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 2025; 25:321-352. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
Ziabska K, Gewartowska M, Frontczak-Baniewicz M, Sypecka J, Ziemka-Nalecz M. The Impact of the Histone Deacetylase Inhibitor-Sodium Butyrate on Complement-Mediated Synapse Loss in a Rat Model of Neonatal Hypoxia-Ischemia. Mol Neurobiol 2025; 62:5216-5233. [PMID: 39531190 PMCID: PMC11880148 DOI: 10.1007/s12035-024-04591-w] [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: 07/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Perinatal asphyxia is one of the most important causes of morbidity and mortality in newborns. One of the key pathogenic factors in hypoxic-ischemic (HI) brain injury is the inflammatory reaction including complement system activation. Over-activated complement stimulates cells to release inflammatory molecules and is involved in the post-ischemic degradation of synaptic connections. On the other hand, complement is also involved in regenerative processes. The histone deacetylase inhibitor (HDACi)-sodium butyrate (SB)-provides reduction of inflammation by decreasing the expression of the proinflammatory factors. The main purpose of this study was to examine the effect of SB treatment on complement activation and synapse elimination after HI. Neonatal HI was induced in Wistar rats pups by unilateral ligation of the common carotid artery followed by 60-min hypoxia (7.6% O2). SB (300 mg/kg) was administered on a 5-day regimen. Our study has shown decreased levels of synapsin I, synaptophysin, and PSD-95 in the hypoxic-ischemic hemisphere, indicating synaptic loss after neonatal HI. Transmission electron microscopy revealed injury of the synaptic structures in the brain after HI. SB treatment increased the level of the synaptic proteins, improved tissue ultrastructure, and reduced degradation of the synapses. Neonatal HI induced mRNA expression of the complement C1q, C3, C5, and C9, and their receptors C3aR and C5aR. The effect of SB was different depending on the time after induction of hypoxic-ischemic damage. Our study demonstrated that neuroprotective effect of SB may be related to the modulation of complement activity after HI brain injury.
Collapse
Affiliation(s)
- Karolina Ziabska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Magdalena Gewartowska
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Malgorzata Frontczak-Baniewicz
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
- Higher School of Engineering and Health in Warsaw, 18 Bitwy Warszawskiej 1920r. Street, 02-366, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland.
| |
Collapse
|
3
|
Tenner AJ, Petrisko TJ. Knowing the enemy: strategic targeting of complement to treat Alzheimer disease. Nat Rev Neurol 2025:10.1038/s41582-025-01073-y. [PMID: 40128350 DOI: 10.1038/s41582-025-01073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/26/2025]
Abstract
The complement system protects against infection, positively responds to tissue damage, clears cell debris, directs and modulates the adaptive immune system, and functions in neuronal development, normal synapse elimination and intracellular metabolism. However, complement also has a role in aberrant synaptic pruning and neuroinflammation - processes that lead to a feedforward loop of inflammation, injury and neuronal death that can contribute to neurodegenerative and neurological disorders, including Alzheimer disease. This Review provides justification, largely from preclinical mouse models but also from correlates with human tissue and biomarkers, for targeting specific complement components for therapeutic intervention in Alzheimer disease. We discuss promising strategies to slow the progression of cognitive loss with minimal undesired effects. The diverse interactions and functions of complement system components can influence biological processes in the healthy and diseased brain; here, these functions are described as a prerequisite to selecting appropriate, safe and effective therapeutic targets for translation to the clinic.
Collapse
Affiliation(s)
- Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA.
| | - Tiffany J Petrisko
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
4
|
Quintanilla B, Zarate CA, Pillai A. Ketamine's mechanism of action with an emphasis on neuroimmune regulation: can the complement system complement ketamine's antidepressant effects? Mol Psychiatry 2024; 29:2849-2858. [PMID: 38575806 PMCID: PMC11804209 DOI: 10.1038/s41380-024-02507-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
Over 300 million people worldwide suffer from major depressive disorder (MDD). Unfortunately, only 30-40% of patients with MDD achieve complete remission after conventional monoamine antidepressant therapy. In recent years, ketamine has revolutionized the treatment of MDD, with its rapid antidepressant effects manifesting within a few hours as opposed to weeks with conventional antidepressants. Many research endeavors have sought to identify ketamine's mechanism of action in mood disorders; while many studies have focused on ketamine's role in glutamatergic modulation, several studies have implicated its role in regulating neuroinflammation. The complement system is an important component of the innate immune response vital for synaptic plasticity. The complement system has been implicated in the pathophysiology of depression, and studies have shown increases in complement component 3 (C3) expression in the prefrontal cortex of suicidal individuals with depression. Given the role of the complement system in depression, ketamine and the complement system's abilities to modulate glutamatergic transmission, and our current understanding of ketamine's anti-inflammatory properties, there is reason to suspect a common link between the complement system and ketamine's mechanism of action. This review will summarize ketamine's anti- inflammatory roles in the periphery and central nervous system, with an emphasis on complement system regulation.
Collapse
Affiliation(s)
- Brandi Quintanilla
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
5
|
Schartz ND, Liang HY, Carvalho K, Chu SH, Mendoza-Arvilla A, Petrisko TJ, Gomez-Arboledas A, Mortazavi A, Tenner AJ. C5aR1 antagonism suppresses inflammatory glial responses and alters cellular signaling in an Alzheimer's disease mouse model. Nat Commun 2024; 15:7028. [PMID: 39147742 PMCID: PMC11327341 DOI: 10.1038/s41467-024-51163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults, and the need for effective, sustainable therapeutic targets is imperative. The complement pathway has been proposed as a therapeutic target. C5aR1 inhibition reduces plaque load, gliosis, and memory deficits in animal models, however, the cellular bases underlying this neuroprotection were unclear. Here, we show that the C5aR1 antagonist PMX205 improves outcomes in the Arctic48 mouse model of AD. A combination of single cell and single nucleus RNA-seq analysis of hippocampi derived from males and females identified neurotoxic disease-associated microglia clusters in Arctic mice that are C5aR1-dependent, while microglial genes associated with synapse organization and transmission and learning were overrepresented in PMX205-treated mice. PMX205 also reduced neurotoxic astrocyte gene expression, but clusters associated with protective responses to injury were unchanged. C5aR1 inhibition promoted mRNA-predicted signaling pathways between brain cell types associated with cell growth and repair, while suppressing inflammatory pathways. Finally, although hippocampal plaque load was unaffected, PMX205 prevented deficits in short-term memory in female Arctic mice. In conclusion, C5aR1 inhibition prevents cognitive loss, limits detrimental glial polarization while permitting neuroprotective responses, as well as leaving most protective functions of complement intact, making C5aR1 antagonism an attractive therapeutic strategy for AD.
Collapse
Affiliation(s)
- Nicole D Schartz
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Heidi Y Liang
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Klebea Carvalho
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Shu-Hui Chu
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Adrian Mendoza-Arvilla
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Tiffany J Petrisko
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Angela Gomez-Arboledas
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Ali Mortazavi
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA.
| |
Collapse
|
6
|
Yang C, Wu X, Feng J, Wu C, Cui X, Wang Z, Yang L. Clinical values of serum C5a in Alzheimer's disease patients with different dementia stages. Neurosci Lett 2024; 836:137833. [PMID: 38796095 DOI: 10.1016/j.neulet.2024.137833] [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: 03/14/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Alzheimer's disease (AD) is characterized by abnormal inflammatory responses, and complement C5a (C5a) is known to initiate inflammation. This study aimed to investigate the associations between serum C5a, inflammatory responses, and cognitive function in AD patients. A total of 242 AD patients and 132 age-matched controls were included. Enzyme-linked immunosorbent assay revealed increased levels of C5a, interleukin (IL)-4, IL-6, IL-10, IL-1β, and tumor necrosis factor (TNF)-α with advancing stages of AD. Pearson correlation coefficient and receiver operating characteristic curve revealed positive correlations between serum C5a levels, inflammatory cytokine levels, Neuropsychiatric Inventory (NPI) and Activities of Daily Living (ADL) scores, and negative correlations with Mini-mental State Examination (MMSE) and Montreal cognitive assessment (MoCA) scores. Serum C5a above 68.68 pg/mL could aid in the diagnosis of AD. Multivariable logistic analysis revealed that serum C5a was an independent risk factor for IL-1β/IL-6/IL-10/TNF-α and an independent protective factor for IL-4. Higher serum C5a levels were associated with lower MMSE and MoCA scores. In conclusion, elevated serum C5a levels were beneficial for AD diagnosis and predictive of inflammation and cognitive dysfunction.
Collapse
Affiliation(s)
- Caijia Yang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xian Wu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jun Feng
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Chunyu Wu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xing Cui
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zeyu Wang
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, China
| | - Lizhen Yang
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
7
|
Liu H, Jiang M, Chen Z, Li C, Yin X, Zhang X, Wu M. The Role of the Complement System in Synaptic Pruning after Stroke. Aging Dis 2024:AD.2024.0373. [PMID: 39012667 DOI: 10.14336/ad.2024.0373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/25/2024] [Indexed: 07/17/2024] Open
Abstract
Stroke is a serious disease that can lead to local neurological dysfunction and cause great harm to the patient's health due to blood cerebral circulation disorder. Synaptic pruning is critical for the normal development of the human brain, which makes the synaptic circuit completer and more efficient by removing redundant synapses. The complement system is considered a key player in synaptic loss and cognitive impairment in neurodegenerative disease. After stroke, the complement system is over-activated, and complement proteins can be labeled on synapses. Microglia and astrocytes can recognize and engulf synapses through corresponding complement receptors. Complement-mediated excessive synaptic pruning can cause post-stroke cognitive impairment (PSCI) and secondary brain damage. This review summarizes the latest progress of complement-mediated synaptic pruning after stroke and the potential mechanisms. Targeting complement-mediated synaptic pruning may be essential for exploring therapeutic strategies for secondary brain injury (SBI) and neurological dysfunction after stroke.
Collapse
Affiliation(s)
- Hongying Liu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| | - Zhiying Chen
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
| | - Chuan Li
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
| | - Xiaorong Zhang
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| | - Moxin Wu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| |
Collapse
|
8
|
Liu Y, Cui W, Liu H, Yao M, Shen W, Miao L, Wei J, Liang X, Zhang Y. Exploring the "gene-metabolite" network of ischemic stroke with blood stasis and toxin syndrome by integrated transcriptomics and metabolomics strategy. Sci Rep 2024; 14:11947. [PMID: 38789486 PMCID: PMC11126742 DOI: 10.1038/s41598-024-61633-y] [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: 01/01/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
A research model combining a disease and syndrome can provide new ideas for the treatment of ischemic stroke. In the field of traditional Chinese medicine, blood stasis and toxin (BST) syndrome is considered an important syndrome seen in patients with ischemic stroke (IS). However, the biological basis of IS-BST syndrome is currently not well understood. Therefore, this study aimed to explore the biological mechanism of IS-BST syndrome. This study is divided into two parts: (1) establishment of an animal model of ischemic stroke disease and an animal model of BST syndrome in ischemic stroke; (2) use of omics methods to identify differentially expressed genes and metabolites in the models. We used middle cerebral artery occlusion (MCAO) surgery to establish the disease model, and utilized carrageenan combined with active dry yeast and MCAO surgery to construct the IS-BST syndrome model. Next, we used transcriptomics and metabolomics methods to explore the differential genes and metabolites in the disease model and IS-BST syndrome model. It is found that the IS-BST syndrome model exhibited more prominent characteristics of IS disease and syndrome features. Both the disease model and the IS-BST syndrome model share some common biological processes, such as thrombus formation, inflammatory response, purine metabolism, sphingolipid metabolism, and so on. Results of the "gene-metabolite" network revealed that the IS-BST syndrome model exhibited more pronounced features of complement-coagulation cascade reactions and amino acid metabolism disorders. Additionally, the "F2 (thrombin)-NMDAR/glutamate" pathway was coupled with the formation process of the blood stasis and toxin syndrome. This study reveals the intricate mechanism of IS-BST syndrome, offering a successful model for investigating the combination of disease and syndrome.
Collapse
Affiliation(s)
- Yue Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wenqiang Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongxi Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Mingjiang Yao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Shen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Lina Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jingjing Wei
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Xiao Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Yunling Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
9
|
Nimmo J, Byrne R, Daskoulidou N, Watkins L, Carpanini S, Zelek W, Morgan B. The complement system in neurodegenerative diseases. Clin Sci (Lond) 2024; 138:387-412. [PMID: 38505993 PMCID: PMC10958133 DOI: 10.1042/cs20230513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Complement is an important component of innate immune defence against pathogens and crucial for efficient immune complex disposal. These core protective activities are dependent in large part on properly regulated complement-mediated inflammation. Dysregulated complement activation, often driven by persistence of activating triggers, is a cause of pathological inflammation in numerous diseases, including neurological diseases. Increasingly, this has become apparent not only in well-recognized neuroinflammatory diseases like multiple sclerosis but also in neurodegenerative and neuropsychiatric diseases where inflammation was previously either ignored or dismissed as a secondary event. There is now a large and rapidly growing body of evidence implicating complement in neurological diseases that cannot be comprehensively addressed in a brief review. Here, we will focus on neurodegenerative diseases, including not only the 'classical' neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, but also two other neurological diseases where neurodegeneration is a neglected feature and complement is implicated, namely, schizophrenia, a neurodevelopmental disorder with many mechanistic features of neurodegeneration, and multiple sclerosis, a demyelinating disorder where neurodegeneration is a major cause of progressive decline. We will discuss the evidence implicating complement as a driver of pathology in these diverse diseases and address briefly the potential and pitfalls of anti-complement drug therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Jacqui Nimmo
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Robert A.J. Byrne
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Lewis M. Watkins
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Sarah M. Carpanini
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Wioleta M. Zelek
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - B. Paul Morgan
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| |
Collapse
|
10
|
Gomez‐Arboledas A, Fonseca MI, Kramar E, Chu S, Schartz ND, Selvan P, Wood MA, Tenner AJ. C5aR1 signaling promotes region- and age-dependent synaptic pruning in models of Alzheimer's disease. Alzheimers Dement 2024; 20:2173-2190. [PMID: 38278523 PMCID: PMC10984438 DOI: 10.1002/alz.13682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Synaptic loss is a hallmark of Alzheimer's disease (AD) that correlates with cognitive decline in AD patients. Complement-mediated synaptic pruning has been associated with this excessive loss of synapses in AD. Here, we investigated the effect of C5aR1 inhibition on microglial and astroglial synaptic pruning in two mouse models of AD. METHODS A combination of super-resolution and confocal and tridimensional image reconstruction was used to assess the effect of genetic ablation or pharmacological inhibition of C5aR1 on the Arctic48 and Tg2576 models of AD. RESULTS Genetic ablation or pharmacological inhibition of C5aR1 partially rescues excessive pre-synaptic pruning and synaptic loss in an age and region-dependent fashion in two mouse models of AD, which correlates with improved long-term potentiation (LTP). DISCUSSION Reduction of excessive synaptic pruning is an additional beneficial outcome of the suppression of C5a-C5aR1 signaling, further supporting its potential as an effective targeted therapy to treat AD. HIGHLIGHTS C5aR1 ablation restores long-term potentiation in the Arctic model of AD. C5aR1 ablation rescues region specific excessive pre-synaptic loss. C5aR1 antagonist, PMX205, rescues VGlut1 loss in the Tg2576 model of AD. C1q tagging is not sufficient to induce VGlut1 microglial ingestion. Astrocytes contribute to excessive pre-synaptic loss at late stages of the disease.
Collapse
Affiliation(s)
- Angela Gomez‐Arboledas
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Maria I. Fonseca
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Enikö Kramar
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Shu‐Hui Chu
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Nicole D. Schartz
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Purnika Selvan
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | - Marcelo A. Wood
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - Andrea J. Tenner
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaSchool of MedicineIrvineCaliforniaUSA
| |
Collapse
|
11
|
Nording H, Baron L, Sauter M, Lübken A, Rawish E, Szepanowski R, von Esebeck J, Sun Y, Emami H, Meusel M, Saraei R, Schanze N, Gorantla SP, von Bubnoff N, Geisler T, von Hundelshausen P, Stellos K, Marquardt J, Sadik CD, Köhl J, Duerschmied D, Kleinschnitz C, Langer HF. Platelets regulate ischemia-induced revascularization and angiogenesis by secretion of growth factor-modulating factors. Blood Adv 2023; 7:6411-6427. [PMID: 37257194 PMCID: PMC10598500 DOI: 10.1182/bloodadvances.2021006891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 06/02/2023] Open
Abstract
In ischemic tissue, platelets can modulate angiogenesis. The specific factors influencing this function, however, are poorly understood. Here, we characterized the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) expressed on platelets as a potent regulator of ischemia-driven revascularization. We assessed the relevance of the anaphylatoxin receptor C5aR1 on platelets in patients with coronary artery disease as well as those with peripheral artery disease and used genetic mouse models to characterize its significance for ischemia and growth factor-driven revascularization. The presence of C5aR1-expressing platelets was increased in the hindlimb ischemia model. Ischemia-driven angiogenesis was significantly improved in C5aR1-/- mice but not in C5-/- mice, suggesting a specific role of C5aR1. Experiments using the supernatant of C5a-stimulated platelets suggested a paracrine mechanism of angiogenesis inhibition by platelets by means of antiangiogenic CXC chemokine ligand 4 (CXCL4, PF4). Lineage-specific C5aR1 deletion verified that the secretion of CXCL4 depends on C5aR1 ligation on platelets. Using C5aR1-/-CXCL4-/- mice, we observed no additional effect in the revascularization response, underscoring a strong dependence of CXCL4 secretion on the C5a-C5aR1-axis. We identified a novel mechanism for inhibition of neovascularization via platelet C5aR1, which was mediated by the release of antiangiogenic CXCL4.
Collapse
Affiliation(s)
- Henry Nording
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Lasse Baron
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Manuela Sauter
- Cardioimmunology Group, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Antje Lübken
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Elias Rawish
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Rebecca Szepanowski
- Department of Neurology and Center for Translational and Behavioral Neurosciences, University Hospital Essen, Essen, Germany
| | - Jacob von Esebeck
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Ying Sun
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Hossein Emami
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Moritz Meusel
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Roza Saraei
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Nancy Schanze
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sivahari Prasad Gorantla
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
| | - Tobias Geisler
- Department of Cardiovascular Medicine, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention, Ludwig Maximilians University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Konstantinos Stellos
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jens Marquardt
- First Department of Medicine, University of Schleswig-Holstein, Lübeck, Germany
| | | | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Schleswig-Holstein, Lübeck, Germany
| | - Daniel Duerschmied
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational and Behavioral Neurosciences, University Hospital Essen, Essen, Germany
| | - Harald F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
- Cardioimmunology Group, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
12
|
Schartz ND, Liang HY, Carvalho K, Chu SH, Mendoza-Arvilla A, Petrisko TJ, Gomez-Arboledas A, Mortazavi A, Tenner AJ. C5aR1 antagonism suppresses inflammatory glial gene expression and alters cellular signaling in an aggressive Alzheimer's model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554306. [PMID: 37662399 PMCID: PMC10473603 DOI: 10.1101/2023.08.22.554306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults, and the need for effective, sustainable therapeutic targets is imperative. Pharmacologic inhibition of C5aR1 reduces plaque load, gliosis and memory deficits in animal models. However, the cellular basis underlying this neuroprotection and which processes were the consequence of amyloid reduction vs alteration of the response to amyloid were unclear. In the Arctic model, the C5aR1 antagonist PMX205 did not reduce plaque load, but deficits in short-term memory in female mice were prevented. Hippocampal single cell and single nucleus RNA-seq clusters revealed C5aR1 dependent and independent gene expression and cell-cell communication. Microglial clusters containing neurotoxic disease-associated microglial genes were robustly upregulated in Arctic mice and drastically reduced with PMX205 treatment, while genes in microglia clusters that were overrepresented in the Arctic-PMX205 vs Arctic group were associated with synapse organization and transmission and learning. PMX205 treatment also reduced some A-1 astrocyte genes. In spite of changes in transcript levels, overall protein levels of some reactive glial markers were relatively unchanged by C5aR1 antagonism, as were clusters associated with protective responses to injury. C5aR1 inhibition promoted signaling pathways associated with cell growth and repair, such as TGFβ and FGF, in Arctic mice, while suppressing inflammatory pathways including PROS, Pecam1, and EPHA. In conclusion, pharmacologic C5aR1 inhibition prevents cognitive loss, limits microglial polarization to a detrimental inflammatory state and permits neuroprotective responses, as well as leaving protective functions of complement intact, making C5aR1 antagonism an attractive therapeutic strategy for individuals with AD.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Heidi Y. Liang
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697
| | - Klebea Carvalho
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697
| | - Shu-Hui Chu
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Adrian Mendoza-Arvilla
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Tiffany J. Petrisko
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Angela Gomez-Arboledas
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
| | - Ali Mortazavi
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697
| | - Andrea J. Tenner
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA 92697
| |
Collapse
|
13
|
Wang L, Sun Y, Kong F, Jiang Y, An M, Jin B, Cao D, Li R, Guan X, Liang S, Abudurexiti S, Gong P. Mild Hypothermia Alleviates Complement C5a-Induced Neuronal Autophagy During Brain Ischemia-Reperfusion Injury After Cardiac Arrest. Cell Mol Neurobiol 2023; 43:1957-1974. [PMID: 36006573 PMCID: PMC11412180 DOI: 10.1007/s10571-022-01275-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/17/2022] [Indexed: 02/06/2023]
Abstract
After restoration of spontaneous circulation (ROSC) following cardiac arrest, complements can be activated and excessive autophagy can contribute to the brain ischemia-reperfusion (I/R) injury. Mild hypothermia (HT) protects against brain I/R injury after ROSC, but the mechanisms have not been fully elucidated. Here, we found that HT significantly inhibited the increases in serum NSE, S100β, and C5a, as well as neurologic deficit scores, TUNEL-positive cells, and autophagic vacuoles in the pig brain cortex after ROSC. The C5a receptor 1 (C5aR1) mRNA and the C5a, C5aR1, Beclin 1, LC3-II, and cleaved caspase-3 proteins were significantly increased, but the P62 protein and the PI3K/Akt/mTOR pathway-related proteins were significantly reduced in pigs after ROSC or neuronal oxygen-glucose deprivation/reoxygenation. HT could significantly attenuate the above changes in NT-treated neurons. Furthermore, C5a treatment induced autophagy and apoptosis and reduced the PI3K/Akt/mTOR pathway-related proteins in cultured neurons, which could be reversed by C5aR1 antagonist PMX205. Our findings demonstrated that C5a could bind to C5aR1 to induce neuronal autophagy during the brain I/R injury, which was associated with the inhibited PI3K/Akt/mTOR pathway. HT could inhibit C5a-induced neuronal autophagy by regulating the C5a-C5aR1 interaction and the PI3K/Akt/mTOR pathway, which might be one of the neuroprotective mechanisms underlying I/R injury. The C5a receptor 1 (C5aR1) mRNA and the C5a, C5aR1, Beclin 1, LC3-II, and cleaved caspase-3 proteins were significantly increased, but the P62 protein and the PI3K/Akt/mTOR pathway-related proteins were significantly reduced in pigs after ROSC or neuronal oxygen-glucose deprivation/reoxygenation. Mild hypothermia (HT) could significantly attenuate the above changes in NT-treated neurons. Furthermore, C5a treatment induced autophagy and apoptosis and reduced the PI3K/Akt/mTOR pathway-related proteins in cultured neurons, which could be reversed by C5aR1 antagonist PMX205. Proposed mechanism by which HT protects against brain I/R injury by repressing C5a-C5aR1-induced excessive autophagy. Complement activation in response to brain I/R injury generates C5a that can interact with C5aR1 to inactivate mTOR, probably through the PI3K-AKT pathway, which can finally lead to autophagy activation. The excessively activated autophagy ultimately contributes to cell apoptosis and brain injury. HT may alleviate complement activation and then reduce C5a-induced autophagy to protect against brain I/R injury. HT, mild hypothermia; I/R, ischemia reperfusion.
Collapse
Affiliation(s)
- Ling Wang
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
- Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Yuanyuan Sun
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Fang Kong
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Yi Jiang
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Mengmeng An
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Beibei Jin
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Da Cao
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Ruifang Li
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Xiaolan Guan
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Shuangshuang Liang
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Subi Abudurexiti
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Ping Gong
- Department of Emergency Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| |
Collapse
|
14
|
Toutonji A, Krieg C, Borucki DM, Mandava M, Guglietta S, Tomlinson S. Mass cytometric analysis of the immune cell landscape after traumatic brain injury elucidates the role of complement and complement receptors in neurologic outcomes. Acta Neuropathol Commun 2023; 11:92. [PMID: 37308987 DOI: 10.1186/s40478-023-01583-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/18/2023] [Indexed: 06/14/2023] Open
Abstract
Following traumatic brain injury (TBI), a neuroinflammatory response can persist for years and contribute to the development of chronic neurological manifestations. Complement plays a central role in post-TBI neuroinflammation, and C3 opsonins and the anaphylatoxins (C3a and C5a) have been implicated in promoting secondary injury. We used single cell mass cytometry to characterize the immune cell landscape of the brain at different time points after TBI. To specifically investigate how complement shapes the post-TBI immune cell landscape, we analyzed TBI brains in the context of CR2-Crry treatment, an inhibitor of C3 activation. We analyzed 13 immune cell types, including peripheral and brain resident cells, and assessed expression of various receptors. TBI modulated the expression of phagocytic and complement receptors on both brain resident and infiltrating peripheral immune cells, and distinct functional clusters were identified within same cell populations that emerge at different phases after TBI. In particular, a CD11c+ (CR4) microglia subpopulation continued to expand over 28 days after injury, and was the only receptor to show continuous increase over time. Complement inhibition affected the abundance of brain resident immune cells in the injured hemisphere and impacted the expression of functional receptors on infiltrating cells. A role for C5a has also been indicated in models of brain injury, and we found significant upregulation of C5aR1 on many immune cell types after TBI. However, we demonstrated experimentally that while C5aR1 is involved in the infiltration of peripheral immune cells into the brain after injury, it does not alone affect histological or behavioral outcomes. However, CR2-Crry improved post-TBI outcomes and reduced resident immune cell populations, as well as complement and phagocytic receptor expression, indicating that its neuroprotective effects are mediated upstream of C5a generation, likely via modulating C3 opsonization and complement receptor expression.
Collapse
Affiliation(s)
- Amer Toutonji
- College of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Carsten Krieg
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Hollings Cancer Center, Charleston, SC, 29425, USA
| | - Davis M Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Mamatha Mandava
- Immune Deficiency Cellular Therapy Program (IDCTP), National Institutes of Health, Bethesda, USA
| | - Silvia Guglietta
- Hollings Cancer Center, Charleston, SC, 29425, USA.
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Ralph Johnson VA Medical Center, Charleston, SC, 29401, USA.
| |
Collapse
|
15
|
Bohlson SS, Tenner AJ. Complement in the Brain: Contributions to Neuroprotection, Neuronal Plasticity, and Neuroinflammation. Annu Rev Immunol 2023; 41:431-452. [PMID: 36750318 DOI: 10.1146/annurev-immunol-101921-035639] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The complement system is an ancient collection of proteolytic cascades with well-described roles in regulation of innate and adaptive immunity. With the convergence of a revolution in complement-directed clinical therapeutics, the discovery of specific complement-associated targetable pathways in the central nervous system, and the development of integrated multi-omic technologies that have all emerged over the last 15 years, precision therapeutic targeting in Alzheimer disease and other neurodegenerative diseases and processes appears to be within reach. As a sensor of tissue distress, the complement system protects the brain from microbial challenge as well as the accumulation of dead and/or damaged molecules and cells. Additional more recently discovered diverse functions of complement make it of paramount importance to design complement-directed neurotherapeutics such that the beneficial roles in neurodevelopment, adult neural plasticity, and neuroprotective functions of the complement system are retained.
Collapse
Affiliation(s)
- Suzanne S Bohlson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA; ,
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA; ,
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, California, USA
| |
Collapse
|
16
|
Evans R, Watkins LM, Hawkins K, Santiago G, Demetriou C, Naughton M, Dittmer M, Rees MI, Fitzgerald D, Morgan BP, Neal JW, Howell OW. Complement activation and increased anaphylatoxin receptor expression are associated with cortical grey matter lesions and the compartmentalised inflammatory response of multiple sclerosis. Front Cell Neurosci 2023; 17:1094106. [PMID: 37032838 PMCID: PMC10073739 DOI: 10.3389/fncel.2023.1094106] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/07/2023] [Indexed: 04/11/2023] Open
Abstract
Background The extent of cortical pathology is an important determinant of multiple sclerosis (MS) severity. Cortical demyelination and neurodegeneration are related to inflammation of the overlying leptomeninges, a more inflammatory CSF milieu and with parenchymal microglia and astroglia activation. These are all components of the compartmentalised inflammatory response. Compartmentalised inflammation is a feature of progressive MS, which is not targeted by disease modifying therapies. Complement is differentially expressed in the MS CSF and complement, and complement receptors, are associated with demyelination and neurodegeneration. Methods To better understand if complement activation in the leptomeninges is associated with underlying cortical demyelination, inflammation, and microglial activation, we performed a neuropathological study of progressive MS (n = 22, 14 females), neuroinflammatory (n = 8), and non-neurological disease controls (n = 10). We then quantified the relative extent of demyelination, connective tissue inflammation, complement, and complement receptor positive microglia/macrophages. Results Complement was elevated at the leptomeninges, subpial, and within and around vessels of the cortical grey matter. The extent of complement C1q immunoreactivity correlated with connective tissue infiltrates, whilst activation products C4d, Bb, and C3b associated with grey matter demyelination, and C3a receptor 1+ and C5a receptor 1+ microglia/macrophages closely apposed C3b labelled cells. The density of C3a receptor 1+ and C5a receptor 1+ cells was increased at the expanding edge of subpial and leukocortical lesions. C5a receptor 1+ cells expressed TNFα, iNOS and contained puncta immunoreactive for proteolipid protein, neurofilament and synaptophysin, suggesting their involvement in grey matter lesion expansion. Interpretation The presence of products of complement activation at the brain surfaces, their association with the extent of underlying pathology and increased complement anaphylatoxin receptor positive microglia/macrophages at expanding cortical grey matter lesions, could represent a target to modify compartmentalised inflammation and cortical demyelination.
Collapse
Affiliation(s)
- Rhian Evans
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis M. Watkins
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Kristen Hawkins
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Gabriella Santiago
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Constantinos Demetriou
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Michelle Naughton
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Marie Dittmer
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Mark I. Rees
- Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| | - Denise Fitzgerald
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - B. Paul Morgan
- School of Medicine, UK Dementia Research Institute Cardiff and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - James W. Neal
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Owain W. Howell
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Owain W. Howell,
| |
Collapse
|
17
|
Shu J, Ren Y, Tan W, Wei W, Zhang L, Chang J. Identification of potential drug targets for vascular dementia and carotid plaques by analyzing underlying molecular signatures shared by them. Front Aging Neurosci 2022; 14:967146. [PMID: 36262886 PMCID: PMC9574221 DOI: 10.3389/fnagi.2022.967146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/05/2022] [Indexed: 11/14/2022] Open
Abstract
Background Vascular dementia (VaD) and carotid atherosclerotic plaques are common in the elderly population, conferring a heavy burden on families and society. Accumulating evidence indicates carotid atherosclerotic plaques to be a risk factor for VaD. However, the underlying mechanisms for this association are mainly unknown. Materials and methods We analyzed temporal cortex gene expression data of the GSE122063 dataset and gene expression data of the GSE163154 dataset to identify commonly differentially expressed genes (DEGs). Then we performed functional enrichment analysis, immune cell infiltration and evaluation, correlation analysis between differentially expressed immune-related genes (DEIRGs) and immune cells, receiver operating characteristic (ROC) analysis, and drug-gene analysis. Results We identified 41 overlapped DEGs between the VaD and carotid atherosclerosis plaque datasets. Functional enrichment analyses revealed that these overlapped DEGs were mainly enriched in inflammatory and immune-related processes. Immunocyte infiltration and evaluation results showed that M0 macrophages, M2 macrophages, and T cells gamma delta had a dominant abundance in carotid atherosclerosis plaque samples, and M0 macrophages showed a significantly different infiltration percentage between the early and advanced stage plaques group. Resting CD4 memory T cells, M2 macrophages, and naive B cells were the top three highest infiltrating fractions in VaD. Furthermore, B cells and NK cells showed a different infiltration percentage between VaD and matched controls. We identified 12 DEIRGs, and the result of correlation analysis revealed that these DEIRGs were closely related to differentially expressed immune cells. We identified five key DEIRGs based on ROC analysis. The drug-gene interaction analysis showed that four drugs (avacopan, CCX354, BMS-817399, and ASK-8007) could be potential drugs for VaD and carotid atherosclerotic plaques treatment. Conclusion Collectively, these findings indicated that inflammatory and immune-related processes be a crucial common pathophysiological mechanism shared by VaD and carotid plaques. This study might provide new insights into common molecular mechanisms between VaD and carotid plaques and potential targets for the treatment.
Collapse
Affiliation(s)
- Jun Shu
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yiqing Ren
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Wen Tan
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Wenshi Wei
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- *Correspondence: Wenshi Wei,
| | - Li Zhang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Li Zhang,
| | - Jie Chang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Jie Chang,
| |
Collapse
|
18
|
Carvalho K, Schartz ND, Balderrama-Gutierrez G, Liang HY, Chu SH, Selvan P, Gomez-Arboledas A, Petrisko TJ, Fonseca MI, Mortazavi A, Tenner AJ. Modulation of C5a-C5aR1 signaling alters the dynamics of AD progression. J Neuroinflammation 2022; 19:178. [PMID: 35820938 PMCID: PMC9277945 DOI: 10.1186/s12974-022-02539-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 06/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The complement system is part of the innate immune system that clears pathogens and cellular debris. In the healthy brain, complement influences neurodevelopment and neurogenesis, synaptic pruning, clearance of neuronal blebs, recruitment of phagocytes, and protects from pathogens. However, excessive downstream complement activation that leads to generation of C5a, and C5a engagement with its receptor C5aR1, instigates a feed-forward loop of inflammation, injury, and neuronal death, making C5aR1 a potential therapeutic target for neuroinflammatory disorders. C5aR1 ablation in the Arctic (Arc) model of Alzheimer's disease protects against cognitive decline and neuronal injury without altering amyloid plaque accumulation. METHODS To elucidate the effects of C5a-C5aR1 signaling on AD pathology, we crossed Arc mice with a C5a-overexpressing mouse (ArcC5a+) and tested hippocampal memory. RNA-seq was performed on hippocampus and cortex from Arc, ArcC5aR1KO, and ArcC5a+ mice at 2.7-10 months and age-matched controls to assess mechanisms involved in each system. Immunohistochemistry was used to probe for protein markers of microglia and astrocytes activation states. RESULTS ArcC5a+ mice had accelerated cognitive decline compared to Arc. Deletion of C5ar1 delayed or prevented the expression of some, but not all, AD-associated genes in the hippocampus and a subset of pan-reactive and A1 reactive astrocyte genes, indicating a separation between genes induced by amyloid plaques alone and those influenced by C5a-C5aR1 signaling. Biological processes associated with AD and AD mouse models, including inflammatory signaling, microglial cell activation, and astrocyte migration, were delayed in the ArcC5aR1KO hippocampus. Interestingly, C5a overexpression also delayed the increase of some AD-, complement-, and astrocyte-associated genes, suggesting the possible involvement of neuroprotective C5aR2. However, these pathways were enhanced in older ArcC5a+ mice compared to Arc. Immunohistochemistry confirmed that C5a-C5aR1 modulation in Arc mice delayed the increase in CD11c-positive microglia, while not affecting other pan-reactive microglial or astrocyte markers. CONCLUSION C5a-C5aR1 signaling in AD largely exerts its effects by enhancing microglial activation pathways that accelerate disease progression. While C5a may have neuroprotective effects via C5aR2, engagement of C5a with C5aR1 is detrimental in AD models. These data support specific pharmacological inhibition of C5aR1 as a potential therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Klebea Carvalho
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697 USA
| | - Nicole D. Schartz
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
| | | | - Heidi Y. Liang
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697 USA
| | - Shu-Hui Chu
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
| | - Purnika Selvan
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
| | - Angela Gomez-Arboledas
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
| | - Tiffany J. Petrisko
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
| | - Maria I. Fonseca
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
| | - Ali Mortazavi
- Department of Developmental & Cell Biology, University of California, Irvine, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology & Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92697-3900 USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA USA
| |
Collapse
|
19
|
Parker SE, Bellingham MC, Woodruff TM. Complement drives circuit modulation in the adult brain. Prog Neurobiol 2022; 214:102282. [DOI: 10.1016/j.pneurobio.2022.102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/24/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022]
|
20
|
Zhang J, Zhou R, Cao G, Zhang Y, Xu H, Yang H. Guhong Injection Prevents Ischemic Stroke-Induced Neuro-Inflammation and Neuron Loss Through Regulation of C5ar1. Front Pharmacol 2022; 13:818245. [PMID: 35387346 PMCID: PMC8979065 DOI: 10.3389/fphar.2022.818245] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
C5ar1 (CD88) has been identified as an important potential therapeutic target for regulating inflammation in ischemic stroke. In this study, the neuroprotective effect of Guhong injection (GHI) on middle cerebral artery occlusion (MCAO)-induced reperfusion injury was assessed and the mechanism was explored by RNA-seq technology. GHI administered for 6 consecutive days significantly decreased body weight loss, infarction rate, neurological deficient scores, and neuron loss but improved rat survival percentage and regional cerebral blood flow after MCAO surgery. Furthermore, we identified inflammation as a vital process and C5AR1 as a vital target in GHI-mediated protection by using RNA-seq analysis. Further experiments confirmed that GHI decreased C5AR1, C5A, CASP3, 8-OHdG, and inflammatory factors such as IL-1β, TNF, IL6, ICAM-1, MMP9, and MCP-1, and enhanced the expression of TIMP1, JAM-A, and laminin. Furthermore, GHI and its major components hydroxysafflower yellow A (HSYA) and aceglutamide (AG) enhanced cell viability and reduced LDH level and C5AR1 expression in a C5A-induced Neuro-2a cell damage model. In general, this study elucidated the mechanism of GHI against ischemic stroke by inhibiting inflammation and highlighted the potential important role of C5AR1 in ischemic stroke. This research provided new insights into the mechanism of GHI in resisting ischemic stroke and benefits of its clinical application.
Collapse
Affiliation(s)
- Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| | - Rui Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangzhao Cao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - He Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Shibata R, Nagoshi N, Kajikawa K, Ito S, Shibata S, Shindo T, Khazaei M, Nori S, Kohyama J, Fehlings MG, Matsumoto M, Nakamura M, Okano H. Administration of C5a receptor antagonist improves the efficacy of human iPSCs-derived NS/PC transplantation in the acute phase of spinal cord injury. J Neurotrauma 2022; 39:667-682. [PMID: 35196890 DOI: 10.1089/neu.2021.0225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Human-induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PCs) transplantation during the acute phase of spinal cord injury (SCI) is not effective due to the inflammatory response occurring immediately after SCI, which negatively impacts transplanted cell survival. Therefore, we chose to study the powerful chemoattractant complement C5a as a method to generate a more favorable transplantation environment. We hypothesized that suppression of the inflammatory response immediately after SCI by C5a receptor antagonist (C5aRA) would improve the efficacy of hiPSC-NS/PCs transplantation for acute phase SCI. Here, we evaluated the influence of C5aRA on the inflammatory reaction during the acute phase after SCI, and observed significant reductions in several inflammatory cytokines, macrophages, neutrophils and apoptotic markers. Next, we divided the SCI mice into 4 groups: i) Phosphate-buffered saline (PBS) only, ii) C5aRA only, iii) PBS + transplantation (PBS+TP), and iv) C5aRA + transplantation (C5aRA+TP). Immediately after SCI, C5aRA or PBS was injected once a day for 4 consecutive days, followed by hiPSC-NS/PC transplantation or PBS into the lesion epicenter on day 4. The C5aRA+TP group had better functional improvement as compared to the PBS only group. The C5aRA+TP group also had a significantly higher cell survival rate compared to the PBS+TP group. This study demonstrates that administration of C5aRA can suppress the inflammatory response during the acute phase of SCI, while improving the survival rate of transplanted hiPSC-NS/PCs as well as enhancing motor functional restoration. hiPSC-NS/PC transplantation with C5aRA is a promising treatment during the acute injury phase for SCI patients.
Collapse
Affiliation(s)
- Reo Shibata
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan.,Keio University School of Medicine, Physiology, Shinjuku-ku, Japan;
| | - Narihito Nagoshi
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan;
| | - Keita Kajikawa
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan;
| | - Shuhei Ito
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan;
| | - Shinsuke Shibata
- Keio University School of Medicine, Electron Microscope Laboratory, Shinjuku-ku, Tokyo, Japan.,Graduate School of Medical and Dental Sciences, Niigata University, Division of Microscopic Anatomy, Niigata, Japan;
| | - Tomoko Shindo
- Keio University School of Medicine, Electron Microscope Laboratory, Shinjuku-ku, Tokyo, Japan;
| | - Mohamad Khazaei
- University Health Network, Division of Genetics and Development, Toronto Western Research Institute, Krembil Neuroscience Program, Toronto, Ontario, Canada;
| | - Satoshi Nori
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan;
| | - Jun Kohyama
- Keio University School of Medicine, Physiology, Shinjuku-ku, Japan;
| | - Michael G Fehlings
- University Health Network, Division of Genetics and Development, Toronto Western Research Institute, Krembil Neuroscience Program, Toronto, Ontario, Canada;
| | - Morio Matsumoto
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan;
| | - Masaya Nakamura
- Keio University School of Medicine, Orthopaedics Surgery, Shinjuku-ku, Japan;
| | - Hideyuki Okano
- Keio University School of Medicine, Physiology, Shinjuku-ku, Japan;
| |
Collapse
|
22
|
Complement as a powerful "influencer" in the brain during development, adulthood and neurological disorders. Adv Immunol 2021; 152:157-222. [PMID: 34844709 DOI: 10.1016/bs.ai.2021.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The complement system was long considered as only a powerful effector arm of the immune system that, while critically protective, could lead to inflammation and cell death if overactivated, even in the central nervous system (CNS). However, in the past decade it has been recognized as playing critical roles in key physiological processes in the CNS, including neurogenesis and synaptic remodeling in the developing and adult brain. Inherent in these processes are the interactions with cells in the brain, and the cascade of interactions and functional consequences that ensue. As a result, investigations of therapeutic approaches for both suppressing excessive complement driven neurotoxicity and aberrant sculpting of neuronal circuits, require broad (and deep) knowledge of the functional activities of multiple components of this highly evolved and regulated system to avoid unintended negative consequences in the clinic. Advances in basic science are beginning to provide a roadmap for translation to therapeutics, with both small molecule and biologics. Here, we present examples of the critical roles of proper complement function in the development and sculpting of the nervous system, and in enabling rapid protection from infection and clearance of dying cells. Microglia are highlighted as important command centers that integrate signals from the complement system and other innate sensors that are programed to provide support and protection, but that direct detrimental responses to aberrant activation and/or regulation of the system. Finally, we present promising research areas that may lead to effective and precision strategies for complement targeted interventions to promote neurological health.
Collapse
|
23
|
Shi Y, Jin Y, Li X, Chen C, Zhang Z, Liu X, Deng Y, Fan X, Wang C. C5aR1 Mediates the Progression of Inflammatory Responses in the Brain of Rats in the Early Stage after Ischemia and Reperfusion. ACS Chem Neurosci 2021; 12:3994-4006. [PMID: 34637270 DOI: 10.1021/acschemneuro.1c00244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
C5a receptor 1 (C5aR1) can induce a strong inflammatory response to an injury. Targeting C5aR1 has emerged as a novel anti-inflammatory therapeutic method. However, the role of C5aR1 in cerebral ischemia and reperfusion (I/R) injury and the definitive mechanism have not been elucidated clearly. Here, we determined whether C5aR1 signaling was essential to the post-ischemic inflammation and brain injury and whether it is a valid target for therapeutic blockade by using soluble receptor antagonist PMX53 in the early stage after I/R injury. In an in vitro model (oxygen and glucose deprivation and reperfusion, OGD/R) and in vivo model (middle cerebral artery occlusion and reperfusion, MCAO/R) of I/R, the neuronal cells of rats showed significantly up-regulated gene expression of C5aR1, and a notable inflammatory response was demonstrated with elevated tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6. Inhibition of C5aR1 by PMX53 treatment significantly reduced cell injury and inflammation and promoted brain function recovery. Further mechanism studies showed that inhibiting C5aR1 by PMX53 protected the rats from MCAO/R injury, decreased cell inflammation, and apoptosis via inhibiting the TLR4 and NF-κB signaling pathway and reducing the production of TNF-α, IL-1β, and IL-6 in MCAO/R rats. In addition, manipulation of the C5aR1 gene expression in vitro displayed that the inflammatory cascade signals including TLR4, TNF-α, IL-1β, and IL-6 were coincidently regulated with the regulation of C5aR1 expression levels. Thus, our results demonstrated a pathogenic role for C5aR1 in the progression of brain injury and inflammation response following I/R injury. Our study clearly demonstrated that C5aR1 inhibition might be an effective treatment strategy for ischemic stroke.
Collapse
Affiliation(s)
- Yunwei Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, Jiangsu, China
| | - Ying Jin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, Jiangsu, China
| | - Xing Li
- Department of ICU, No. 1 People’s Hospital of Yancheng City, The Fourth Affiliated Hospital of Nantong University, Yancheng 224000, Jiangsu, China
| | - Chen Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, Jiangsu, China
| | - Zhihong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, Jiangsu, China
| | - Yijun Deng
- Department of ICU, No. 1 People’s Hospital of Yancheng City, The Fourth Affiliated Hospital of Nantong University, Yancheng 224000, Jiangsu, China
| | - Xingjuan Fan
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
24
|
Gomez-Arboledas A, Acharya MM, Tenner AJ. The Role of Complement in Synaptic Pruning and Neurodegeneration. Immunotargets Ther 2021; 10:373-386. [PMID: 34595138 PMCID: PMC8478425 DOI: 10.2147/itt.s305420] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
The complement system, an essential part of the innate immune system, is composed of a group of secreted and membrane proteins that collectively participate in maintaining the function of the healthy and diseased brain. However, an inappropriate activation of the complement system has been related to an inflammatory response in multiple diseases, such as stroke, traumatic brain injury, multiple sclerosis, and Alzheimer's disease, as well as Zika infection and radiotherapy. In addition, C1q and C3 (initial activation components of the complement cascade) have been shown to play a key beneficial role in the refinement of synaptic circuits during developmental stages and adult plasticity. Nevertheless, excessive synaptic pruning in the adult brain can be detrimental and has been associated with synaptic loss in several pathological conditions. In this brief review, we will discuss the role of the complement system in synaptic pruning as well as its contribution to neurodegeneration and cognitive deficits. We also mention potential therapeutic approaches to target the complement system to treat several neuroinflammatory diseases and unintended consequences of radiotherapy.
Collapse
Affiliation(s)
- Angela Gomez-Arboledas
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
25
|
The complement cascade in the regulation of neuroinflammation, nociceptive sensitization, and pain. J Biol Chem 2021; 297:101085. [PMID: 34411562 PMCID: PMC8446806 DOI: 10.1016/j.jbc.2021.101085] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 01/13/2023] Open
Abstract
The complement cascade is a key component of the innate immune system that is rapidly recruited through a cascade of enzymatic reactions to enable the recognition and clearance of pathogens and promote tissue repair. Despite its well-understood role in immunology, recent studies have highlighted new and unexpected roles of the complement cascade in neuroimmune interaction and in the regulation of neuronal processes during development, aging, and in disease states. Complement signaling is particularly important in directing neuronal responses to tissue injury, neurotrauma, and nerve lesions. Under physiological conditions, complement-dependent changes in neuronal excitability, synaptic strength, and neurite remodeling promote nerve regeneration, tissue repair, and healing. However, in a variety of pathologies, dysregulation of the complement cascade leads to chronic inflammation, persistent pain, and neural dysfunction. This review describes recent advances in our understanding of the multifaceted cross-communication that takes place between the complement system and neurons. In particular, we focus on the molecular and cellular mechanisms through which complement signaling regulates neuronal excitability and synaptic plasticity in the nociceptive pathways involved in pain processing in both health and disease. Finally, we discuss the future of this rapidly growing field and what we believe to be the significant knowledge gaps that need to be addressed.
Collapse
|
26
|
Complement C4 Is Reduced in iPSC-Derived Astrocytes of Autism Spectrum Disorder Subjects. Int J Mol Sci 2021; 22:ijms22147579. [PMID: 34299197 PMCID: PMC8305914 DOI: 10.3390/ijms22147579] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
In recent years, accumulating evidence has shown that the innate immune complement system is involved in several aspects of normal brain development and in neurodevelopmental disorders, including autism spectrum disorder (ASD). Although abnormal expression of complement components was observed in post-mortem brain samples from individuals with ASD, little is known about the expression patterns of complement molecules in distinct cell types in the developing autistic brain. In the present study, we characterized the mRNA and protein expression profiles of a wide range of complement system components, receptors and regulators in induced pluripotent stem cell (iPSC)-derived neural progenitor cells, neurons and astrocytes of individuals with ASD and neurotypical controls, which constitute in vitro cellular models that recapitulate certain features of both human brain development and ASD pathophysiology. We observed that all the analyzed cell lines constitutively express several key complement molecules. Interestingly, using different quantification strategies, we found that complement C4 mRNA and protein are expressed in significantly lower levels by astrocytes derived from ASD individuals compared to control astrocytes. As astrocytes participate in synapse elimination, and diminished C4 levels have been linked to defective synaptic pruning, our findings may contribute to an increased understanding of the atypically enhanced brain connectivity in ASD.
Collapse
|
27
|
Garred P, Tenner AJ, Mollnes TE. Therapeutic Targeting of the Complement System: From Rare Diseases to Pandemics. Pharmacol Rev 2021; 73:792-827. [PMID: 33687995 PMCID: PMC7956994 DOI: 10.1124/pharmrev.120.000072] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.
Collapse
Affiliation(s)
- Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Andrea J Tenner
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| | - Tom E Mollnes
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (P.G.); Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, California (A.J.T.); and Research Laboratory, Nordland Hospital, Bodø, Norway, Faculty of Health Sciences, K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway (T.E.M.); Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (T.E.M.); and Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway (T.E.M.)
| |
Collapse
|
28
|
Keragala CB, Woodruff TM, Liu Z, Niego B, Ho H, McQuilten Z, Medcalf RL. Tissue-Type Plasminogen Activator and Tenecteplase-Mediated Increase in Blood Brain Barrier Permeability Involves Cell Intrinsic Complement. Front Neurol 2020; 11:577272. [PMID: 33363504 PMCID: PMC7753024 DOI: 10.3389/fneur.2020.577272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/09/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Tissue-type plasminogen activator (t-PA) has been the mainstay of therapeutic thrombolysis for patients with acute ischaemic stroke (AIS). However, t-PA can cause devastating intracerebral hemorrhage. t-PA can also influence the CNS in part by modulation of BBB permeability. Complement activation also occurs after AIS and has also been reported to increase BBB permeability. The complement components, C3 and C5, can also be activated by t-PA via plasmin formation and cell intrinsic complement may be involved in this process. Tenecteplase (TNK-tPA) is a t-PA variant with a longer plasma half-life, yet the ability of TNK-tPA to modulate the BBB and complement is less clear. Aim: To evaluate the effect of C5 and C5a-receptor 1 (C5aR1) inhibitors on t-PA- and TNK-tPA-mediated opening of the BBB. Methods: We used an in vitro model of the BBB where human brain endothelial cells and human astrocytes were co-cultured on the opposite sides of a porous membrane assembled in transwell inserts. The luminal (endothelial) compartment was stimulated with t-PA or TNK-tPA together with plasminogen, in the presence of PMX205 (a non-competitive C5aR1 antagonist), Avacopan (a competitive C5aR1 antagonist) or Eculizumab (a humanized monoclonal inhibitor of human C5). BBB permeability was assessed 5 and 24 h later. Immunofluorescence was also used to detect changes in C5 and C5aR1 expression in endothelial cells and astrocytes. Results: PMX205, but not Avacopan or Eculizumab, blocked t-PA-mediated increase in BBB permeability at both the 5 and 24 h time points. PMX205 also blocked TNK-tPA-mediated increase in BBB permeability. Immunofluorescence analysis revealed intracellular staining of C5 in both cell types. C5aR1 expression was also detected on the cell surfaces and also located intracellularly in both cell types. Conclusion: t-PA and TNK-tPA-mediated increase in BBB permeability involves C5aR1 receptor activation from cell-derived C5a. Selective inhibitors of C5aR1 may have therapeutic potential in AIS.
Collapse
Affiliation(s)
- Charithani B Keragala
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Zikou Liu
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Be'eri Niego
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Heidi Ho
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Zoe McQuilten
- Transfusion Research Unit, Department of Epidemiology and Preventative Medicine, Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia
| | - Robert L Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
29
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|
30
|
Gavriilaki M, Kimiskidis VK, Gavriilaki E. Precision Medicine in Neurology: The Inspirational Paradigm of Complement Therapeutics. Pharmaceuticals (Basel) 2020; 13:E341. [PMID: 33114553 PMCID: PMC7693884 DOI: 10.3390/ph13110341] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Precision medicine has emerged as a central element of healthcare science. Complement, a component of innate immunity known for centuries, has been implicated in the pathophysiology of numerous incurable neurological diseases, emerging as a potential therapeutic target and predictive biomarker. In parallel, the innovative application of the first complement inhibitor in clinical practice as an approved treatment of myasthenia gravis (MG) and neuromyelitis optica spectrum disorders (NMOSD) related with specific antibodies raised hope for the implementation of personalized therapies in detrimental neurological diseases. A thorough literature search was conducted through May 2020 at MEDLINE, EMBASE, Cochrane Library and ClinicalTrials.gov databases based on medical terms (MeSH)" complement system proteins" and "neurologic disease". Complement's role in pathophysiology, monitoring of disease activity and therapy has been investigated in MG, multiple sclerosis, NMOSD, spinal muscular atrophy, amyotrophic lateral sclerosis, Parkinson, Alzheimer, Huntington disease, Guillain-Barré syndrome, chronic inflammatory demyelinating polyneuropathy, stroke, and epilepsy. Given the complexity of complement diagnostics and therapeutics, this state-of-the-art review aims to provide a brief description of the complement system for the neurologist, an overview of novel complement inhibitors and updates of complement studies in a wide range of neurological disorders.
Collapse
Affiliation(s)
- Maria Gavriilaki
- Postgraduate Course, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasilios K. Kimiskidis
- Postgraduate Course, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, 57010 Thessaloniki, Greece;
| |
Collapse
|
31
|
Gravina G, Svedin P, Ardalan M, Levy O, Ek CJ, Mallard C, Lai JCY. Staphylococcus epidermidis Sensitizes Perinatal Hypoxic-Ischemic Brain Injury in Male but Not Female Mice. Front Immunol 2020; 11:516. [PMID: 32373108 PMCID: PMC7186320 DOI: 10.3389/fimmu.2020.00516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Staphylococcus epidermidis is the most common nosocomial infection and the predominant pathogen in late-onset sepsis in preterm infants. Infection and inflammation are linked to neurological and developmental sequelae and bacterial infections increase the vulnerability of the brain to hypoxia-ischemia (HI). We thus tested the hypothesis that S. epidermidis exacerbates HI neuropathology in neonatal mice. Methods: Male and female C57Bl/6 mice were injected intraperitoneally with sterile saline or 3.5 × 107 colony-forming units of S. epidermidis on postnatal day (PND) 4 and then subjected to HI on PND5 (24 h after injection) or PND9 (5 d after injection) by left carotid artery ligation and exposure to 10% O2. White and gray matter injury was assessed on PND14-16. In an additional group of animals, the plasma, brain, and liver were collected on PND5 or PND9 after infection to evaluate cytokine and chemokine profiles, C5a levels and C5 signaling. Results: HI induced 24 h after injection of S. epidermidis resulted in greater gray and white matter injury compared to saline injected controls in males, but not in females. Specifically, males demonstrated increased gray matter injury in the cortex and striatum, and white matter loss in the subcortical region, hippocampal fimbria and striatum. In contrast, there was no potentiation of brain injury when HI occurred 5 d after infection in either sex. In the plasma, S. epidermidis-injected mice demonstrated increased levels of pro- and anti-inflammatory cytokines and chemokines and a reduction of C5a at 24 h, but not 5 d after infection. Brain CCL2 levels were increased in both sexes 24 h after infection, but increased only in males at 5 d post infection. Conclusion: Ongoing S. epidermidis infection combined with neonatal HI increases the vulnerability of the developing brain in male but not in female mice. These sex-dependent effects were to a large extent independent of expression of systemic cytokines or brain CCL2 expression. Overall, we provide new insights into how systemic S. epidermidis infection affects the developing brain and show that the time interval between infection and HI is a critical sensitizing factor in males.
Collapse
Affiliation(s)
- Giacomo Gravina
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
32
|
Zhu H, Meissner LE, Byrnes C, Tuymetova G, Tifft CJ, Proia RL. The Complement Regulator Susd4 Influences Nervous-System Function and Neuronal Morphology in Mice. iScience 2020; 23:100957. [PMID: 32179479 PMCID: PMC7075988 DOI: 10.1016/j.isci.2020.100957] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 10/31/2019] [Accepted: 02/25/2020] [Indexed: 01/02/2023] Open
Abstract
The SUSD4 (Sushi domain-containing protein 4) gene encodes a complement inhibitor that is frequently deleted in 1q41q42 microdeletion syndrome, a multisystem congenital disorder that includes neurodevelopmental abnormalities. To understand SUSD4's role in the mammalian nervous system, we analyzed Susd4 knockout (KO) mice. Susd4 KO mice exhibited significant defects in motor performance and significantly higher levels of anxiety-like behaviors. Susd4 KO brain had abnormal “hairy” basket cells surrounding Purkinje neurons within the cerebellum and significantly reduced dendritic spine density in hippocampal pyramidal neurons. Neurons and oligodendrocyte lineage cells of wild-type mice were found to express Susd4 mRNA. Protein expression of the complement component C1q was increased in the brains of Susd4 KO mice. Our data indicate that SUSD4 plays an important role in neuronal functions, possibly via the complement pathway, and that SUSD4 deletion may contribute to the nervous system abnormalities in patients with 1q41q42 deletions. Susd4 is expressed in neurons and oligodendrocyte lineage cells Susd4 knockout mice have abnormal hippocampal and cerebellar neuronal morphologies Susd4 knockout mice exhibit anxiety-like behaviors and impaired motor function Susd4 knockout mice have elevated brain levels of the complement component C1q
Collapse
Affiliation(s)
- Hongling Zhu
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Laura E Meissner
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Colleen Byrnes
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Galina Tuymetova
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Cynthia J Tifft
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA.
| |
Collapse
|
33
|
Lo MW, Woodruff TM. Complement: Bridging the innate and adaptive immune systems in sterile inflammation. J Leukoc Biol 2020; 108:339-351. [PMID: 32182389 DOI: 10.1002/jlb.3mir0220-270r] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The complement system is a collection of soluble and membrane-bound proteins that together act as a powerful amplifier of the innate and adaptive immune systems. Although its role in infection is well established, complement is becoming increasingly recognized as a key contributor to sterile inflammation, a chronic inflammatory process often associated with noncommunicable diseases. In this context, damaged tissues release danger signals and trigger complement, which acts on a range of leukocytes to augment and bridge the innate and adaptive immune systems. Given the detrimental effect of chronic inflammation, the complement system is therefore well placed as an anti-inflammatory drug target. In this review, we provide a general outline of the sterile activators, effectors, and targets of the complement system and a series of examples (i.e., hypertension, cancer, allograft transplant rejection, and neuroinflammation) that highlight complement's ability to bridge the 2 arms of the immune system.
Collapse
Affiliation(s)
- Martin W Lo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
34
|
Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial Cells: Role of the Immune Response in Ischemic Stroke. Front Immunol 2020; 11:294. [PMID: 32174916 PMCID: PMC7055422 DOI: 10.3389/fimmu.2020.00294] [Citation(s) in RCA: 377] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke, which accounts for 75-80% of all strokes, is the predominant cause of morbidity and mortality worldwide. The post-stroke immune response has recently emerged as a new breakthrough target in the treatment strategy for ischemic stroke. Glial cells, including microglia, astrocytes, and oligodendrocytes, are the primary components of the peri-infarct environment in the central nervous system (CNS) and have been implicated in post-stroke immune regulation. However, increasing evidence suggests that glial cells exert beneficial and detrimental effects during ischemic stroke. Microglia, which survey CNS homeostasis and regulate innate immune responses, are rapidly activated after ischemic stroke. Activated microglia release inflammatory cytokines that induce neuronal tissue injury. By contrast, anti-inflammatory cytokines and neurotrophic factors secreted by alternatively activated microglia are beneficial for recovery after ischemic stroke. Astrocyte activation and reactive gliosis in ischemic stroke contribute to limiting brain injury and re-establishing CNS homeostasis. However, glial scarring hinders neuronal reconnection and extension. Neuroinflammation affects the demyelination and remyelination of oligodendrocytes. Myelin-associated antigens released from oligodendrocytes activate peripheral T cells, thereby resulting in the autoimmune response. Oligodendrocyte precursor cells, which can differentiate into oligodendrocytes, follow an ischemic stroke and may result in functional recovery. Herein, we discuss the mechanisms of post-stroke immune regulation mediated by glial cells and the interaction between glial cells and neurons. In addition, we describe the potential roles of various glial cells at different stages of ischemic stroke and discuss future intervention targets.
Collapse
Affiliation(s)
- Shenbin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
35
|
Tenner AJ. Complement-Mediated Events in Alzheimer's Disease: Mechanisms and Potential Therapeutic Targets. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:306-315. [PMID: 31907273 PMCID: PMC6951444 DOI: 10.4049/jimmunol.1901068] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022]
Abstract
An estimated 5.7 million Americans suffer from Alzheimer's disease in the United States, with no disease-modifying treatments to prevent or treat cognitive deficits associated with the disease. Genome-wide association studies suggest that an enhancement of clearance mechanisms and/or promotion of an anti-inflammatory response may slow or prevent disease progression. Increasing awareness of distinct roles of complement components in normal brain development and function and in neurodegenerative disorders align with complement-mediated responses, and thus, thorough understanding of these molecular pathways is needed to facilitate successful therapeutic design. Both beneficial and detrimental effects of C1q as well as contributions to local inflammation by C5a-C5aR1 signaling in brain highlight the need for precision of therapeutic design. The potential benefit of β-amyloid clearance from the circulation via CR1-mediated mechanisms is also reviewed. Therapies that suppress inflammation while preserving protective effects of complement could be tested now to slow the progression of this debilitating disease.
Collapse
Affiliation(s)
- Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697;
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697;
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697; and
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697
| |
Collapse
|
36
|
van Dijk BJ, Meijers JCM, Kloek AT, Knaup VL, Rinkel GJE, Morgan BP, van der Kamp MJ, Osuka K, Aronica E, Ruigrok YM, van de Beek D, Brouwer M, Pekna M, Hol EM, Vergouwen MDI. Complement C5 Contributes to Brain Injury After Subarachnoid Hemorrhage. Transl Stroke Res 2019; 11:678-688. [PMID: 31811640 PMCID: PMC7340633 DOI: 10.1007/s12975-019-00757-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/29/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
Previous studies showed that complement activation is associated with poor functional outcome after aneurysmal subarachnoid hemorrhage (SAH). We investigated whether complement activation is underlying brain injury after aneurysmal SAH (n = 7) and if it is an appropriate treatment target. We investigated complement expression in brain tissue of aneurysmal SAH patients (n = 930) and studied the role of common genetic variants in C3 and C5 genes in outcome. We analyzed plasma levels (n = 229) to identify the functionality of a single nucleotide polymorphism (SNP) associated with outcome. The time course of C5a levels was measured in plasma (n = 31) and CSF (n = 10). In an SAH mouse model, we studied the extent of microglia activation and cell death in wild-type mice, mice lacking the C5a receptor, and in mice treated with C5-specific antibodies (n = 15 per group). Brain sections from aneurysmal SAH patients showed increased presence of complement components C1q and C3/C3b/iC3B compared to controls. The complement component 5 (C5) SNP correlated with C5a plasma levels and poor disease outcome. Serial measurements in CSF revealed that C5a was > 1400-fold increased 1 day after aneurysmal SAH and then gradually decreased. C5a in plasma was 2-fold increased at days 3–10 after aneurysmal SAH. In the SAH mouse model, we observed a ≈ 40% reduction in both microglia activation and cell death in mice lacking the C5a receptor, and in mice treated with C5-specific antibodies. These data show that C5 contributes to brain injury after experimental SAH, and support further study of C5-specific antibodies as novel treatment option to reduce brain injury and improve prognosis after aneurysmal SAH.
Collapse
Affiliation(s)
- Bart J van Dijk
- UMC Utrecht Brain Center, Department of Translational Neurosciences, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands.,UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands
| | - Joost C M Meijers
- Department of Experimental Vascular Medicine, Academic Medical Center, Meibergdreef 9, Amsterdam, The Netherlands.,Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, Amsterdam, The Netherlands
| | - Anne T Kloek
- Department of Neurology, Amsterdam Neuroscience, Academic Medical Center, Meibergdreef 9, Amsterdam, The Netherlands
| | - Veronique L Knaup
- Department of Experimental Vascular Medicine, Academic Medical Center, Meibergdreef 9, Amsterdam, The Netherlands
| | - Gabriel J E Rinkel
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands
| | - B Paul Morgan
- Systems Immunity Research Institute, Cardiff University, Heath Park, Cardiff, UK
| | - Marije J van der Kamp
- UMC Utrecht Brain Center, Department of Translational Neurosciences, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands
| | - Koji Osuka
- Department of Neurological Surgery, Aichi Medical University, 1-1 Karimatayazako, Aichi, Japan
| | - Eleonora Aronica
- Department of Neuropathology, Academic Medical Center, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ynte M Ruigrok
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands
| | - Diederik van de Beek
- Department of Neurology, Amsterdam Neuroscience, Academic Medical Center, Meibergdreef 9, Amsterdam, The Netherlands
| | - Matthijs Brouwer
- Department of Neurology, Amsterdam Neuroscience, Academic Medical Center, Meibergdreef 9, Amsterdam, The Netherlands
| | - Marcela Pekna
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 9A, Gothenburg, Sweden
| | - Elly M Hol
- UMC Utrecht Brain Center, Department of Translational Neurosciences, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands.,Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, The Netherlands
| | - Mervyn D I Vergouwen
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands.
| |
Collapse
|
37
|
Lee JD, Coulthard LG, Woodruff TM. Complement dysregulation in the central nervous system during development and disease. Semin Immunol 2019; 45:101340. [PMID: 31708347 DOI: 10.1016/j.smim.2019.101340] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/15/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
The complement cascade is an important arm of the immune system that plays a key role in protecting the central nervous system (CNS) from infection. Recently, it has also become clear that complement proteins have fundamental roles in the developing and aging CNS that are distinct from their roles in immunity. During neurodevelopment, complement signalling is involved in diverse processes including neural tube closure, neural progenitor proliferation and differentiation, neuronal migration, and synaptic pruning. In acute neurotrauma and ischamic brain injury, complement drives inflammation and neuronal death, but also neuroprotection and regeneration. In diseases of the aging CNS including dementias and motor neuron disease, chronic complement activation is associated with glial activation, and synapse and neuron loss. Proper regulation of complement is thus essential to allow for an appropriately developed CNS and prevention of excessive damage following neurotrauma or during neurodegeneration. This review provides a comprehensive overview of the evidence for functional roles of complement in brain formation, and its dysregulation during acute and chronic disease. We also provide working models for how complement can lead to neurodevelopmental disorders such as schizophrenia and autism, and either protect, or propagate neurodegenerative diseases including Alzheimer's disease and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Liam G Coulthard
- Royal Brisbane and Women's Hospital, Herston, Australia; School of Clinical Medicine, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
38
|
Li S, Jiang D, Rosenkrans ZT, Barnhart TE, Ehlerding EB, Ni D, Engle JW, Cai W. Aptamer-Conjugated Framework Nucleic Acids for the Repair of Cerebral Ischemia-Reperfusion Injury. NANO LETTERS 2019; 19:7334-7341. [PMID: 31518140 PMCID: PMC6876547 DOI: 10.1021/acs.nanolett.9b02958] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Effective therapy for protecting dying neurons against cerebral ischemia-reperfusion injury (IRI) represents a substantial challenge in the treatment of ischemic strokes. Oxidative stress coupled with excessive inflammation is the main culprit for brain IRI that results in neuronal damage and disability. Specifically, complement component 5a (C5a) exacerbates the vicious cycle between oxidative stress and inflammatory responses. Herein, we propose that a framework nucleic acid (FNA) conjugated with anti-C5a aptamers (aC5a) can selectively reduce C5a-mediated neurotoxicity and effectively alleviate oxidative stress in the brain. Intrathecal injection of the aC5a-conjugated FNA (aC5a-FNA) was applied for the treatment of rats with ischemic strokes. Positron emission tomography (PET) imaging was performed to investigate the accumulation of aC5a-FNA in the penumbra and its therapeutic efficacy. Results demonstrated that aC5a-FNA could rapidly penetrate different brain regions after brain IRI. Furthermore, aC5a-FNA effectively protected neurons from brain IRI, as verified by serum tests, tissue staining, biomarker detection, and functional assessment. The protective effect of aC5a-FNA against cerebral IRI in living animals may pave the way for the translation of FNA from bench to bedside and broaden the horizon of FNA in the field of biomedicine.
Collapse
Affiliation(s)
- Shiyong Li
- Department of Rehabilitation, Second Affiliated Hospital of Nanchang University, Jiangxi 330006, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Zachary T. Rosenkrans
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Todd E. Barnhart
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Emily B. Ehlerding
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Dalong Ni
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jonathan W. Engle
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
39
|
Clarke AR, Christophe BR, Khahera A, Sim JL, Connolly ES. Therapeutic Modulation of the Complement Cascade in Stroke. Front Immunol 2019; 10:1723. [PMID: 31417544 PMCID: PMC6682670 DOI: 10.3389/fimmu.2019.01723] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/09/2019] [Indexed: 01/22/2023] Open
Abstract
Stroke is a leading cause of death and disability worldwide and an increasing number of ischemic stroke patients are undergoing pharmacological and mechanical reperfusion. Both human and experimental models of reperfused ischemic stroke have implicated the complement cascade in secondary tissue injury. Most data point to the lectin and alternative pathways as key to activation, and C3a and C5a binding of their receptors as critical effectors of injury. During periods of thrombolysis use to treat stroke, acute experimental complement cascade blockade has been found to rescue tissue and improves functional outcome. Blockade of the complement cascade during the period of tissue reorganization, repair, and recovery is by contrast not helpful and in fact is likely to be deleterious with emerging data suggesting downstream upregulation of the cascade might even facilitate recovery. Successful clinical translation will require the right clinical setting and pharmacologic strategies that are capable of targeting the key effectors early while not inhibiting delayed repair. Early reports in a variety of disease states suggest that such pharmacologic strategies appear to have a favorable risk profile and offer substantial hope for patients.
Collapse
Affiliation(s)
- Alison R Clarke
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - Brandon R Christophe
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - Anadjeet Khahera
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - Justin L Sim
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| | - E Sander Connolly
- Cerebrovascular Research Laboratory, Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
40
|
The Novel C5aR Antagonist DF3016A Protects Neurons Against Ischemic Neuroinflammatory Injury. Neurotox Res 2019; 36:163-174. [PMID: 30953275 PMCID: PMC6570783 DOI: 10.1007/s12640-019-00026-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 12/25/2022]
Abstract
The central nervous system (CNS) constitutively expresses complement (C) membrane receptors and complement proteins, including the component C5a. This is a crucial terminal effector of the C cascade, mostly involved in pain and neuroinflammatory conditions. Aberrant activation of C5a protein and its receptor C5aR has been reported to play a critical role in neurodegenerative diseases, with important clinical consequences. Here we have investigated the effects of DF3016A, a novel selective C5aR antagonist, able to penetrate the blood-brain barrier (BBB), on cortical neurons exposed to oxygen-glucose deprivation-reoxygenation (OGD/R), a neuroinflammation-related process. We demonstrated that a mild ischemic insult induces an early upregulation of C5aR associated with the over-production of pro-inflammatory cytokines and the over-expression of the transcriptional regulatory factor miR-181. Furthermore, we report the first experimental evidence of the effect of DF3016A, modulating complement component C5a, on neurons in a model of injury. Interestingly, DF3016A protects neuronal viability by restoring intracellular calcium levels, thus opposing the increase in pro-inflammatory cytokine levels and miR-181 expression. Based on our results, we suggest that DF3016A is a novel C5aR antagonist promoting protective effects against OGD/R-induced damage that could be a new therapeutic approach to controlling CNS neuroinflammatory conditions.
Collapse
|
41
|
Ma Y, Liu Y, Zhang Z, Yang GY. Significance of Complement System in Ischemic Stroke: A Comprehensive Review. Aging Dis 2019; 10:429-462. [PMID: 31011487 PMCID: PMC6457046 DOI: 10.14336/ad.2019.0119] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system is an essential part of innate immunity, typically conferring protection via eliminating pathogens and accumulating debris. However, the defensive function of the complement system can exacerbate immune, inflammatory, and degenerative responses in various pathological conditions. Cumulative evidence indicates that the complement system plays a critical role in the pathogenesis of ischemic brain injury, as the depletion of certain complement components or the inhibition of complement activation could reduce ischemic brain injury. Although multiple candidates modulating or inhibiting complement activation show massive potential for the treatment of ischemic stroke, the clinical availability of complement inhibitors remains limited. The complement system is also involved in neural plasticity and neurogenesis during cerebral ischemia. Thus, unexpected side effects could be induced if the systemic complement system is inhibited. In this review, we highlighted the recent concepts and discoveries of the roles of different kinds of complement components, such as C3a, C5a, and their receptors, in both normal brain physiology and the pathophysiology of brain ischemia. In addition, we comprehensively reviewed the current development of complement-targeted therapy for ischemic stroke and discussed the challenges of bringing these therapies into the clinic. The design of future experiments was also discussed to better characterize the role of complement in both tissue injury and recovery after cerebral ischemia. More studies are needed to elucidate the molecular and cellular mechanisms of how complement components exert their functions in different stages of ischemic stroke to optimize the intervention of targeting the complement system.
Collapse
Affiliation(s)
- Yuanyuan Ma
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqun Liu
- 3Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijun Zhang
- 2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Pekny M, Wilhelmsson U, Tatlisumak T, Pekna M. Astrocyte activation and reactive gliosis-A new target in stroke? Neurosci Lett 2018; 689:45-55. [PMID: 30025833 DOI: 10.1016/j.neulet.2018.07.021] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/03/2018] [Accepted: 07/14/2018] [Indexed: 11/27/2022]
Abstract
Stroke is an acute insult to the central nervous system (CNS) that triggers a sequence of responses in the acute, subacute as well as later stages, with prominent involvement of astrocytes. Astrocyte activation and reactive gliosis in the acute stage of stroke limit the tissue damage and contribute to the restoration of homeostasis. Astrocytes also control many aspects of neural plasticity that is the basis for functional recovery. Here, we discuss the concept of intermediate filaments (nanofilaments) and the complement system as two handles on the astrocyte responses to injury that both present attractive opportunities for novel treatment strategies modulating astrocyte functions and reactive gliosis.
Collapse
Affiliation(s)
- Milos Pekny
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Newcastle, Newcastle, NSW, Australia.
| | - Ulrika Wilhelmsson
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden
| | - Turgut Tatlisumak
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcela Pekna
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
43
|
Tenner AJ, Stevens B, Woodruff TM. New tricks for an ancient system: Physiological and pathological roles of complement in the CNS. Mol Immunol 2018; 102:3-13. [PMID: 29958698 DOI: 10.1016/j.molimm.2018.06.264] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022]
Abstract
While the mechanisms underlying the functions of the complement system in the central nervous system (CNS) and systemically, namely opsonization, chemotaxis, membrane lysis, and regulation of inflammation are the same, the plethora of functions that complement orchestrates in the central nervous system (CNS) is complex. Strictly controlled expression of complement effector molecules, regulators and receptors across the gamut of life stages (embryogenesis, development and maturation, aging and disease) dictate fascinating contributions for this ancient system. Furthermore, it is becoming apparent that complement functions differ widely across distinct brain regions. This review provides a comprehensive overview of the newly identified roles for complement in the brain, including its roles in CNS development and function, during aging and in the processes of neurodegeneration. The diversity and selectively of beneficial and detrimental activities of complement, while challenging, should lead to precision targeting of specific components to provide disease modifying treatments for devastating psychiatric and neurodegenerative disorders that are still without effective treatment.
Collapse
Affiliation(s)
- Andrea J Tenner
- Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, CA, United States.
| | - Beth Stevens
- F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Department of Neurobiology, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
44
|
Gu Z, Yan S, Cheong S, Cao Z, Zuo H, Thomas AC, Rolfe BE, Xu ZP. Layered double hydroxide nanoparticles: Impact on vascular cells, blood cells and the complement system. J Colloid Interface Sci 2018; 512:404-410. [DOI: 10.1016/j.jcis.2017.10.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
|
45
|
Jabłoński M, Mazur JK, Tarnowski M, Dołęgowska B, Pędziwiatr D, Kubiś E, Budkowska M, Sałata D, Wysiecka JP, Kazimierczak A, Reginia A, Ratajczak MZ, Samochowiec J. Mobilization of Peripheral Blood Stem Cells and Changes in the Concentration of Plasma Factors Influencing their Movement in Patients with Panic Disorder. Stem Cell Rev Rep 2017; 13:217-225. [PMID: 27914035 PMCID: PMC5380702 DOI: 10.1007/s12015-016-9700-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this paper we examined whether stem cells and factors responsible for their movement may serve as new biological markers of anxiety disorders. The study was carried out on a group of 30 patients diagnosed with panic disorder (examined before and after treatment), compared to 30 healthy individuals forming the control group. We examined the number of circulating HSCs (hematopoetic stem cells) (Lin−/CD45 +/CD34 +) and HSCs (Lin−/CD45 +/AC133 +), the number of circulating VSELs (very small embryonic-like stem cells) (Lin−/CD45−/CD34 +) and VSELs (Lin−/CD45−/AC133 +), as well as the concentration of complement components: C3a, C5a and C5b-9, SDF-1 (stromal derived factor) and S1P (sphingosine-1-phosphate). Significantly lower levels of HSCs (Lin−/CD45 +/AC133 +) have been demonstrated in the patient group compared to the control group both before and after treatment. The level of VSELs (Lin−/CD45−/CD133 +) was significantly lower in the patient group before treatment as compared to the patient group after treatment. The levels of factors responsible for stem cell movement were significantly lower in the patient group compared to the control group before and after treatment. It was concluded that the study of stem cells and factors associated with their movement can be useful in the diagnostics of panic disorder, as well as differentiating between psychotic and anxiety disorders.
Collapse
Affiliation(s)
- Marcin Jabłoński
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460, Szczecin, Poland
| | - Jolanta Kucharska Mazur
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460, Szczecin, Poland.
| | - Maciej Tarnowski
- Department of Physiology, Pomeranian University of Medicine, Szczecin, Poland
| | - Barbara Dołęgowska
- Department of Medical Analytics, Pomeranian University of Medicine, Szczecin, Poland
| | - Daniel Pędziwiatr
- Department of Physiology, Pomeranian University of Medicine, Szczecin, Poland
| | - Ewa Kubiś
- Department of Physiology, Pomeranian University of Medicine, Szczecin, Poland
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian University of Medicine, Szczecin, Poland
| | - Daria Sałata
- Department of Medical Analytics, Pomeranian University of Medicine, Szczecin, Poland
| | - Justyna Pełka Wysiecka
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460, Szczecin, Poland
| | | | - Artur Reginia
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460, Szczecin, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Biology Program at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460, Szczecin, Poland
| |
Collapse
|
46
|
Hernandez MX, Namiranian P, Nguyen E, Fonseca MI, Tenner AJ. C5a Increases the Injury to Primary Neurons Elicited by Fibrillar Amyloid Beta. ASN Neuro 2017; 9:1759091416687871. [PMID: 28078911 PMCID: PMC5298486 DOI: 10.1177/1759091416687871] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
C5aR1, the proinflammatory receptor for C5a, is expressed in the central nervous system on microglia, endothelial cells, and neurons. Previous work demonstrated that the C5aR1 antagonist, PMX205, decreased amyloid pathology and suppressed cognitive deficits in two Alzheimer's Disease (AD) mouse models. However, the cellular mechanisms of this protection have not been definitively demonstrated. Here, primary cultured mouse neurons treated with exogenous C5a show reproducible loss of MAP-2 staining in a dose-dependent manner within 24 hr of treatment, indicative of injury to neurons. This injury is prevented by the C5aR1 antagonist PMX53, a close analog of PMX205. Furthermore, primary neurons derived from C5aR1 null mice exhibited no MAP-2 loss after exposure to the highest concentration of C5a tested. Primary mouse neurons treated with both 100 nM C5a and 5 µM fibrillar amyloid beta (fAβ), to model what occurs in the AD brain, showed increased MAP-2 loss relative to either C5a or fAβ alone. Blocking C5aR1 with PMX53 (100 nM) blocked the loss of MAP2 in these primary neurons to the level seen with fAβ alone. Similar experiments with primary neurons derived from C5aR1 null mice showed a loss of MAP-2 due to fAβ treatment. However, the addition of C5a to the cultures did not enhance the loss of MAP-2 and the addition of PMX53 to the cultures did not change the MAP-2 loss in response to fAβ. Thus, at least part of the beneficial effects of C5aR1 antagonist in AD mouse models may be due to protection of neurons from the toxic effects of C5a.
Collapse
Affiliation(s)
- Michael X Hernandez
- 1 Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, USA
| | - Pouya Namiranian
- 2 Department of Molecular Biology and Biochemistry, University of California, Irvine, USA
| | - Eric Nguyen
- 2 Department of Molecular Biology and Biochemistry, University of California, Irvine, USA
| | - Maria I Fonseca
- 2 Department of Molecular Biology and Biochemistry, University of California, Irvine, USA
| | - Andrea J Tenner
- 1 Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, USA.,2 Department of Molecular Biology and Biochemistry, University of California, Irvine, USA.,3 Department of Neurobiology and Behavior, University of California, Irvine, USA
| |
Collapse
|
47
|
Peterson SL, Nguyen HX, Mendez OA, Anderson AJ. Complement Protein C3 Suppresses Axon Growth and Promotes Neuron Loss. Sci Rep 2017; 7:12904. [PMID: 29018286 PMCID: PMC5635131 DOI: 10.1038/s41598-017-11410-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/22/2017] [Indexed: 01/29/2023] Open
Abstract
The inflammatory response to spinal cord injury (SCI) involves localization and activation of innate and adaptive immune cells and proteins, including the complement cascade. Complement C3 is important for the classical, alternative, and lectin pathways of complement activation, and its cleavage products C3a and C3b mediate several functions in the context of inflammation, but little is known about the potential functions of C3 on regeneration and survival of injured neurons after SCI. We report that 6 weeks after dorsal hemisection with peripheral conditioning lesion, C3-/- mice demonstrated a 2-fold increase in sensory axon regeneration in the spinal cord in comparison to wildtype C3+/+ mice. In vitro, addition of C3 tripled both myelin-mediated neurite outgrowth inhibition and neuron loss versus myelin alone, and ELISA experiments revealed that myelin serine proteases cleave C3 to generate active fragments. Addition of purified C3 cleavage products to cultured neurons suggested that C3b is responsible for the growth inhibitory and neurotoxic or anti-adhesion activities of C3. These data indicate that C3 reduces neurite outgrowth and neuronal viability in vitro and restricts axon regeneration in vivo, and demonstrate a novel, non-traditional role for this inflammatory protein in the central nervous system.
Collapse
Affiliation(s)
- Sheri L Peterson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA, 92697, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Hal X Nguyen
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA, 92697, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Oscar A Mendez
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA, 92697, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA, 92697, USA. .,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA. .,Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, 92697, USA. .,Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
48
|
Hernandez MX, Jiang S, Cole TA, Chu SH, Fonseca MI, Fang MJ, Hohsfield LA, Torres MD, Green KN, Wetsel RA, Mortazavi A, Tenner AJ. Prevention of C5aR1 signaling delays microglial inflammatory polarization, favors clearance pathways and suppresses cognitive loss. Mol Neurodegener 2017; 12:66. [PMID: 28923083 PMCID: PMC5604420 DOI: 10.1186/s13024-017-0210-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/12/2017] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Pharmacologic inhibition of C5aR1, a receptor for the complement activation proinflammatory fragment, C5a, suppressed pathology and cognitive deficits in Alzheimer's disease (AD) mouse models. To validate that the effect of the antagonist was specifically via C5aR1 inhibition, mice lacking C5aR1 were generated and compared in behavior and pathology. In addition, since C5aR1 is primarily expressed on cells of the myeloid lineage, and only to a lesser extent on endothelial cells and neurons in brain, gene expression in microglia isolated from adult brain at multiple ages was compared across all genotypes. METHODS C5aR1 knock out mice were crossed to the Arctic AD mouse model, and characterized for pathology and for behavior performance in a hippocampal dependent memory task. CX3CR1GFP and CCR2RFP reporter mice were bred to C5aR1 sufficient and knockout wild type and Arctic mice to enable sorting of microglia (GFP-positive, RFP-negative) isolated from adult brain at 2, 5, 7 and 10 months of age followed by RNA-seq analysis. RESULTS A lack of C5aR1 prevented behavior deficits at 10 months, although amyloid plaque load was not altered. Immunohistochemical analysis showed no CCR2+ monocytes/macrophages near the plaques in the Arctic brain with or without C5aR1. Microglia were sorted from infiltrating monocytes (GFP and RFP-positive) for transcriptome analysis. RNA-seq analysis identified inflammation related genes as differentially expressed, with increased expression in the Arctic mice relative to wild type and decreased expression in the Arctic/C5aR1KO relative to Arctic. In addition, phagosomal-lysosomal gene expression was increased in the Arctic mice relative to wild type but further increased in the Arctic/C5aR1KO mice. A decrease in neuronal complexity was seen in hippocampus of 10 month old Arctic mice at the time that correlates with the behavior deficit, both of which were rescued in the Arctic/C5aR1KO. CONCLUSIONS These data are consistent with microglial polarization in the absence of C5aR1 signaling reflecting decreased induction of inflammatory genes and enhancement of degradation/clearance pathways, which is accompanied by preservation of CA1 neuronal complexity and hippocampal dependent cognitive function. These results provide links between microglial responses and loss of cognitive performance and, combined with the previous pharmacological approach to inhibit C5aR1 signaling, support the potential of this receptor as a novel therapeutic target for AD in humans.
Collapse
Affiliation(s)
- Michael X Hernandez
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, CA, USA
| | - Shan Jiang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Tracy A Cole
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Present Address: Ionis Pharmaceuticals Inc., Carlsbad, CA, 92010, USA
| | - Shu-Hui Chu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Maria I Fonseca
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Melody J Fang
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Maria D Torres
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Rick A Wetsel
- Research Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas-Houston, Houston, TX, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
49
|
Zhu M, He X, Wang XH, Qiu W, Xing W, Guo W, An TC, Ao LQ, Hu XT, Li Z, Liu XP, Xiao N, Yu J, Huang H, Xu X. Complement C5a induces mesenchymal stem cell apoptosis during the progression of chronic diabetic complications. Diabetologia 2017; 60:1822-1833. [PMID: 28577176 DOI: 10.1007/s00125-017-4316-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/25/2017] [Indexed: 12/28/2022]
Abstract
AIMS/HYPOTHESIS Regeneration and repair mediated by mesenchymal stem cells (MSCs) are key self-protection mechanisms against diabetic complications, a reflection of diabetes-related cell/tissue damage and dysfunction. MSC abnormalities have been reported during the progression of diabetic complications, but little is known about whether a deficiency in these cells plays a role in the pathogenesis of this disease. In addition to MSC resident sites, peripheral circulation is a major source of MSCs that participate in the regeneration and repair of damaged tissue. Therefore, we investigated whether there is a deficiency of circulating MSC-like cells in people with diabetes and explored the underlying mechanisms. METHODS The abundance of MSC-like cells in peripheral blood was evaluated by FACS. Selected diabetic and non-diabetic serum (DS and NDS, respectively) samples were used to mimic diabetic and non-diabetic microenvironments, respectively. The proliferation and survival of MSCs under different serum conditions were analysed using several detection methods. The survival of MSCs in diabetic microenvironments was also investigated in vivo using leptin receptor mutant (Lepr db/db ) mice. RESULTS Our data showed a significant decrease in the abundance of circulating MSC-like cells, which was correlated with complications in individuals with type 2 diabetes. DS strongly impaired the proliferation and survival of culture-expanded MSCs through the complement system but not through exposure to high glucose levels. DS-induced MSC apoptosis was mediated, at least in part, by the complement C5a-dependent upregulation of Fas-associated protein with death domain (FADD) and the Bcl-2-associated X protein (BAX)/B cell lymphoma 2 (Bcl-2) ratio, which was significantly inhibited by neutralising C5a or by the pharmacological or genetic inhibition of the C5a receptor (C5aR) on MSCs. Moreover, blockade of the C5a/C5aR pathway significantly inhibited the apoptosis of transplanted MSCs in Lepr db/db recipient mice. CONCLUSIONS/INTERPRETATION C5a-dependent apoptotic death is probably involved in MSC deficiency and in the progression of complications in individuals with type 2 diabetes. Therefore, anticomplement therapy may be a novel intervention for diabetic complications.
Collapse
Affiliation(s)
- Ming Zhu
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Xiao He
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Xiao-Hui Wang
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
- Department of Histology and Embryology, Medical College of Qingdao University, Qingdao, People's Republic of China
| | - Wei Qiu
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Wei Xing
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Wei Guo
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Tian-Chen An
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Luo-Quan Ao
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Xue-Ting Hu
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Zhan Li
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Xiao-Ping Liu
- Department of Histology and Embryology, Medical College of Qingdao University, Qingdao, People's Republic of China
| | - Nan Xiao
- Ninth Department, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Jian Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hong Huang
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China.
| | - Xiang Xu
- First Department, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital and Research Institute of Surgery, Third Military Medical University, No. 10 Changjiang Branch Road, Daping Street, Yuzhong District, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
50
|
Luo C, Ouyang MW, Fang YY, Li SJ, Zhou Q, Fan J, Qin ZS, Tao T. Dexmedetomidine Protects Mouse Brain from Ischemia-Reperfusion Injury via Inhibiting Neuronal Autophagy through Up-Regulating HIF-1α. Front Cell Neurosci 2017; 11:197. [PMID: 28729825 PMCID: PMC5498477 DOI: 10.3389/fncel.2017.00197] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/22/2017] [Indexed: 11/14/2022] Open
Abstract
Stroke is the leading cause of death in China and produces a heavy socio-economic burden in the past decades. Previous studies have shown that dexmedetomidine (DEX) is neuroprotective after cerebral ischemia. However, the role of autophagy during DEX-mediated neuroprotection after cerebral ischemia is still unknown. In this study, we found that post-conditioning with DEX and DEX+3-methyladenine (3-MA) (autophagy inhibitor) reduced brain infarct size and improved neurological deficits compared with DEX+RAPA (autophagy inducer) 24 h after transient middle cerebral artery artery occlusion (tMCAO) model in mice. DEX inhibited the neuronal autophagy in the peri-ischemic brain, and increased viability and decreased apoptosis of primary cultured neurons in oxygen-glucose deprivation (OGD) model. DEX induced expression of Bcl-1 and p62, while reduced the expression of microtubule-associated protein 1 light chain 3 (LC3) and Beclin 1 in primary cultured neurons through inhibition of apoptosis and autophagy. Meanwhile, DEX promoted the expression of hypoxia-inducible factor-1α (HIF-1α) both in vivo and in vitro, and 2-Methoxyestradiol (2ME2), an inhibitor of HIF-1α, could reverse DEX-induced autophagic inhibition. In conclusion, our study suggests that post-conditioning with DEX at the beginning of reperfusion protects mouse brain from ischemia-reperfusion injury via inhibition of neuronal autophagy by upregulation of HIF-1α, which provides a potential therapeutic treatment for acute ischemic injury.
Collapse
Affiliation(s)
- Cong Luo
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Ming-Wen Ouyang
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical UniversityGuangzhou, China
| | - Ying-Ying Fang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou, China
| | - Shu-Ji Li
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhou, China
| | - Quan Zhou
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Jun Fan
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Zai-Sheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| | - Tao Tao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical UniversityGuangzhou, China
| |
Collapse
|