetic silencing. RESULTS Exploring signaling mechanisms that underlie TNF-induced effects on neuronal excitability, we found that the complex assembly of fibroblast growth factor 14 (FGF14) and the voltage-gated Na+ (Nav) channel 1.6 (Nav1.6) is increased upon tumor necrosis factor receptor 1 (TNFR1) stimulation via Janus Kinase 2 (JAK2). On account of the dependency of hyperinflammatory experimental cerebral malaria (eCM) on TNF, we performed patch-clamp studies in slices from eCM mice and showed that Plasmodium chabaudi infection augments Nav1.6 channel conductance of CA1 pyramidal neurons through the TNFR1-JAK2-FGF14-Nav1.6 signaling network, which leads to hyperexcitability. Hyperexcitability of CA1 pyramidal neurons caused by infection was mitigated via an anti-TNF antibody and genetic silencing of FGF14 in CA1. Furthermore, knockdown of FGF14 in CA1 reduced sickness behavior caused by infection. CONCLUSIONS FGF14 may represent a therapeutic target for mitigating consequences of TNF-mediated neuroinflammation.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Nadia D Domingo
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Cynthia M Tapia
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Paul A Wadsworth
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Mate Marosi
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yosef Avchalumov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Chanida Fongsaran
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Leandra Koff
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jessica Di Re
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Catherine M Sampson
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Timothy J Baumgartner
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Pingyuan Wang
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Paula P Villarreal
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Clinical Sciences Program, The Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Olivia D Solomon
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sonja J Stutz
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Aditi
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jacob Porter
- Department of Chemistry, University of Texas, Austin, TX, 78712, USA
| | - Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07301, USA
| | - Brendan Prideaux
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Erin H Seeley
- Department of Chemistry, University of Texas, Austin, TX, 78712, USA
| | - Parimal Samir
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kelley T Dineley
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Gracie Vargas
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jia Zhou
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Irma Cisneros
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07301, USA.
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
10
|
Bellingacci L, Canonichesi J, Mancini A, Parnetti L, Di Filippo M. Cytokines, synaptic plasticity and network dynamics: a matter of balance. Neural Regen Res 2023; 18:2569-2572. [PMID: 37449591 DOI: 10.4103/1673-5374.371344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
The modern view of the immune system as a sensitizing and modulating machinery of the central nervous system is now well recognized. However, the specific mechanisms underlying this fine crosstalk have yet to be fully disentangled. To control cognitive function and behavior, the two systems are engaged in a subtle interacting act. In this scenario, a dual action of pro-inflammatory cytokines in the modulation of brain network connections is emerging. Pro-inflammatory cytokines are indeed required to express physiological plasticity in the hippocampal network while being detrimental when over-expressed during uncontrolled inflammatory processes. In this dynamic equilibrium, synaptic functioning and the performance of neural networks are ensured by maintaining an appropriate balance between pro- and anti-inflammatory molecules in the central nervous system microenvironment.
Collapse
Affiliation(s)
- Laura Bellingacci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jacopo Canonichesi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
11
|
Wang J, Qiu F, Zhang Z, Liu Y, Zhou Q, Dai S, Xiang S, Wei C. Clostridium butyricum Alleviates DEHP Plasticizer-Induced Learning and Memory Impairment in Mice via Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18524-18537. [PMID: 37963287 DOI: 10.1021/acs.jafc.3c03533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) plasticizer, a well-known environmental and food pollutant, has neurotoxicity. However, it is unknown whether DEHP leads to learning and memory impairment through gut-brain axis and whether Clostridium butyricum can alleviate this impairment. Here, C57BL/6 mice were exposed to DEHP and treated with C. butyricum. Learning and memory abilities were evaluated through the Morris water maze. The levels of synaptic proteins, inflammatory cytokines, and 5-hydroxytryptamine (5-HT) were detected by immunohistochemistry or ELISA. Gut microbiota were analyzed through 16S rRNA sequencing. C. butyricum alleviated DEHP-induced learning and memory impairment and restored synaptic proteins. It significantly relieved DEHP-induced inflammation and recovered 5-HT levels. C. butyricum recovered the richness of the gut microbiota decreased by DEHP, with the Bifidobacterium genus increasing the most. Overall, C. butyricum alleviated DEHP-induced learning and memory impairment due to reduced inflammation and increased 5-HT secretion, which was partly attributed to the recovery of gut microbiota.
Collapse
Affiliation(s)
- Jin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Feng Qiu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Zilong Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Yu Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Qian Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Siyu Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| |
Collapse
|
12
|
Alipour V, Shojaei A, Rezaei M, Mirnajafi-Zadeh J, Azizi H. Intergenerational consequences of adolescent morphine exposure on learning and memory. Neurosci Lett 2023; 808:137303. [PMID: 37196975 DOI: 10.1016/j.neulet.2023.137303] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/01/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Drug addiction is a worldwide social and medical disorder. More than 50 percent of drug abusers start their substance abuse in adolescence between the ages of 15-19. Adolescence is a sensitive and crucial period for the development and maturity of the brain. Chronic exposure to morphine, particularly during this period, lead to long-lasting effects, including effects that extend to the next generation. The current study examined the intergenerational effects of paternal morphine exposure during adolescence on learning and memory. In this study, male Wistar rats were exposed to increasing doses of morphine (5-25 mg/kg, s.c.) or saline for 10 days at postnatal days (PND) 30-39 during adolescence. Following a 20-day drug-free period, the treated male rats were mated with naïve females. Adult male offspring (PND 60-80) were tested for working memory, novel object recognition memory, spatial memory, and passive avoidance memory using the Y-Maze, novel object recognition, Morris water maze, and shuttle box tests, respectively. The spontaneous alternation (as measured in the Y-Maze test) was significantly less in the morphine-sired group compared to the saline-sired one. The offspring showed significantly less discrimination index in the novel object recognition test when compared to the control group. Morphine-sired offspring tended to spend significantly more time in the target quadrant and less escape latency in the Morris water maze on probe day when compared to the saline-sired ones. The offspring showed significantly less step-through latency to enter the dark compartment compared to the control group when measured in the shuttle box test. Paternal exposure to morphine during adolescence impaired working, novel object recognition, and passive avoidance memory in male offspring. Spatial memory changed in the morphine-sired group compared to the saline-sired one.
Collapse
Affiliation(s)
- Vida Alipour
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Shojaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
13
|
Gruol DL, Calderon D, French K, Melkonian C, Huitron-Resendiz S, Cates-Gatto C, Roberts AJ. Neuroimmune interactions with binge alcohol drinking in the cerebellum of IL-6 transgenic mice. Neuropharmacology 2023; 228:109455. [PMID: 36775097 PMCID: PMC10029700 DOI: 10.1016/j.neuropharm.2023.109455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
The neuroimmune system of the brain, which is comprised primarily of astrocytes and microglia, regulates a variety of homeostatic mechanisms that underlie normal brain function. Numerous conditions, including alcohol consumption, can disrupt this regulatory process by altering brain levels of neuroimmune factors. Alcohol and neuroimmune factors, such as proinflammatory cytokines IL-6 and TNF-alpha, act at similar targets in the brain, including excitatory and inhibitory synaptic transmission. Thus, alcohol-induced production of IL-6 and/or TNF-alpha could be important contributing factors to the effects of alcohol on the brain. Recent studies indicate that IL-6 plays a role in alcohol drinking and the effects of alcohol on the brain activity following the cessation of alcohol consumption (post-alcohol period), however information on these topics is limited. Here we used homozygous and heterozygous female and male transgenic mice with increased astrocyte expression of IL-6 to examined further the interactions between alcohol and IL-6 with respect to voluntary alcohol drinking, brain activity during the post-alcohol period, IL-6 signal transduction, and expression of synaptic proteins. Wildtype littermates (WT) served as controls. The transgenic mice model brain neuroimmune status with respect to IL-6 in subjects with a history of persistent alcohol use. Results showed a genotype dependent reduction in voluntary alcohol consumption in the Drinking in the Dark protocol and in frequency-dependent relationships between brain activity in EEG recordings during the post-alcohol period and alcohol consumption. IL-6, TNF-alpha, IL-6 signal transduction partners pSTAT3 and c/EBP beta, and synaptic proteins were shown to play a role in these genotypic effects.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Delilah Calderon
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Katharine French
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Claudia Melkonian
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | - Chelsea Cates-Gatto
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
14
|
Akıncı B, Oğul ÖE, Hanoğlu L, Kulaç B, Ören D, Ulu O, Basançelebi B. Evaluation of cognitive functions in adult individuals with COVID-19. Neurol Sci 2023; 44:793-802. [PMID: 36574178 PMCID: PMC9793347 DOI: 10.1007/s10072-022-06562-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE Cognitive deficits that are associated with coronavirus disease 2019 (COVID-19) and occur in the acute period are gaining importance. While most studies have focused on the elderly severely affected during acute infection, it remains unclear whether mild to moderate COVID-19 results in cognitive deficits in young patients. This study aims to evaluate the post-infection cognitive functions of young adults with mild to moderate symptoms of COVID-19. METHODS A total of 100 adults with similar age and educational background were included in the study. Half of those had been infected with COVID-19 in the last 60 days (N = 50), and the other half had not (N = 50). Global cognitive skills of the participants were evaluated through Montreal Cognitive Assessment Scale (MoCA) and Clock-Drawing Test; memory functions with Öktem Verbal Memory Processes Test (Ö-VMPT); attention span with Digit Span Test; executive functions with Fluency Tests, Stroop Test, and Trail Making Test; visual perceptual skills with Rey Osterrieth Complex Figure Test (ROCF); and neuropsychiatric status with Neuropsychiatric Inventory (NPI). Evaluations were performed in the experimental group for 21 to 60 days from the onset of the disease, and throughout the study, in the control group. RESULTS It was found that global cognitive skills, verbal memory, visual memory, executive function, and neuropsychiatric status were affected during COVID-19 (p < 0.05). CONCLUSION When the cases were analyzed according to disease severity, no relationship was found between cognitive deficits and disease severity.
Collapse
Affiliation(s)
- Büşra Akıncı
- Department of Physical Therapy and Rehabilitation, Medipol Mega University Hospital, Bagcilar, 34214, Istanbul, Turkey.
| | - Özden Erkan Oğul
- Department of Occupational Therapy, Faculty of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| | - Lütfü Hanoğlu
- Department of Neurology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Betül Kulaç
- Department of Physical Therapy and Rehabilitation, Medipol Mega University Hospital, Bagcilar, 34214 Istanbul, Turkey
| | - Damla Ören
- Department of Physical Therapy and Rehabilitation, Pendik Medipol University Hospital, Istanbul, Turkey
| | - Oğulcan Ulu
- Faculty of Science and Letters, Psychology, Beykent University, Istanbul, Turkey
| | - Berkan Basançelebi
- Department of Electroneurophysiology, Health Vocational School Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
15
|
De Felice E, Gonçalves de Andrade E, Golia MT, González Ibáñez F, Khakpour M, Di Castro MA, Garofalo S, Di Pietro E, Benatti C, Brunello N, Tascedda F, Kaminska B, Limatola C, Ragozzino D, Tremblay ME, Alboni S, Maggi L. Microglial diversity along the hippocampal longitudinal axis impacts synaptic plasticity in adult male mice under homeostatic conditions. J Neuroinflammation 2022; 19:292. [PMID: 36482444 PMCID: PMC9730634 DOI: 10.1186/s12974-022-02655-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.
Collapse
Affiliation(s)
- E. De Felice
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Gonçalves de Andrade
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. T. Golia
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - F. González Ibáñez
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - M. Khakpour
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. A. Di Castro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - S. Garofalo
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Di Pietro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - C. Benatti
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N. Brunello
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F. Tascedda
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - B. Kaminska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - C. Limatola
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - D. Ragozzino
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy ,grid.417778.a0000 0001 0692 3437Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - M. E. Tremblay
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - S. Alboni
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - L. Maggi
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
16
|
Zhang NY, Wang TH, Chou CH, Wu KC, Yang CR, Kung FL, Lin CJ. Ibuprofen treatment ameliorates memory deficits in rats with collagen-induced arthritis by normalizing aberrant MAPK/NF-κB and glutamatergic pathways. Eur J Pharmacol 2022; 933:175256. [PMID: 36088983 DOI: 10.1016/j.ejphar.2022.175256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 12/29/2022]
Abstract
Many studies have indicated that the risk of cognitive impairment is higher in patients with rheumatoid arthritis (RA). Additionally, patients with RA may have a lower incidence of cognitive impairment with long-term use of ibuprofen. This study was aimed at investigating the impacts of RA on memory function and the mechanisms that ibuprofen may exhibit to improve memory function in rats with collagen-induced arthritis (CIA). Ibuprofen (30 mg/kg) was given twice daily to CIA rats for two weeks starting from Day 18 following the first immunization. Memory function was measured by the Morris water maze (MWM) test and long-term potentiation (LTP). The proinflammatory cytokine levels and downstream signaling pathways, including mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB), were examined. Furthermore, the glutamatergic system, including glutamate transporters/receptors and brain extracellular levels of glutamate, was investigated. The results showed that the impaired learning memory in CIA rats, examined by the MWM test and LTP, can be ameliorated by ibuprofen treatment. Along with the improvement in memory deficits, ibuprofen attenuated both neuroinflammation and the associated elevated levels of phosphorylated p38, JNK, and p65 in the hippocampus of CIA rats. In addition, the decreased excitatory amino acid transporter 2 level, the increased extracellular glutamate, and the upregulated hippocampal NMDA receptor 2B of CIA rats were all normalized by ibuprofen treatment. These findings suggest that the effect of ibuprofen on the memory improvement in CIA rats is associated with the normalization of the activated MAPK and NF-κB pathways and the aberrant glutamatergic system.
Collapse
Affiliation(s)
- Nai-You Zhang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Hsuan Wang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsuan Chou
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Chen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fan-Lu Kung
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
17
|
Chen Y, Yang W, Chen F, Cui L. COVID-19 and cognitive impairment: neuroinvasive and blood‒brain barrier dysfunction. J Neuroinflammation 2022; 19:222. [PMID: 36071466 PMCID: PMC9450840 DOI: 10.1186/s12974-022-02579-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global pandemic. Although COVID-19 was initially described as a respiratory disease, there is growing evidence that SARS-CoV-2 is able to invade the brains of COVID-19 patients and cause cognitive impairment. It has been reported that SARS-CoV-2 may have invasive effects on a variety of cranial nerves, including the olfactory, trigeminal, optic, and vagus nerves, and may spread to other brain regions via infected nerve endings, retrograde transport, and transsynaptic transmission. In addition, the blood-brain barrier (BBB), composed of neurovascular units (NVUs) lining the brain microvasculature, acts as a physical barrier between nerve cells and circulating cells of the immune system and is able to regulate the transfer of substances between the blood and brain parenchyma. Therefore, the BBB may be an important structure for the direct and indirect interaction of SARS-CoV-2 with the brain via the blood circulation. In this review, we assessed the potential involvement of neuroinvasion under the SARS-CoV-2 infection, and the potential impact of BBB disorder under SARS-CoV-2 infection on cognitive impairment.
Collapse
Affiliation(s)
- Yanting Chen
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Wenren Yang
- Department of Trauma Center, Hengyang Medical School, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China
| | - Feng Chen
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Lili Cui
- Department of Neurology, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China.
| |
Collapse
|
18
|
Palmas MF, Etzi M, Pisanu A, Camoglio C, Sagheddu C, Santoni M, Manchinu MF, Pala M, Fusco G, De Simone A, Picci L, Mulas G, Spiga S, Scherma M, Fadda P, Pistis M, Simola N, Carboni E, Carta AR. The Intranigral Infusion of Human-Alpha Synuclein Oligomers Induces a Cognitive Impairment in Rats Associated with Changes in Neuronal Firing and Neuroinflammation in the Anterior Cingulate Cortex. Cells 2022; 11:cells11172628. [PMID: 36078036 PMCID: PMC9454687 DOI: 10.3390/cells11172628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is a complex pathology causing a plethora of non-motor symptoms besides classical motor impairments, including cognitive disturbances. Recent studies in the PD human brain have reported microgliosis in limbic and neocortical structures, suggesting a role for neuroinflammation in the development of cognitive decline. Yet, the mechanism underlying the cognitive pathology is under investigated, mainly for the lack of a valid preclinical neuropathological model reproducing the disease’s motor and non-motor aspects. Here, we show that the bilateral intracerebral infusion of pre-formed human alpha synuclein oligomers (H-αSynOs) within the substantia nigra pars compacta (SNpc) offers a valid model for studying the cognitive symptoms of PD, which adds to the classical motor aspects previously described in the same model. Indeed, H-αSynOs-infused rats displayed memory deficits in the two-trial recognition task in a Y maze and the novel object recognition (NOR) test performed three months after the oligomer infusion. In the anterior cingulate cortex (ACC) of H-αSynOs-infused rats the in vivo electrophysiological activity was altered and the expression of the neuron-specific immediate early gene (IEG) Npas4 (Neuronal PAS domain protein 4) and the AMPA receptor subunit GluR1 were decreased. The histological analysis of the brain of cognitively impaired rats showed a neuroinflammatory response in cognition-related regions such as the ACC and discrete subareas of the hippocampus, in the absence of any evident neuronal loss, supporting a role of neuroinflammation in cognitive decline. We found an increased GFAP reactivity and the acquisition of a proinflammatory phenotype by microglia, as indicated by the increased levels of microglial Tumor Necrosis Factor alpha (TNF-α) as compared to vehicle-infused rats. Moreover, diffused deposits of phospho-alpha synuclein (p-αSyn) and Lewy neurite-like aggregates were found in the SNpc and striatum, suggesting the spreading of toxic protein within anatomically interconnected areas. Altogether, we present a neuropathological rat model of PD that is relevant for the study of cognitive dysfunction featuring the disease. The intranigral infusion of toxic oligomeric species of alpha-synuclein (α-Syn) induced spreading and neuroinflammation in distant cognition-relevant regions, which may drive the altered neuronal activity underlying cognitive deficits.
Collapse
Affiliation(s)
| | - Michela Etzi
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Augusta Pisanu
- National Research Council, Institute of Neuroscience, 09040 Cagliari, Italy
| | - Chiara Camoglio
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Maria Francesca Manchinu
- Istituto Di Ricerca Genetica e Biomedica Del Consiglio Nazionale Delle Ricerche, 09040 Monserrato, Italy
| | - Mauro Pala
- Istituto Di Ricerca Genetica e Biomedica Del Consiglio Nazionale Delle Ricerche, 09040 Monserrato, Italy
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Alfonso De Simone
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Luca Picci
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Maria Scherma
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, 09040 Cagliari, Italy
- Correspondence:
| |
Collapse
|
19
|
Mezo-González CE, Daher Abdi A, Reyes-Castro LA, Olvera Hernández S, Almeida C, Croyal M, Aguesse A, Gavioli EC, Zambrano E, Bolaños-Jiménez F. Learning Deficits Induced by High-Calorie Feeding in the Rat are Associated With Impaired Brain Kynurenine Pathway Metabolism. Int J Tryptophan Res 2022; 15:11786469221111116. [PMID: 35846874 PMCID: PMC9277427 DOI: 10.1177/11786469221111116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
In addition to be a primary risk factor for type 2 diabetes and cardiovascular
disease, obesity is associated with learning disabilities. Here we examined
whether a dysregulation of the kynurenine pathway (KP) of tryptophan (Trp)
metabolism might underlie the learning deficits exhibited by obese individuals.
The KP is initiated by the enzymatic conversion of Trp into kynurenine (KYN) by
indoleamine 2,3-dioxygenase (IDO). KYN is further converted to several signaling
molecules including quinolinic acid (QA) which has a negative impact on
learning. Wistar rats were fed either standard chow or made obese by exposure to
a free choice high-fat high-sugar (fcHFHS) diet. Their learning capacities were
evaluated using a combination of the novel object recognition and the novel
object location tasks, and the concentrations of Trp and KYN-derived metabolites
in several brain regions determined by ultra-performance liquid
chromatography-tandem mass spectrometry. Male, but not female, obese rats
exhibited reduced learning capacity characterized by impaired encoding along
with increased hippocampal concentrations of QA, Xanthurenic acid (XA),
Nicotinamide (Nam), and oxidized Nicotinamide Adenine Dinucleotide (NAD+). In
contrast, no differences were detected in the serum levels of Trp or KP
metabolites. Moreover, obesity enhanced the expression in the hippocampus and
frontal cortex of kynurenine monooxygenase (KMO), an enzyme involved in the
production of QA from kynurenine. QA stimulates the glutamatergic system and its
increased production leads to cognitive impairment. These results suggest that
the deleterious effects of obesity on cognition are sex dependent and that
altered KP metabolism might contribute to obesity-associated learning
disabilities.
Collapse
Affiliation(s)
| | - Amran Daher Abdi
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Université de Nantes, Nantes France
| | - Luis Antonio Reyes-Castro
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Université de Nantes, Nantes France.,Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Sandra Olvera Hernández
- UMR Physiologie des Adaptations Nutritionnelles, INRAE - Université de Nantes, Nantes France.,Medical and Psychology School, Autonomous University of Baja California, Tijuana, Mexico
| | - Clarissa Almeida
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Mikaël Croyal
- CRNH-O Mass Spectrometry Core Facility, Nantes, France.,Université de Nantes, CNRS, INSERM, L'institut du Thorax, Nantes, France.,Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | | | - Elaine Cristina Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | |
Collapse
|
20
|
Baek SH, Kim H, Kim JW, Ryu S, Lee JY, Kim JM, Shin IS, Kim SW. Association between Peripheral Inflammatory Cytokines and Cognitive Function in Patients with First-Episode Schizophrenia. J Pers Med 2022; 12:jpm12071137. [PMID: 35887634 PMCID: PMC9317024 DOI: 10.3390/jpm12071137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022] Open
Abstract
In this study, we investigated the impact of inflammatory cytokines on the cognitive performance of patients with schizophrenia. The included patients met the criteria for schizophrenia spectrum disorder and were aged between 15 and 40 years, with a duration of illness ≤1 year. Plasma tumor necrosis factor (TNF)-α; interferon-γ; and interleukin (IL)-1β, IL-6, IL-8, IL-10, and IL-12 levels were measured. A computerized neurocognitive battery, measures for social cognitive function, and clinical measures were administered. A total of 174 patients with first-episode psychosis were enrolled. The TNF-α level was negatively correlated with scores on the digit span, verbal learning, and Wisconsin card sorting tests, and the number of correct responses on the continuous performance test (CR-CPT), whereas a positive correlation was detected with the trail making test (TMT)-B time. The interferon-γ level was negatively correlated with performance on the false belief and visual learning tests. The IL-1β level was positively correlated with the TMT-A time and CPT reaction time, whereas it was negatively correlated with the CR-CPT and performance on the visual learning and social cognitive tests. The IL-12 level was negatively correlated with the CR-CPT and false belief test. Our results suggest that proinflammatory cytokines are associated with cognitive impairment in patients with schizophrenia.
Collapse
Affiliation(s)
- Seon-Hwa Baek
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Honey Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Ju-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Seunghyong Ryu
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Ju-Yeon Lee
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Jae-Min Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Il-Seon Shin
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju 61419, Korea; (S.-H.B.); (H.K.); (J.-W.K.); (S.R.); (J.-Y.L.); (J.-M.K.); (I.-S.S.)
- Mindlink, Gwangju Bukgu Mental Health Center, Gwangju 61220, Korea
- Correspondence: ; Tel.: +82-62-220-6148
| |
Collapse
|
21
|
Neuroimmune Mechanisms Underlying Neuropathic Pain: The Potential Role of TNF-α-Necroptosis Pathway. Int J Mol Sci 2022; 23:ijms23137191. [PMID: 35806192 PMCID: PMC9266916 DOI: 10.3390/ijms23137191] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
The neuroimmune mechanism underlying neuropathic pain has been extensively studied. Tumor necrosis factor-alpha (TNF-α), a key pro-inflammatory cytokine that drives cytokine storm and stimulates a cascade of other cytokines in pain-related pathways, induces and modulates neuropathic pain by facilitating peripheral (primary afferents) and central (spinal cord) sensitization. Functionally, TNF-α controls the balance between cell survival and death by inducing an inflammatory response and two programmed cell death mechanisms (apoptosis and necroptosis). Necroptosis, a novel form of programmed cell death, is receiving increasing attraction and may trigger neuroinflammation to promote neuropathic pain. Chronic pain is often accompanied by adverse pain-associated emotional reactions and cognitive disorders. Overproduction of TNF-α in supraspinal structures such as the anterior cingulate cortex (ACC) and hippocampus plays an important role in pain-associated emotional disorders and memory deficits and also participates in the modulation of pain transduction. At present, studies reporting on the role of the TNF-α–necroptosis pathway in pain-related disorders are lacking. This review indicates the important research prospects of this pathway in pain modulation based on its role in anxiety, depression and memory deficits associated with other neurodegenerative diseases. In addition, we have summarized studies related to the underlying mechanisms of neuropathic pain mediated by TNF-α and discussed the role of the TNF-α–necroptosis pathway in detail, which may represent an avenue for future therapeutic intervention.
Collapse
|
22
|
Lyra E Silva NM, Barros-Aragão FGQ, De Felice FG, Ferreira ST. Inflammation at the crossroads of COVID-19, cognitive deficits and depression. Neuropharmacology 2022; 209:109023. [PMID: 35257690 PMCID: PMC8894741 DOI: 10.1016/j.neuropharm.2022.109023] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Natalia M Lyra E Silva
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada.
| | - Fernanda G Q Barros-Aragão
- D'OR Institute for Research & Education, RJ, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil.
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada; D'OR Institute for Research & Education, RJ, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil
| | - Sergio T Ferreira
- D'OR Institute for Research & Education, RJ, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, RJ, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ, Brazil
| |
Collapse
|
23
|
Pathway-specific TNF-mediated metaplasticity in hippocampal area CA1. Sci Rep 2022; 12:1746. [PMID: 35110639 PMCID: PMC8810872 DOI: 10.1038/s41598-022-05844-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/19/2022] [Indexed: 01/29/2023] Open
Abstract
Long-term potentiation (LTP) is regulated in part by metaplasticity, the activity-dependent alterations in neural state that coordinate the direction, amplitude, and persistence of future synaptic plasticity. Previously, we documented a heterodendritic metaplasticity effect whereby high-frequency priming stimulation in stratum oriens (SO) of hippocampal CA1 suppressed subsequent LTP in the stratum radiatum (SR). The cytokine tumor necrosis factor (TNF) mediated this heterodendritic metaplasticity in wild-type rodents and in a mouse model of Alzheimer’s disease. Here, we investigated whether LTP at other afferent synapses to CA1 pyramidal cells were similarly affected by priming stimulation. We found that priming stimulation in SO inhibited LTP only in SR and not in a second independent pathway in SO, nor in stratum lacunosum moleculare (SLM). Synapses in SR were also more sensitive than SO or SLM to the LTP-inhibiting effects of pharmacological TNF priming. Neither form of priming was sex-specific, while the metaplasticity effects were absent in TNFR1 knock-out mice. Our findings demonstrate an unexpected pathway specificity for the heterodendritic metaplasticity in CA1. That Schaffer collateral/commissural synapses in SR are particularly susceptible to such metaplasticity may reflect an important control of information processing in this pathway in addition to its sensitivity to neuroinflammation under disease conditions.
Collapse
|
24
|
Stranahan AM. Visceral adiposity, inflammation, and hippocampal function in obesity. Neuropharmacology 2021; 205:108920. [PMID: 34902347 DOI: 10.1016/j.neuropharm.2021.108920] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/09/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
The 'apple-shaped' anatomical pattern that accompanies visceral adiposity increases risk for multiple chronic diseases, including conditions that impact the brain, such as diabetes and hypertension. However, distinguishing between the consequences of visceral obesity, as opposed to visceral adiposity-associated metabolic and cardiovascular pathologies, presents certain challenges. This review summarizes current literature on relationships between adipose tissue distribution and cognition in preclinical models and highlights unanswered questions surrounding the potential role of tissue- and cell type-specific insulin resistance in these effects. While gaps in knowledge persist related to insulin insensitivity and cognitive impairment in obesity, several recent studies suggest that cells of the neurovascular unit contribute to hippocampal synaptic dysfunction, and this review interprets those findings in the context of progressive metabolic dysfunction in the CNS. Signalling between cerebrovascular endothelial cells, astrocytes, microglia, and neurons has been linked with memory deficits in visceral obesity, and this article describes the cellular changes in each of these populations with respect to their role in amplification or diminution of peripheral signals. The picture emerging from these studies, while incomplete, implicates pro-inflammatory cytokines, insulin resistance, and hyperglycemia in various stages of obesity-induced hippocampal dysfunction. As in the parable of the five blind wanderers holding different parts of an elephant, considerable work remains in order to assemble a model for the underlying mechanisms linking visceral adiposity with age-related cognitive decline.
Collapse
Affiliation(s)
- Alexis M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
25
|
Bettcher BM, Tansey MG, Dorothée G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease - a research prospectus. Nat Rev Neurol 2021; 17:689-701. [PMID: 34522039 PMCID: PMC8439173 DOI: 10.1038/s41582-021-00549-x] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Dysregulation of the immune system is a cardinal feature of Alzheimer disease (AD), and a considerable body of evidence indicates pathological alterations in central and peripheral immune responses that change over time. Considering AD as a systemic immune process raises important questions about how communication between the peripheral and central compartments occurs and whether this crosstalk represents a therapeutic target. We established a whitepaper workgroup to delineate the current status of the field and to outline a research prospectus for advancing our understanding of peripheral-central immune crosstalk in AD. To guide the prospectus, we begin with an overview of seminal clinical observations that suggest a role for peripheral immune dysregulation and peripheral-central immune communication in AD, followed by formative animal data that provide insights into possible mechanisms for these clinical findings. We then present a roadmap that defines important next steps needed to overcome conceptual and methodological challenges, opportunities for future interdisciplinary research, and suggestions for translating promising mechanistic studies into therapeutic interventions.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Behavioral Neurology Section, Department of Neurology, University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Malú G Tansey
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
| | - Guillaume Dorothée
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Team "Immune System and Neuroinflammation", Hôpital Saint-Antoine, Paris, France
| | - Michael T Heneka
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
26
|
Gupta A, Singh AK, Kumar R, Jamieson S, Pandey AK, Bishayee A. Neuroprotective Potential of Ellagic Acid: A Critical Review. Adv Nutr 2021; 12:1211-1238. [PMID: 33693510 PMCID: PMC8321875 DOI: 10.1093/advances/nmab007] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/02/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Ellagic acid (EA) is a dietary polyphenol present in various fruits, vegetables, herbs, and nuts. It exists either independently or as part of complex structures, such as ellagitannins, which release EA and several other metabolites including urolithins following absorption. During the past few decades, EA has drawn considerable attention because of its vast range of biological activities as well as its numerous molecular targets. Several studies have reported that the oxidative stress-lowering potential of EA accounts for its broad-spectrum pharmacological attributes. At the biochemical level, several mechanisms have also been associated with its therapeutic action, including its efficacy in normalizing lipid metabolism and lipidemic profile, regulating proinflammatory mediators, such as IL-6, IL-1β, and TNF-α, upregulating nuclear factor erythroid 2-related factor 2 and inhibiting NF-κB action. EA exerts appreciable neuroprotective activity by its free radical-scavenging action, iron chelation, initiation of several cell signaling pathways, and alleviation of mitochondrial dysfunction. Numerous in vivo studies have also explored the neuroprotective attribute of EA against various neurotoxins in animal models. Despite the increasing number of publications with experimental evidence, a critical analysis of available literature to understand the full neuroprotective potential of EA has not been performed. The present review provides up-to-date, comprehensive, and critical information regarding the natural sources of EA, its bioavailability, metabolism, neuroprotective activities, and underlying mechanisms of action in order to encourage further studies to define the clinical usefulness of EA for the management of neurological disorders.
Collapse
Affiliation(s)
- Ashutosh Gupta
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Ramesh Kumar
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Sarah Jamieson
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| | - Abhay Kumar Pandey
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| |
Collapse
|
27
|
Manrique-Suárez V, Macaya L, Contreras MA, Parra N, Maura R, González A, Toledo JR, Sánchez O. Design and characterization of a novel dimeric blood-brain barrier penetrating TNFα inhibitor. Proteins 2021; 89:1508-1521. [PMID: 34219271 DOI: 10.1002/prot.26173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022]
Abstract
Tumor necrosis factor-alpha (TNFα) inhibitors could prevent neurological disorders systemically, but their design generally relies on molecules unable to cross the blood-brain barrier (BBB). This research was aimed to design and characterize a novel TNFα inhibitor based on the angiopeptide-2 as a BBB shuttle molecule fused to the extracellular domain of human TNFα receptor 2 and a mutated vascular endothelial growth factor (VEGF) dimerization domain. This new chimeric protein (MTV) would be able to trigger receptor-mediated transcytosis across the BBB via low-density lipoprotein receptor-related protein-1 (LRP-1) and inhibit the cytotoxic effect of TNFα more efficiently because of its dimeric structure. Stably transformed CHO cells successfully expressed MTV, and its purification by Immobilized-Metal Affinity Chromatography (IMAC) rendered high purity degree. Mutated VEGF domain included in MTV did not show cell proliferation or angiogenic activities measured by scratch and aortic ring assays, which corroborate that the function of this domain is restricted to dimerization. The pairs MTV-TNFα (Kd 279 ± 40.9 nM) and MTV-LRP1 (Kd 399 ± 50.5 nM) showed high affinity by microscale thermophoresis, and a significant increase in cell survival was observed after blocking TNFα with MTV in a cell cytotoxicity assay. Also, the antibody staining in CHOK1 and bEnd3 cells demonstrated the adhesion of MTV to the LRP1 receptor located in the cell membrane. These results provide compelling evidence for the proper functioning of the three main domains of MTV individually, which encourage us to continue the research with this new molecule as a potential candidate for the systemic treatment of neurological disorders.
Collapse
Affiliation(s)
- Viana Manrique-Suárez
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Luis Macaya
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Maria Angélica Contreras
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Natalie Parra
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Rafael Maura
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Alaín González
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile.,Faculty of Basic Sciences, University of Medellin, Medellin, Colombia
| | - Jorge R Toledo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, School of Biological Science, Universidad de Concepción, Concepcion, Chile.,Center of Biotechnology and Biomedicine Spa, Concepción, Chile
| | - Oliberto Sánchez
- Recombinant Biopharmaceuticals Laboratory, Pharmacology Department, School of Biological Sciences, University of Concepcion, Concepcion, Chile.,Center of Biotechnology and Biomedicine Spa, Concepción, Chile
| |
Collapse
|
28
|
Awogbindin IO, Ben-Azu B, Olusola BA, Akinluyi ET, Adeniyi PA, Di Paolo T, Tremblay MÈ. Microglial Implications in SARS-CoV-2 Infection and COVID-19: Lessons From Viral RNA Neurotropism and Possible Relevance to Parkinson's Disease. Front Cell Neurosci 2021; 15:670298. [PMID: 34211370 PMCID: PMC8240959 DOI: 10.3389/fncel.2021.670298] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/05/2021] [Indexed: 12/24/2022] Open
Abstract
Since December 2019, humankind has been experiencing a ravaging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak, the second coronavirus pandemic in a decade after the Middle East respiratory syndrome coronavirus (MERS-CoV) disease in 2012. Infection with SARS-CoV-2 results in Coronavirus disease 2019 (COVID-19), which is responsible for over 3.1 million deaths worldwide. With the emergence of a second and a third wave of infection across the globe, and the rising record of multiple reinfections and relapses, SARS-CoV-2 infection shows no sign of abating. In addition, it is now evident that SARS-CoV-2 infection presents with neurological symptoms that include early hyposmia, ischemic stroke, meningitis, delirium and falls, even after viral clearance. This may suggest chronic or permanent changes to the neurons, glial cells, and/or brain vasculature in response to SARS-CoV-2 infection or COVID-19. Within the central nervous system (CNS), microglia act as the central housekeepers against altered homeostatic states, including during viral neurotropic infections. In this review, we highlight microglial responses to viral neuroinfections, especially those with a similar genetic composition and route of entry as SARS-CoV-2. As the primary sensor of viral infection in the CNS, we describe the pathogenic and neuroinvasive mechanisms of RNA viruses and SARS-CoV-2 vis-à-vis the microglial means of viral recognition. Responses of microglia which may culminate in viral clearance or immunopathology are also covered. Lastly, we further discuss the implication of SARS-CoV-2 CNS invasion on microglial plasticity and associated long-term neurodegeneration. As such, this review provides insight into some of the mechanisms by which microglia could contribute to the pathophysiology of post-COVID-19 neurological sequelae and disorders, including Parkinson's disease, which could be pervasive in the coming years given the growing numbers of infected and re-infected individuals globally.
Collapse
Affiliation(s)
- Ifeoluwa O. Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Nigeria
| | - Babatunde A. Olusola
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Elizabeth T. Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Philip A. Adeniyi
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Therese Di Paolo
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Sanguino‐Gómez J, Buurstede JC, Abiega O, Fitzsimons CP, Lucassen PJ, Eggen BJL, Lesuis SL, Meijer OC, Krugers HJ. An emerging role for microglia in stress‐effects on memory. Eur J Neurosci 2021; 55:2491-2518. [PMID: 33724565 PMCID: PMC9373920 DOI: 10.1111/ejn.15188] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/13/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Stressful experiences evoke, among others, a rapid increase in brain (nor)epinephrine (NE) levels and a slower increase in glucocorticoid hormones (GCs) in the brain. Microglia are key regulators of neuronal function and contain receptors for NE and GCs. These brain cells may therefore potentially be involved in modulating stress effects on neuronal function and learning and memory. In this review, we discuss that stress induces (1) an increase in microglial numbers as well as (2) a shift toward a pro‐inflammatory profile. These microglia have (3) impaired crosstalk with neurons and (4) disrupted glutamate signaling. Moreover, microglial immune responses after stress (5) alter the kynurenine pathway through metabolites that impair glutamatergic transmission. All these effects could be involved in the impairments in memory and in synaptic plasticity caused by (prolonged) stress, implicating microglia as a potential novel target in stress‐related memory impairments.
Collapse
Affiliation(s)
| | - Jacobus C. Buurstede
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Oihane Abiega
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems Section Molecular Neurobiology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
- Program in Neurosciences and Mental Health Hospital for Sick Children Toronto ON Canada
| | - Onno C. Meijer
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
30
|
Takahashi S, Fukushima H, Yu Z, Tomita H, Kida S. Tumor necrosis factor α negatively regulates the retrieval and reconsolidation of hippocampus-dependent memory. Brain Behav Immun 2021; 94:79-88. [PMID: 33677026 DOI: 10.1016/j.bbi.2021.02.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/28/2021] [Accepted: 02/28/2021] [Indexed: 01/05/2023] Open
Abstract
Neural inflammation is associated with cognitive decline, especially learning and memory. Tumor necrosis factor α (TNFα) is a major cytokine generated during neuroinflammation. Previous studies indicated that TNFα impairs hippocampus-dependent memory including contextual fear and spatial memories. However, it is unknown which memory processes are impaired by TNFα. Here, we show that TNFα blocked the retrieval and reconsolidation of contextual fear and spatial memories. Micro-infusion of TNFα into the dorsal hippocampus at 6-18 h before retrieval impaired the retrieval of contextual fear memory, although micro-infusion before contextual fear conditioning had no effect on memory formation. Interestingly, hippocampal TNFα micro-infusion before memory retrieval decreased freezing responses, even at 24 h after retrieval, suggesting that TNFα impairs the reconsolidation of contextual fear memory. Similarly, hippocampal TNFα micro-infusion impaired the retrieval and reconsolidation of spatial memory in the Morris water maze. Consistent with these observations, hippocampal TNFα micro-infusion before retrieval blocked the induction of c-fos expression in the hippocampus, which is a marker of neural activation, in response to the retrieval of contextual fear memory. Collectively, our findings indicate that TNFα negatively regulates the retrieval and reconsolidation of hippocampus-dependent memory.
Collapse
Affiliation(s)
- Shohei Takahashi
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan; Department of Bioscience, Faculty of Life Sciences, Tokyo University of Agriculture, Tokyo 156-8502, Japan
| | - Hotaka Fukushima
- Department of Bioscience, Faculty of Life Sciences, Tokyo University of Agriculture, Tokyo 156-8502, Japan
| | - Zhiqian Yu
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Satoshi Kida
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan; Department of Bioscience, Faculty of Life Sciences, Tokyo University of Agriculture, Tokyo 156-8502, Japan.
| |
Collapse
|
31
|
Alvarez Cooper I, Beecher K, Chehrehasa F, Belmer A, Bartlett SE. Tumour Necrosis Factor in Neuroplasticity, Neurogenesis and Alcohol Use Disorder. Brain Plast 2020; 6:47-66. [PMID: 33680846 PMCID: PMC7903009 DOI: 10.3233/bpl-190095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder is a pervasive and detrimental condition that involves changes in neuroplasticity and neurogenesis. Alcohol activates the neuroimmune system and alters the inflammatory status of the brain. Tumour necrosis factor (TNF) is a well characterised neuroimmune signal but its involvement in alcohol use disorder is unknown. In this review, we discuss the variable findings of TNF's effect on neuroplasticity and neurogenesis. Acute ethanol exposure reduces TNF release while chronic alcohol intake generally increases TNF levels. Evidence suggests TNF potentiates excitatory transmission, promotes anxiety during alcohol withdrawal and is involved in drug use in rodents. An association between craving for alcohol and TNF is apparent during withdrawal in humans. While anti-inflammatory therapies show efficacy in reversing neurogenic deficit after alcohol exposure, there is no evidence for TNF's essential involvement in alcohol's effect on neurogenesis. Overall, defining TNF's role in alcohol use disorder is complicated by poor understanding of its variable effects on synaptic transmission and neurogenesis. While TNF may be of relevance during withdrawal, the neuroimmune system likely acts through a larger group of inflammatory cytokines to alter neuroplasticity and neurogenesis. Understanding the individual relevance of TNF in alcohol use disorder awaits a more comprehensive understanding of TNF's effects within the brain.
Collapse
Affiliation(s)
- Ignatius Alvarez Cooper
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
| | - Kate Beecher
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
| | - Arnauld Belmer
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Selena E. Bartlett
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
32
|
Kogan S, Ospina LH, Mittal VA, Kimhy D. The impact of inflammation on neurocognition and risk for psychosis: a critical review. Eur Arch Psychiatry Clin Neurosci 2020; 270:793-802. [PMID: 31620871 PMCID: PMC7160015 DOI: 10.1007/s00406-019-01073-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022]
Abstract
Neurocognitive difficulties are highly prevalent among people with schizophrenia and have been linked to increased inflammation, as well as dysfunction and disability. Poor neurocognitive functioning has also been documented in individuals at clinical high risk for psychosis (CHR) and a burgeoning literature point to alterations in inflammation markers in this population. However, there is limited information regarding the putative link between inflammation and neurocognition in CHR individuals, and the potential role of inflammation in the development of cognitive difficulties and psychosis. As previous reports indicate that early treatment in schizophrenia is associated with better outcomes, there is an urgent need to identify neurobiological mechanisms underlying cognitive deterioration and psychosis in CHR individuals to provide them with care prior to significant cognitive and functional declines. To address this gap in the literature, we review and summarize the relevant literatures on inflammation and neurocognitive dysfunction in schizophrenia and CHR individuals, point to remaining gaps, and suggest directions for future research.
Collapse
Affiliation(s)
- Sophia Kogan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1230, New York, NY, 10029, USA
| | - Luz H Ospina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1230, New York, NY, 10029, USA
| | - Vijay A Mittal
- Department of Psychology, Northwestern University, Evanston, IL, USA
| | - David Kimhy
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1230, New York, NY, 10029, USA.
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
33
|
Heir R, Stellwagen D. TNF-Mediated Homeostatic Synaptic Plasticity: From in vitro to in vivo Models. Front Cell Neurosci 2020; 14:565841. [PMID: 33192311 PMCID: PMC7556297 DOI: 10.3389/fncel.2020.565841] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Since it was first described almost 30 years ago, homeostatic synaptic plasticity (HSP) has been hypothesized to play a key role in maintaining neuronal circuit function in both developing and adult animals. While well characterized in vitro, determining the in vivo roles of this form of plasticity remains challenging. Since the discovery that the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) mediates some forms of HSP, it has been possible to probe some of the in vivo contribution of TNF-mediated HSP. Work from our lab and others has found roles for TNF-HSP in a variety of functions, including the developmental plasticity of sensory systems, models of drug addiction, and the response to psychiatric drugs.
Collapse
Affiliation(s)
- Renu Heir
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC, Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC, Canada
| |
Collapse
|
34
|
Wang D. Tumor Necrosis Factor-Alpha Alters Electrophysiological Properties of Rabbit Hippocampal Neurons. J Alzheimers Dis 2020; 68:1257-1271. [PMID: 30909246 DOI: 10.3233/jad-190043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have shown tumor necrosis factor-alpha (TNF-α) may impact neurodegeneration in Alzheimer's disease (AD) by regulating amyloid-β and tau pathogenesis. However, it is unclear whether TNF-α has a role in a cholesterol-fed rabbit model of AD or TNF-α affects the electrophysiological properties of rabbit hippocampus. This study was designed to investigate whether long-term feeding of cholesterol diet known to induce AD pathology regulates TNF-α expression in the hippocampus and whether TNF-α would modulate electrophysiological properties of rabbit hippocampal CA1 neurons. TNF-α ELISA showed dietary cholesterol increased hippocampal TNF-α expression in a dose-dependent manner. Whole-cell recordings revealed TNF-α altered the membrane properties of rabbit hippocampal CA1 neurons, which was characterized by a decrease in after-hyperpolarization amplitudes; Field potential recordings showed TNF-α inhibited long-term potentiation but did not influence presynaptic function. Interestingly, TNF-α did not significantly affect the after-hyperpolarization amplitudes of hippocampal CA1 neurons from cholesterol fed rabbits compared to normal chow fed rabbits. In conclusion, dietary cholesterol generated an in vivo model of chronic TNF-α elevation and TNF-α may underlie the learning and memory changes previously seen in the rabbit model of AD by acting as a bridge between dietary cholesterol and brain function and directly modulating the electrophysiological properties of hippocampal CA1 neurons.
Collapse
Affiliation(s)
- Desheng Wang
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA.,Rockefeller Neuroscience Institute, Morgantown, WV, USA
| |
Collapse
|
35
|
Falcicchia C, Tozzi F, Arancio O, Watterson DM, Origlia N. Involvement of p38 MAPK in Synaptic Function and Dysfunction. Int J Mol Sci 2020; 21:ijms21165624. [PMID: 32781522 PMCID: PMC7460549 DOI: 10.3390/ijms21165624] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Many studies have revealed a central role of p38 MAPK in neuronal plasticity and the regulation of long-term changes in synaptic efficacy, such as long-term potentiation (LTP) and long-term depression (LTD). However, p38 MAPK is classically known as a responsive element to stress stimuli, including neuroinflammation. Specific to the pathophysiology of Alzheimer’s disease (AD), several studies have shown that the p38 MAPK cascade is activated either in response to the Aβ peptide or in the presence of tauopathies. Here, we describe the role of p38 MAPK in the regulation of synaptic plasticity and its implication in an animal model of neurodegeneration. In particular, recent evidence suggests the p38 MAPK α isoform as a potential neurotherapeutic target, and specific inhibitors have been developed and have proven to be effective in ameliorating synaptic and memory deficits in AD mouse models.
Collapse
Affiliation(s)
- Chiara Falcicchia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
| | - Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy;
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA;
| | | | - Nicola Origlia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-3153193
| |
Collapse
|
36
|
Effects of Different Continuous Aerobic Training Protocols in a Heterozygous Mouse Model of Niemann-Pick Type C Disease. J Funct Morphol Kinesiol 2020; 5:jfmk5030053. [PMID: 33467268 PMCID: PMC7739240 DOI: 10.3390/jfmk5030053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 01/10/2023] Open
Abstract
The positive effects of physical activity on cognitive functions are widely known. Aerobic training is known to promote the expression of neurotrophins, thus inducing an increase in the development and survival of neurons, as well as enhancing synaptic plasticity. Based on this evidence, in the present study, we analyze the effects of two different types of aerobic training, progressive continuous (PC) and varying continuous (VC), on synaptic and muscular plasticity in heterozygous mice carrying the genetic mutation for Niemann-Pick type C disease. We also analyze the effects on synaptic plasticity by extracellular recordings in vitro in mouse hippocampal slices, while the morphological structure of muscle tissue was studied by transmission electron microscopy. Our results show a modulation of synaptic plasticity that varies according to the type of training protocol used, and only the VC protocol administered twice a week, has a significantly positive effect on long-term potentiation. On the contrary, ultrastructural analysis of muscle tissue shows an improvement in cellular conditions in all trained mice. These results confirm the beneficial effects of exercise on quality of life, supporting the hypothesis that physical activity could represent an alternative therapeutic strategy for patients with Niemann-Pick type C disease.
Collapse
|
37
|
Administration of Bacterial Lipopolysaccharide during Early Postnatal Ontogenesis Induces Transient Impairment of Long-Term Synaptic Plasticity Associated with Behavioral Abnormalities in Young Rats. Pharmaceuticals (Basel) 2020; 13:ph13030048. [PMID: 32197321 PMCID: PMC7151710 DOI: 10.3390/ph13030048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 01/31/2023] Open
Abstract
Infectious diseases in early postnatal ontogenesis often result in cognitive impairments, particularly learning and memory. The essential foundation of learning and memory is long-term synaptic plasticity, which depends on N-methyl-D-aspartate (NMDA) receptors. In the present study, bacterial infection was modeled by treating rat pups with bacterial lipopolysaccharide (LPS, 25 µg/kg) three times, during either the first or the third week of life. These time points are critical for the maturation of NMDA receptors. We assessed the effects of LPS treatments on the properties of long-term potentiation (LTP) in the CA1 hippocampus of young (21–23 days) and adolescent (51–55 days) rats. LTP magnitude was found to be significantly reduced in both groups of young rats, which also exhibited investigative and motor behavior disturbances in the open field test. No changes were observed in the main characteristics of synaptic transmission, although the LTP induction mechanism was disturbed. In rats treated with LPS during the third week, the NMDA-dependent form of LTP was completely suppressed, and LTP switched to the Type 1 metabotropic glutamate receptor (mGluR1)-dependent form. These impairments of synaptic plasticity and behavior were temporary. In adolescent rats, no difference was observed in LTP properties between the control and experimental groups. Lastly, the investigative and motor behavior parameters in both groups of adult rats were similar.
Collapse
|
38
|
Contreras JA, Aslanyan V, Sweeney MD, Sanders LMJ, Sagare AP, Zlokovic BV, Toga AW, Han SD, Morris JC, Fagan A, Massoumzadeh P, Benzinger TL, Pa J. Functional connectivity among brain regions affected in Alzheimer's disease is associated with CSF TNF-α in APOE4 carriers. Neurobiol Aging 2020; 86:112-122. [PMID: 31870643 PMCID: PMC7205323 DOI: 10.1016/j.neurobiolaging.2019.10.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/20/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023]
Abstract
It is now recognized that understanding how neuroinflammation affects brain function may provide new insights into Alzheimer's pathophysiology. Tumor necrosis factor (TNF)-α, an inflammatory cytokine marker, has been implicated in Alzheimer's disease (AD), as it can impair neuronal function through suppression of long-term potentiation. Our study investigated the relationship between cerebrospinal fluid TNF-α and functional connectivity (FC) in a cohort of 64 older adults (μ age = 69.76 years; 30 cognitively normal, 34 mild AD). Higher cerebrospinal fluid TNF-α levels were associated with lower FC among brain regions important for high-level decision-making, inhibitory control, and memory. This effect was moderated by apolipoprotein E-ε4 (APOE4) status. Graph theory metrics revealed there were significant differences between APOE4 carriers at the node level, and by diagnosis at the network level suggesting global brain network dysfunction in participants with AD. These findings suggest proinflammatory mechanisms may contribute to reduced FC in regions important for high-level cognition. Future studies are needed to understand the role of inflammation on brain function and clinical progression, especially in APOE4 carriers.
Collapse
Affiliation(s)
- Joey Annette Contreras
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Vahan Aslanyan
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Melanie D Sweeney
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Lianne M J Sanders
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA; Department of Human Movement Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Abhay P Sagare
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - S Duke Han
- Family Medicine, Neurology, Psychology, and Gerontology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - John C Morris
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; Department of Neurology, Washington University, St Louis, MO, USA
| | - Anne Fagan
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; Department of Neurology, Washington University, St Louis, MO, USA
| | - Parinaz Massoumzadeh
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA
| | - Tammie L Benzinger
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; Department of Radiology and Neurological Surgery, Washington University, St. Louis, MO, USA
| | - Judy Pa
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
39
|
Lathe R, Singadia S, Jordan C, Riedel G. The interoceptive hippocampus: Mouse brain endocrine receptor expression highlights a dentate gyrus (DG)-cornu ammonis (CA) challenge-sufficiency axis. PLoS One 2020; 15:e0227575. [PMID: 31940330 PMCID: PMC6961916 DOI: 10.1371/journal.pone.0227575] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
The primeval function of the mammalian hippocampus (HPC) remains uncertain. Implicated in learning and memory, spatial navigation, and neuropsychological disorders, evolutionary theory suggests that the HPC evolved from a primeval chemosensory epithelium. Deficits in sensing of internal body status ('interoception') in patients with HPC lesions argue that internal sensing may be conserved in higher vertebrates. We studied the expression patterns in mouse brain of 250 endocrine receptors that respond to blood-borne ligands. Key findings are (i) the proportions and levels of endocrine receptor expression in the HPC are significantly higher than in all other comparable brain regions. (ii) Surprisingly, the distribution of endocrine receptor expression within mouse HPC was found to be highly structured: receptors signaling 'challenge' are segregated in dentate gyrus (DG), whereas those signaling 'sufficiency' are principally found in cornu ammonis (CA) regions. Selective expression of endocrine receptors in the HPC argues that interoception remains a core feature of hippocampal function. Further, we report that ligands of DG receptors predominantly inhibit both synaptic potentiation and neurogenesis, whereas CA receptor ligands conversely promote both synaptic potentiation and neurogenesis. These findings suggest that the hippocampus acts as an integrator of body status, extending its role in context-dependent memory encoding from 'where' and 'when' to 'how I feel'. Implications for anxiety and depression are discussed.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, University of Edinburgh Medical School, Little France, Edinburgh, Scotland, United Kingdom
- * E-mail: (RL); (GR)
| | - Sheena Singadia
- Division of Behavioral Neuroscience, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, United Kingdom
| | - Crispin Jordan
- Division of Biomedical Sciences, University of Edinburgh Medical School, George Square, Edinburgh, Scotland, United Kingdom
| | - Gernot Riedel
- Division of Behavioral Neuroscience, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, United Kingdom
- * E-mail: (RL); (GR)
| |
Collapse
|
40
|
Bourgognon JM, Cavanagh J. The role of cytokines in modulating learning and memory and brain plasticity. Brain Neurosci Adv 2020; 4:2398212820979802. [PMID: 33415308 PMCID: PMC7750764 DOI: 10.1177/2398212820979802] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/18/2020] [Indexed: 12/28/2022] Open
Abstract
Cytokines are proteins secreted in the central nervous system by neurons, microglia, astrocytes and infiltrating peripheral immune cells under physiological and pathological conditions. Over the last 20 years, a growing number of reports have investigated the effects of these molecules on brain plasticity. In this review, we describe how the key cytokines interleukin 1β, interleukin 6 and tumour necrosis factor α were found to support long-term plasticity and learning and memory processes in physiological conditions. In contrast, during inflammation where cytokines levels are elevated such as in models of brain injury or infection, depression or neurodegeneration, the effects of cytokines are mostly detrimental to memory mechanisms, associated behaviours and homeostatic plasticity.
Collapse
Affiliation(s)
| | - Jonathan Cavanagh
- Institute of Infection, Immunity &
Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
41
|
Alnefeesi Y, Siegel A, Lui LMW, Teopiz KM, Ho RCM, Lee Y, Nasri F, Gill H, Lin K, Cao B, Rosenblat JD, McIntyre RS. Impact of SARS-CoV-2 Infection on Cognitive Function: A Systematic Review. Front Psychiatry 2020; 11:621773. [PMID: 33643083 PMCID: PMC7902710 DOI: 10.3389/fpsyt.2020.621773] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
The prevalence and etiology of COVID-19's impact on brain health and cognitive function is poorly characterized. With mounting reports of delirium, systemic inflammation, and evidence of neurotropism, a statement on cognitive impairment among COVID-19 cases is needed. A substantial literature has demonstrated that inflammation can severely disrupt brain function, suggesting an immune response, a cytokine storm, as a possible cause of neurocognitive impairments. In this light, the aim of the present study was to summarize the available knowledge of the impact of COVID-19 on cognition (i.e., herein, we broadly define cognition reflecting the reporting on this topic in the literature) during the acute and recovery phases of the disease, in hospitalized patients and outpatients with confirmed COVID-19 status. A systematic review of the literature identified six studies which document the prevalence of cognitive impairment, and one which quantifies deficits after recovery. Pooling the samples of the included studies (total sample n = 644) at three standards of quality produced conservative estimates of cognitive impairment ranging from 43.0 to 66.8% prevalence in hospitalized COVID-19 patients only, as no studies which report on outpatients met criteria for inclusion in the main synthesis. The most common impairment reported was delirium and frequent reports of elevated inflammatory markers suggest etiology. Other studies have demonstrated that the disease involves marked increases in IL-6, TNFα, and IL-1β; cytokines known to have a profound impact on working memory and attention. Impairment of these cognitive functions is a characteristic aspect of delirium, which suggests these cytokines as key mediators in the etiology of COVID-19 induced cognitive impairments. Researchers are encouraged to assay inflammatory markers to determine the potential role of inflammation in mediating the disturbance of cognitive function in individuals affected by COVID-19.
Collapse
Affiliation(s)
- Yazen Alnefeesi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Ashley Siegel
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Kayla M Teopiz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Roger C M Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Flora Nasri
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Kangguang Lin
- Department of Affective Disorder, the Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou Medical University, Guangzhou, China.,Laboratory of Emotion and Cognition, the Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou Medical University, Guangzhou, China
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology, University of Toronto, Toronto, ON, Canada.,Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| |
Collapse
|
42
|
Tumor Necrosis Factor-α-Mediated Metaplastic Inhibition of LTP Is Constitutively Engaged in an Alzheimer's Disease Model. J Neurosci 2019; 39:9083-9097. [PMID: 31570539 DOI: 10.1523/jneurosci.1492-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
Abstract
LTP, a fundamental mechanism of learning and memory, is a highly regulated process. One form of regulation is metaplasticity (i.e., the activity-dependent and long-lasting changes in neuronal state that orchestrate the direction, magnitude, and persistence of future synaptic plasticity). We have previously described a heterodendritic metaplasticity effect, whereby strong high-frequency priming stimulation in stratum oriens inhibits subsequent LTP in the stratum radiatum of hippocampal area CA1, potentially by engagement of the enmeshed astrocytic network. This effect may occur due to neuron-glia interactions in response to priming stimulation that leads to the release of gliotransmitters. Here we found in male rats that TNFα and associated signal transduction enzymes, but not interleukin-1β (IL-1β), were responsible for mediating the metaplasticity effect. Replacing priming stimulation with TNFα incubation reproduced these effects. As TNFα levels are elevated in Alzheimer's disease, we examined whether heterodendritic metaplasticity is dysregulated in a transgenic mouse model of the disease, either before or after amyloid plaque formation. We showed that TNFα and IL-1β levels were significantly increased in aged but not young transgenic mice. Although control LTP was impaired in the young transgenic mice, it was not TNFα-dependent. In the older transgenic mice, however, LTP was impaired in a way that occluded further reduction by heterosynaptic metaplasticity, whereas LTP was entirely rescued by incubation with a TNFα antibody, but not an IL-1β antibody. Thus, TNFα mediates a heterodendritic metaplasticity in healthy rodents that becomes constitutively and selectively engaged in a mouse model of Alzheimer's disease.SIGNIFICANCE STATEMENT The proinflammatory cytokine TNFα is known to be capable of inhibiting LTP and is upregulated several-fold in brain tissue, serum, and CSF of Alzheimer's disease (AD) patients. However, the mechanistic roles played by TNFα in plasticity and AD remain poorly understood. Here we show that TNFα and its downstream signaling molecules p38 MAPK, ERK, and JNK contribute fundamentally to a long-range metaplastic inhibition of LTP in rats. Moreover, the impaired LTP in aged APP/PS1 mice is rescued by incubation with a TNFα antibody. Thus, there is an endogenous engagement of the metaplasticity mechanism in this mouse model of AD, supporting the idea that blocking TNFα might be of therapeutic benefit in the disease.
Collapse
|
43
|
Ethanol Induction of Innate Immune Signals Across BV2 Microglia and SH-SY5Y Neuroblastoma Involves Induction of IL-4 and IL-13. Brain Sci 2019; 9:brainsci9090228. [PMID: 31510019 PMCID: PMC6770440 DOI: 10.3390/brainsci9090228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023] Open
Abstract
Innate immune signaling molecules, such as Toll-like receptors (TLRs), cytokines and transcription factor NFκB, are increased in post-mortem human alcoholic brain and may play roles in alcohol dependence and neurodegeneration. Innate immune signaling involves microglia -neuronal signaling which while poorly understood, may impact learning and memory. To investigate mechanisms of ethanol induction of innate immune signaling within and between brain cells, we studied immortalized BV2 microglia and SH-SY5Y human neuroblastoma to model microglial and neuronal signaling. Cells were treated alone or in co-culture using a Transwell system, which allows transfer of soluble mediators. We determined immune signaling mRNA using real-time polymerase chain reaction. Ethanol induced innate immune genes in both BV2 and SH-SY5Y cultured alone, with co-culture altering gene expression at baseline and following ethanol exposure. Co-culture blunted ethanol-induced high mobility group box protein 1 (HMGB1)-TLR responses, corresponding with reduced ethanol induction of several proinflammatory NFκB target genes. In contrast, co-culture resulted in ethanol upregulation of cytokines IL-4 and IL-13 in BV2 and corresponding receptors, that is, IL-4 and IL-13 receptors, in SH-SY5Y, suggesting induction of a novel signaling pathway. Co-culture reduction in HMGB1-TLR levels occurs in parallel with reduced proinflammatory gene induction and increased IL-4 and IL-13 ligands and receptors. Findings from these immortalized and tumor-derived cell lines could provide insight into microglial-neuronal interactions via release of soluble mediators in vivo.
Collapse
|
44
|
Activation of microglia in acute hippocampal slices affects activity-dependent long-term potentiation and synaptic tagging and capture in area CA1. Neurobiol Learn Mem 2019; 163:107039. [DOI: 10.1016/j.nlm.2019.107039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
|
45
|
Matta SM, Hill-Yardin EL, Crack PJ. The influence of neuroinflammation in Autism Spectrum Disorder. Brain Behav Immun 2019; 79:75-90. [PMID: 31029798 DOI: 10.1016/j.bbi.2019.04.037] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social communication and restricted or repetitive behaviours. The clinical presentation of ASD is highly variable and diagnosis is based on the presence of impaired social communication and repetitive and/or restricted behaviours. Although the precise pathophysiologies underlying ASD are unclear, growing evidence supports a role for dysregulated neuroinflammation. The potential involvement of microglia and astrocytes reactive to inflammatory stimuli in ASD has generated much interest due to their varied roles including in mounting an immune response and regulating synaptic function. Increased numbers of reactive microglial and astrocytes in both ASD postmortem tissue and animal models have been reported. Whether dysregulation of glial subtypes exacerbates alterations in neural connectivity in the brain of autistic patients is not well explored. A role for the gut-brain axis involving microbial-immune-neuronal cross talk is also a growing area of neuroinflammation research. Greater understanding of these interactions under patho/physiological conditions and the identification of consistent immune profile abnormalities can potentially lead to more reliable diagnostic measures and treatments in ASD.
Collapse
Affiliation(s)
- Samantha M Matta
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elisa L Hill-Yardin
- School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; Department of Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
46
|
Abstract
Innate immune signaling is an important feature in the pathology of alcohol use disorders. Alcohol abuse causes persistent innate immune activation in the brain. This is seen in postmortem human alcoholic brain specimens, as well as in primate and rodent models of alcohol consumption. Further, in vitro models of alcohol exposure in neurons and glia also demonstrate innate immune activation. The activation of the innate immune system seems to be important in the development of alcohol use pathology, as anti-immune therapies reduce pathology and ethanol self-administration in rodent models. Further, innate immune activation has been identified in each of the stages of addiction: binge/intoxication, withdrawal/negative affect, and preoccupation/craving. This suggests that innate immune activation may play a role both in the development and maintenance of alcoholic pathology. In this chapter, we discuss the known contributions of innate immune signaling in the pathology of alcohol use disorders, and present potential therapeutic interventions that may be beneficial for alcohol use disorders.
Collapse
Affiliation(s)
- Leon G Coleman
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
47
|
Lindsey LP, Daphney CM, Oppong-Damoah A, Uchakin PN, Abney SE, Uchakina ON, Khusial RD, Akil A, Murnane KS. The cannabinoid receptor 2 agonist, β-caryophyllene, improves working memory and reduces circulating levels of specific proinflammatory cytokines in aged male mice. Behav Brain Res 2019; 372:112012. [PMID: 31173795 DOI: 10.1016/j.bbr.2019.112012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/30/2022]
Abstract
Age-related cognitive decline has been associated with proinflammatory cytokines, yet the precise relationship between cognitive decline and cytokine load remains to be elucidated. β-caryophyllene (BCP) is a cannabinoid receptor 2 (CB2) agonist with established anti-inflammatory effects that is known to improve memory and increase lifespan. It is of interest to explore the potential of BCP to reduce age-related cognitive decline and proinflammatory cytokine load. In this study, we assessed changes in circulating cytokines across the lifespan, memory performance in young and aged mice, and the effects of BCP on memory function and cytokine load. The plasma levels of 12 cytokines were assessed in male Swiss-Webster mice at 3, 12, and 18 months of age using multiplexed flow cytometry. Working memory was compared in 3 and 12 month-old mice using spontaneous alternations. A dose-response function (100-300 mg/kg, subchronic administration) for BCP-induced memory restoration was determined in 3- and 12- month-old mice. Finally, the effects on cytokine levels of the peak memory enhancing dose of BCP were assessed in 18- month-old mice. Circulating levels of several cytokines significantly increased with age. Multilinear regression analysis showed that IL-23 levels were most strongly associated with age. Aged mice showed deficits in working memory and higher levels of IL-23, both of which were reversed by BCP treatment. BCP appears to reverse age-associated impairments in memory and modulates cytokine production. IL-23 may play a significant role in the aging process, and future research should determine whether it has utility as a biomarker for novel anti-aging therapeutics.
Collapse
Affiliation(s)
- Lindsey Phillips Lindsey
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Cedrick Maceo Daphney
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Aboagyewaah Oppong-Damoah
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Peter Nikolaevich Uchakin
- Department of Biomedical Sciences, Mercer University School of Medicine, Mercer University Health Sciences Center, Macon, GA, USA
| | - Sarah E Abney
- Department of Biomedical Sciences, Mercer University School of Medicine, Mercer University Health Sciences Center, Macon, GA, USA
| | - Olga N Uchakina
- Department of Biomedical Sciences, Mercer University School of Medicine, Mercer University Health Sciences Center, Macon, GA, USA
| | - Richard Darien Khusial
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Ayman Akil
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA
| | - Kevin Sean Murnane
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, GA, USA.
| |
Collapse
|
48
|
Maggio N, Vlachos A. Tumor necrosis factor (TNF) modulates synaptic plasticity in a concentration-dependent manner through intracellular calcium stores. J Mol Med (Berl) 2018; 96:1039-1047. [PMID: 30073573 DOI: 10.1007/s00109-018-1674-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/22/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022]
Abstract
The role of inflammatory signaling pathways in synaptic plasticity has long been identified. Yet, it remains unclear how inflammatory cytokines assert their pleiotropic effects on neural plasticity. Moreover, the neuronal targets through which inflammatory cytokines assert their effects on plasticity remain not well-understood. In an attempt to learn more about the plasticity-modulating effects of the pro-inflammatory cytokine tumor necrosis factor (TNF), we used two-pathway long-term potentiation (LTP) experiments at Schaffer collateral-CA1 synapses to test for concentration-dependent effects of TNF on synaptic plasticity. We report that high concentrations of TNF (1 μg/mL) impair the ability of mouse CA1 pyramidal neurons to express synaptic plasticity without affecting baseline synaptic transmission and/or previously established LTP. Interestingly, 100 ng/mL of TNF has no apparent effect on LTP, while low concentrations (1 ng/mL) promote the ability of neurons to express LTP. These dose-dependent metaplastic effects of TNF are modulated by intracellular calcium stores: Pharmacological activation of intracellular calcium stores with ryanodine (10 μM) reverses the negative effects of TNF[high], and the plasticity-promoting effects of TNF[low] are blocked when intracellular calcium stores are depleted with thapsigargin (1 μM). Consistent with this result, TNF does not promote plasticity in synaptopodin-deficient preparations, which show deficits in neuronal calcium store-mediated synaptic plasticity. Thus, we propose that TNF mediates its pleiotropic effects on synaptic plasticity in a concentration-dependent manner through signaling pathways that are modulated by intracellular calcium stores and require the presence of synaptopodin. These results demonstrate that TNF can act as mediator of metaplasticity, which is of considerable relevance in the context of brain diseases associated with increased TNF levels and alterations in synaptic plasticity. KEY MESSAGES • TNF modulates the ability of neurons to express synaptic plasticity. • High concentrations of TNF impair synaptic plasticity. • Low concentrations of TNF improve synaptic plasticity. • TNF does not affect previously established long-term potentiation. • Plasticity effects of TNF are modulated by intracellular calcium stores.
Collapse
Affiliation(s)
- Nicola Maggio
- Department of Neurology and Sagol Center for Neurosciences, Sheba Medical Center, 52621, Ramat Gan, Israel. .,Department of Neurology, The Chaim Sheba Medical Center, 52621, Tel HaShomer, Israel. .,Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, 52621, Tel HaShomer, Israel. .,Department of Neurology and Neurosurgery, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 52621, Tel Aviv, Israel.
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
49
|
Ignatowski TA, Spengler RN. Targeting tumor necrosis factor in the brain relieves neuropathic pain. World J Anesthesiol 2018; 7:10-19. [DOI: 10.5313/wja.v7.i2.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/28/2018] [Accepted: 06/02/2018] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain is a chronic syndrome caused by direct damage to or disease of the somatosensory nervous system. The lack of safe, adequate and sustained pain relief offered by present analgesic treatments is most alarming. While many treatment options are available to manage chronic pain, such as antidepressants, non-steroidal anti-inflammatory agents, opioids, and anticonvulsants, chronic neuropathic pain remains largely unmanaged. Compounding the dilemma of ineffective chronic pain treatments is the need to provide relief from suffering and yet not contribute to the scourge of drug abuse. A recent epidemic of addiction and accidental drug prescription overdoses parallel the increased use of opioid treatment, even though opioids are rarely an effective treatment of relieving chronic pain. To make matters worse, opioids may contribute to exacerbating pain, and side-effects such as cognitive impairment, nausea, constipation, development of tolerance, as well as their potential for addiction and overdose deaths exist. Clearly, there is an urgent need for alternative, non-opiate treatment of chronic pain. Innovative discoveries of pertinent brain mechanisms and functions are key to developing effective, safe treatments. Pioneering work has revealed the essential effects of the pleiotropic mediator tumor necrosis factor (TNF) on brain functioning. These studies establish that TNF inhibits norepinephrine release from hippocampal neurons, and show that excess TNF production within the hippocampus occurs during neuropathic pain, which mobilizes additional mechanisms that further inhibit norepinephrine release. Significantly, it has been verified that elevated levels of TNF in the brain are actually required for neuropathic pain development. Since TNF decreases norepinephrine release in the brain, enhanced TNF levels would prevent engagement of the norepinephrine descending inhibitory neuronal pain pathways. Increased levels of TNF in the brain are therefore critical to the development of neuropathic pain. Therefore, strategies that decrease this enhanced TNF expression in the brain will have superior analgesic efficacy. We propose this novel approach of targeting the pathologically high levels of brain TNF as an effective strategy in the treatment of the devastating syndrome of chronic pain.
Collapse
Affiliation(s)
- Tracey A Ignatowski
- Department of Pathology and Anatomical Sciences and Program for Neuroscience, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14203, United States
- NanoAxis, LLC, Clarence, NY 14031, United States
| | | |
Collapse
|
50
|
Synaptic transmission and excitability during hypoxia with inflammation and reoxygenation in hippocampal CA1 neurons. Neuropharmacology 2018; 138:20-31. [PMID: 29775678 DOI: 10.1016/j.neuropharm.2018.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/19/2018] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
Although a number of experimental and clinical studies have shown that hypoxia typically accompanies acute inflammatory responses, the combinatorial effect of the two insults on basic neural function has not been thoroughly investigated. Previous studies have predominantly suggested that hypoxia reduces network activity; however, several studies suggest the opposite effect. Of note, inflammation is known to increase neural activity. In the current study, we examined the effects of limited oxygen in combination with an inflammatory stimulus, as well as the effects of reoxygenation, on synaptic transmission and excitability. We observed a significant reduction of both synaptic transmission and excitability when hypoxia and inflammation occurred in combination, whereas reoxygenation caused hyperexcitability of neurons. Further, we found that the observed reduction in synaptic transmission was due to compromised presynaptic release efficiency based on an adenosine-receptor-dependent increase in synaptic facilitation. Excitability changes in both directions were attributable to dynamic regulation of the hyperpolarization-activated cation current (Ih) and to changes in the input resistance and the voltage difference between resting membrane potential and action potential threshold. We found that zatebradine, an Ih current inhibitor, reduced the fluctuation in excitability, suggesting that it may have potential as a drug to ameliorate reperfusion brain injury.
Collapse
|