1
|
Yang F, Gao W, Wang J, Li X, Li H. Progress of Chinese Medicine in Regulating Microglial Polarization against Alzheimer's Disease. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2255-2275. [PMID: 39721955 DOI: 10.1142/s0192415x24500873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD), the predominant form of dementia, is a neurodegenerative disorder of the central nervous system (CNS) characterized by a subtle onset and a spectrum of cognitive and functional declines. The clinical manifestation of AD encompasses memory deficits, cognitive deterioration, and behavioral disturbances, culminating in a severe impairment of daily living skills. Despite its high prevalence, accounting for 60-70% of all dementia cases, there remains an absence of curative therapeutics. Microglia (MG), the resident immune cells of the CNS, exhibit a bifurcated role in AD pathogenesis. Functioning in a neuroprotective capacity, MGs express scavenger receptors, facilitating the clearance of [Formula: see text]-amyloid protein (A[Formula: see text]) and cellular debris. Conversely, aberrant activation of MGs can lead to the secretion of pro-inflammatory cytokines, thereby propagating neuroinflammatory responses that are detrimental to neuronal integrity. The dynamics of MG activation and the ensuing neuroinflammation are pivotal in the evolution of AD. Chinese medicine (CM), a treasure trove of traditional Chinese cultural practices, has demonstrated significant potential in the therapeutic management of AD. Over the past triennium, CM has garnered considerable research attention for its multifaceted approaches to AD, including the regulation of MG polarization. This review synthesizes current knowledge on the origins, polarization dynamics, and mechanistic interplay of MG with AD pathology. It further explores the nexus between MG polarization and cardinal pathological hallmarks of AD, such as A[Formula: see text] plaque deposition, hyperphosphorylation of tau, synaptic plasticity impairments, neuroinflammation, and brain-gut-axis dysregulation. The review also encapsulates the therapeutic strategies of CM, which encompass monomers, formulae, and acupuncture. These strategies modulate MG polarization in the context of AD treatment, thereby providing a robust theoretical framework in which to conduct future investigative endeavors in both the clinical and preclinical realms.
Collapse
Affiliation(s)
- Fengge Yang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Wei Gao
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- Jiangsu College of Nursing Jiangsu, Huaian, Huaiyin 223001, P. R. China
| | - Junting Wang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| | - Xue Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Honglin Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| |
Collapse
|
2
|
Cheng Q, Fan Y, Zhang P, Liu H, Han J, Yu Q, Wang X, Wu S, Lu Z. Biomarkers of Synaptic Degeneration in Alzheimer's Disease. Ageing Res Rev 2024:102642. [PMID: 39701184 DOI: 10.1016/j.arr.2024.102642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Synapse has been considered a critical neuronal structure in the procession of Alzheimer's disease (AD), attacked by two pathological molecule aggregates (amyloid-β and phosphorylated tau) in the brain, disturbing synaptic homeostasis before disease manifestation and subsequently causing synaptic degeneration. Recently, evidence has emerged indicating that soluble oligomeric amyloid-β (AβO) and tau exert direct toxicity on synapses, causing synaptic damage. Synaptic degeneration is closely linked to cognitive decline in AD, even in the asymptomatic stages of AD. Therefore, the identification of novel, specific, and sensitive biomarkers involved in synaptic degeneration holds significant promise for early diagnosis of AD, reducing synaptic degeneration and loss, and controlling the progression of AD. Currently, a range of biomarkers in cerebrospinal fluid (CSF), such as synaptosome-associated protein 25 (SNAP-25), synaptotagmin-1, growth-associated protein-43 (GAP-43), and neurogranin (Ng), along with functional brain imaging techniques, can detect variations in synaptic density, offering high sensitivity and specificity for Alzheimer's disease (AD) diagnosis. However, these methods face challenges, including invasiveness, high cost, and limited accessibility. In contrast, biomarkers found in blood or urine provide a minimally invasive, cost-effective, and more accessible alternative to traditional diagnostic methods. Notably, neuron-derived exosomes in blood, which contain synaptic proteins, show variations in concentration that can serve as indicators of synaptic injury, providing an additional, less invasive approach to AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Yiou Fan
- Laboratory and Quality Management Department, Centers for Disease Control and Prevention of Shandong, Jinan, Shandong, China
| | - Pengfei Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Huan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Jialin Han
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Qian Yu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Xueying Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Shuang Wu
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China
| | - Zhiming Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong, China.
| |
Collapse
|
3
|
Tillmon H, Soteros BM, Shen L, Cong Q, Wollet M, General J, Chin H, Lee JB, Carreno FR, Morilak DA, Kim JH, Sia GM. Complement and microglia activation mediate stress-induced synapse loss in layer 2/3 of the medial prefrontal cortex in male mice. Nat Commun 2024; 15:9803. [PMID: 39532876 PMCID: PMC11557709 DOI: 10.1038/s41467-024-54007-5] [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: 11/28/2023] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Spatially heterogeneous synapse loss is a characteristic of many psychiatric and neurological disorders, but the underlying mechanisms are unclear. Here, we show that spatially-restricted complement activation mediates stress-induced heterogeneous microglia activation and synapse loss localized to the upper layers of the medial prefrontal cortex (mPFC) in male mice. Single cell RNA sequencing also reveals a stress-associated microglia state marked by high expression of the apolipoprotein E gene (Apoehigh) localized to the upper layers of the mPFC. Mice lacking complement component C3 are protected from stress-induced layer-specific synapse loss, and the Apoehigh microglia population is markedly reduced in the mPFC of these mice. Furthermore, C3 knockout mice are also resilient to stress-induced anhedonia and working memory behavioral deficits. Our findings suggest that region-specific complement and microglia activation can contribute to the disease-specific spatially restricted patterns of synapse loss and clinical symptoms found in many brain diseases.
Collapse
Affiliation(s)
- Haven Tillmon
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Breeanne M Soteros
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Liang Shen
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Qifei Cong
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
- Clinical Research Center of Neurological Disease, Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mackenna Wollet
- Department of Cellular and Integrative Physiology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Julianne General
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Hanna Chin
- University of Rochester, Rochester, NY, 14627, USA
| | - John Beichen Lee
- Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Flavia R Carreno
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - David A Morilak
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veteran's Health Care System, San Antonio, TX, 78229, USA
| | - Jun Hee Kim
- Department of Cellular and Integrative Physiology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gek Ming Sia
- Department of Cellular and Integrative Physiology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
4
|
Qin Y, Lin W, Ren Y. Ferroptosis involvement in the neurotoxicity of flunitrazepam in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 276:107128. [PMID: 39467492 DOI: 10.1016/j.aquatox.2024.107128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
In recent years, psychoactive drugs such as benzodiazepines (BZDs) have been frequently detected in water environments, however, there is still limited understanding regarding their potential impact on neurological health and underlying mechanisms. This study evaluated the neurotoxicity of the typical BZD drug flunitrazepam (FLZ, 0.2 and 5 μg/L) in zebrafish embryos and adults, and investigated the relationship between ferroptosis and FLZ-induced neurotoxicity. The results indicated that acute exposure to FLZ significantly inhibited zebrafish embryo hatching and promotes death, induced larval deformities, and led to abnormal neurobehavioral responses in larvae, likely due to ferroptosis induction. Results from a 30-day subacute exposure to FLZ showed that it decreased motor function and induced cognitive impairment in adult zebrafish. Immunofluorescence of brain tissues revealed a reduction in neurons in the telencephalon and an increase in microglia in the mesencephalon of the zebrafish exposed to FLZ. The ultrastructure of brain mitochondria showed serious damage. Besides, FLZ exposure increased iron levels, reduced GSH/GSSG and increased LPO in brain tissue, which is related to the abnormal expression of genes associated with ferroptosis. In the rescue experiments with co-exposure to deferoxamine (DFO), the motor-related parameters and biochemical indexes related to ferroptosis were restored, suggesting that FLZ can induce ferroptosis. The molecular docking results indicated that FLZ had a higher affinity with transferrin. This study elucidates the close relationship between ferroptosis and FLZ-induced neurotoxicity, which is significant for understanding the physiological damage caused by psychoactive substances and assessing environmental risks.
Collapse
Affiliation(s)
- Yingjun Qin
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Wenting Lin
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Yuan Ren
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China; The Key Lab of Pollution Control and Ecosystem Restoration in Industry Clusters, Ministry of Education, Guangzhou 510006, PR China; The Key Laboratory of Environmental Protection and Eco-Remediation of Guangdong Regular Higher Education Institutions, Guangzhou 510006, PR China.
| |
Collapse
|
5
|
Shen Y, Timsina J, Heo G, Beric A, Ali M, Wang C, Yang C, Wang Y, Western D, Liu M, Gorijala P, Budde J, Do A, Liu H, Gordon B, Llibre-Guerra JJ, Joseph-Mathurin N, Perrin RJ, Maschi D, Wyss-Coray T, Pastor P, Renton AE, Surace EI, Johnson ECB, Levey AI, Alvarez I, Levin J, Ringman JM, Allegri RF, Seyfried N, Day GS, Wu Q, Fernández MV, Tarawneh R, McDade E, Morris JC, Bateman RJ, Goate A, Ibanez L, Sung YJ, Cruchaga C. CSF proteomics identifies early changes in autosomal dominant Alzheimer's disease. Cell 2024; 187:6309-6326.e15. [PMID: 39332414 PMCID: PMC11531390 DOI: 10.1016/j.cell.2024.08.049] [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: 01/31/2024] [Revised: 07/02/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024]
Abstract
In this high-throughput proteomic study of autosomal dominant Alzheimer's disease (ADAD), we sought to identify early biomarkers in cerebrospinal fluid (CSF) for disease monitoring and treatment strategies. We examined CSF proteins in 286 mutation carriers (MCs) and 177 non-carriers (NCs). The developed multi-layer regression model distinguished proteins with different pseudo-trajectories between these groups. We validated our findings with independent ADAD as well as sporadic AD datasets and employed machine learning to develop and validate predictive models. Our study identified 137 proteins with distinct trajectories between MCs and NCs, including eight that changed before traditional AD biomarkers. These proteins are grouped into three stages: early stage (stress response, glutamate metabolism, neuron mitochondrial damage), middle stage (neuronal death, apoptosis), and late presymptomatic stage (microglial changes, cell communication). The predictive model revealed a six-protein subset that more effectively differentiated MCs from NCs, compared with conventional biomarkers.
Collapse
Affiliation(s)
- Yuanyuan Shen
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Gyujin Heo
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Aleksandra Beric
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Ciyang Wang
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Yueyao Wang
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Daniel Western
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Anh Do
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian Gordon
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jorge J Llibre-Guerra
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University St Louis, St Louis, MO 63110, USA
| | - Richard J Perrin
- Department of Pathology and Immunology, Washington University St. Louis, St. Louis, MO 63110, USA
| | - Dario Maschi
- Department of Cell Biology and Physiology, Washington University St. Louis, St. Louis, MO 63110, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Pau Pastor
- Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona 08916, Spain
| | - Alan E Renton
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ezequiel I Surace
- Laboratory of Neurodegenerative Diseases, Institute of Neurosciences (INEU-Fleni-CONICET), Buenos Aires, Argentina
| | - Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA 30307, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Ignacio Alvarez
- Department of Neurology, University Hospital Mútua de Terrassa and Fundació Docència i Recerca Mútua de Terrassa, Terrassa 08221, Barcelona, Spain
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich 80336, Germany; German Center for Neurodegenerative Diseases, site Munich, Munich 80336, Germany
| | - John M Ringman
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA 90033, USA
| | - Ricardo Francisco Allegri
- Department of Cognitive Neurology, Neuropsychology and Neuropsychiatry, FLENI, Buenos Aires, Argentina
| | - Nicholas Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Gregg S Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL 32224, USA
| | - Qisi Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Rawan Tarawneh
- The University of New Mexico, Albuquerque, NM 87131, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alison Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA; NeuroGenomics and Informatics, Washington University, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
6
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. iScience 2024; 27:110800. [PMID: 39310747 PMCID: PMC11416532 DOI: 10.1016/j.isci.2024.110800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement pathway dysregulation in PV cells drives disease pathogenesis, we have developed a transgenic line that permits cell-type specific overexpression of the schizophrenia-associated C4 gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific alterations in anxiety-like behavior and deficits in synaptic connectivity and excitability of PFC PV cells. Using a computational model, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that perturbations of this neuroimmune gene in fast-spiking neurons are especially detrimental to circuits associated with anxiety-like behavior. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics MS Program, Boston University, Boston, MA, USA
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, USA
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- NeuroTechnology Center (NTC), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
7
|
Bai Q, Shao E, Ma D, Jiao B, Scheetz SD, Hartnett-Scott KA, Ilin VA, Aizenman E, Kofler J, Burton EA. A human Tau expressing zebrafish model of progressive supranuclear palsy identifies Brd4 as a regulator of microglial synaptic elimination. Nat Commun 2024; 15:8195. [PMID: 39294122 PMCID: PMC11410960 DOI: 10.1038/s41467-024-52173-0] [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: 07/11/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is an incurable neurodegenerative disease characterized by 4-repeat (0N/4R)-Tau protein accumulation in CNS neurons. We generated transgenic zebrafish expressing human 0N/4R-Tau to investigate PSP pathophysiology. Tau zebrafish replicated multiple features of PSP, including: decreased survival; hypokinesia; impaired optokinetic responses; neurodegeneration; neuroinflammation; synapse loss; and Tau hyperphosphorylation, misfolding, mislocalization, insolubility, truncation, and oligomerization. Using automated assays, we screened 147 small molecules for activity in rescuing neurological deficits in Tau zebrafish. (+)JQ1, a bromodomain inhibitor, improved hypokinesia, survival, microgliosis, and brain synapse elimination. A heterozygous brd4+/- mutant reducing expression of the bromodomain protein Brd4 similarly rescued these phenotypes. Microglial phagocytosis of synaptic material was decreased by (+)JQ1 in both Tau zebrafish and rat primary cortical cultures. Microglia in human PSP brains expressed Brd4. Our findings implicate Brd4 as a regulator of microglial synaptic elimination in tauopathy and provide an unbiased approach for identifying mechanisms and therapeutic targets in PSP.
Collapse
Affiliation(s)
- Qing Bai
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Enhua Shao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Denglei Ma
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Binxuan Jiao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Seth D Scheetz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Karen A Hartnett-Scott
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Vladimir A Ilin
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Elias Aizenman
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Edward A Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatrics Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
8
|
Shen FS, Liu C, Sun HZ, Chen XY, Xue Y, Chen L. Emerging evidence of context-dependent synapse elimination by phagocytes in the CNS. J Leukoc Biol 2024; 116:511-522. [PMID: 38700080 DOI: 10.1093/jleuko/qiae098] [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: 11/16/2023] [Revised: 03/09/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Precise synapse elimination is essential for the establishment of a fully developed neural circuit during brain development and higher function in adult brain. Beyond immune and nutrition support, recent groundbreaking studies have revealed that phagocytic microglia and astrocytes can actively and selectively eliminate synapses in normal and diseased brains, thereby mediating synapse loss and maintaining circuit homeostasis. Multiple lines of evidence indicate that the mechanisms of synapse elimination by phagocytic glia are not universal but rather depend on specific contexts and detailed neuron-glia interactions. The mechanism of synapse elimination by phagocytic glia is dependent on neuron-intrinsic factors and many innate immune and local apoptosis-related molecules. During development, microglial synapse engulfment in the visual thalamus is primarily influenced by the classic complement pathway, whereas in the barrel cortex, the fractalkine pathway is dominant. In Alzheimer's disease, microglia employ complement-dependent mechanisms for synapse engulfment in tauopathy and early β-amyloid pathology, but microglia are not involved in synapse loss at late β-amyloid stages. Phagocytic microglia also engulf synapses in a complement-dependent way in schizophrenia, anxiety, and stress. In addition, phagocytic astrocytes engulf synapses in a MEGF10-dependent way during visual development, memory, and stroke. Furthermore, the mechanism of a phenomenon that phagocytes selectively eliminate excitatory and inhibitory synapses is also emphasized in this review. We hypothesize that elucidating context-dependent synapse elimination by phagocytic microglia and astrocytes may reveal the molecular basis of synapse loss in neural disorders and provide a rationale for developing novel candidate therapies that target synapse loss and circuit homeostasis.
Collapse
Affiliation(s)
- Fang-Shuai Shen
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Cui Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Hui-Zhe Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Xin-Yi Chen
- Department of International Medicine, No. 16 Jiangsu Road, Shinan District, Affiliated Hospital of Qingdao University 266000, Qingdao, China
| | - Yan Xue
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| |
Collapse
|
9
|
Akinlaja YO, Nishiyama A. Glial modulation of synapse development and plasticity: oligodendrocyte precursor cells as a new player in the synaptic quintet. Front Cell Dev Biol 2024; 12:1418100. [PMID: 39258226 PMCID: PMC11385347 DOI: 10.3389/fcell.2024.1418100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/09/2024] [Indexed: 09/12/2024] Open
Abstract
Synaptic communication is an important process in the central nervous system that allows for the rapid and spatially specified transfer of signals. Neurons receive various synaptic inputs and generate action potentials required for information transfer, and these inputs can be excitatory or inhibitory, which collectively determines the output. Non-neuronal cells (glial cells) have been identified as crucial participants in influencing neuronal activity and synaptic transmission, with astrocytes forming tripartite synapses and microglia pruning synapses. While it has been known that oligodendrocyte precursor cells (OPCs) receive neuronal inputs, whether they also influence neuronal activity and synaptic transmission has remained unknown for two decades. Recent findings indicate that OPCs, too, modulate neuronal synapses. In this review, we discuss the roles of different glial cell types at synapses, including the recently discovered involvement of OPCs in synaptic transmission and synapse refinement, and discuss overlapping roles played by multiple glial cell types.
Collapse
Affiliation(s)
- Yetunde O Akinlaja
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Institute of Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
- Institute of Systems Genomics, University of Connecticut, Storrs, CT, United States
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Institute of Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
- Institute of Systems Genomics, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
10
|
Scott-Hewitt N, Mahoney M, Huang Y, Korte N, Yvanka de Soysa T, Wilton DK, Knorr E, Mastro K, Chang A, Zhang A, Melville D, Schenone M, Hartigan C, Stevens B. Microglial-derived C1q integrates into neuronal ribonucleoprotein complexes and impacts protein homeostasis in the aging brain. Cell 2024; 187:4193-4212.e24. [PMID: 38942014 DOI: 10.1016/j.cell.2024.05.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/08/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
Neuroimmune interactions mediate intercellular communication and underlie critical brain functions. Microglia, CNS-resident macrophages, modulate the brain through direct physical interactions and the secretion of molecules. One such secreted factor, the complement protein C1q, contributes to complement-mediated synapse elimination in both developmental and disease models, yet brain C1q protein levels increase significantly throughout aging. Here, we report that C1q interacts with neuronal ribonucleoprotein (RNP) complexes in an age-dependent manner. Purified C1q protein undergoes RNA-dependent liquid-liquid phase separation (LLPS) in vitro, and the interaction of C1q with neuronal RNP complexes in vivo is dependent on RNA and endocytosis. Mice lacking C1q have age-specific alterations in neuronal protein synthesis in vivo and impaired fear memory extinction. Together, our findings reveal a biophysical property of C1q that underlies RNA- and age-dependent neuronal interactions and demonstrate a role of C1q in critical intracellular neuronal processes.
Collapse
Affiliation(s)
- Nicole Scott-Hewitt
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Matthew Mahoney
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Youtong Huang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nils Korte
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - T Yvanka de Soysa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel K Wilton
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Emily Knorr
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kevin Mastro
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Allison Chang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Allison Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - David Melville
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Monica Schenone
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christina Hartigan
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Investigator, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
11
|
María Del Rocío PB, Palomares Bralo M, Vanhooydonck M, Hamerlinck L, D'haene E, Leimbacher S, Jacobs EZ, De Cock L, D'haenens E, Dheedene A, Malfait Z, Vantomme L, Silva A, Rooney K, Santos-Simarro F, Lleuger-Pujol R, García-Miñaúr S, Losantos-García I, Menten B, Gestri G, Ragge N, Sadikovic B, Bogaert E, Syx D, Callewaert B, Vergult S. Loss-of-function of the Zinc Finger Homeobox 4 ( ZFHX4) gene underlies a neurodevelopmental disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311381. [PMID: 39148819 PMCID: PMC11326360 DOI: 10.1101/2024.08.07.24311381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
8q21.11 microdeletions encompassing the gene encoding transcription factor ZFHX4, have previously been associated by us with a syndromic form of intellectual disability, hypotonia, decreased balance and hearing loss. Here, we report on 57 individuals, 52 probands and 5 affected family members, with protein truncating variants (n=36), (micro)deletions (n=20) or an inversion (n=1) affecting ZFHX4 with variable developmental delay and intellectual disability, distinctive facial characteristics, morphological abnormalities of the central nervous system, behavioral alterations, short stature, hypotonia, and occasionally cleft palate and anterior segment dysgenesis. The phenotypes associated with 8q21.11 microdeletions and ZFHX4 intragenic loss-of-function variants largely overlap, identifying ZFHX4 as the main driver for the microdeletion syndrome, although leukocyte-derived DNA shows a mild common methylation profile for (micro)deletions only. We identify ZFHX4 as a transcription factor that is increasingly expressed during human brain development and neuronal differentiation. Furthermore, ZFHX4 interacting factors identified via IP-MS in neural progenitor cells, suggest an important role for ZFHX4 in cellular and developmental pathways, especially during histone modifications, cytosolic transport and development. Additionally, using CUT&RUN, we observed that ZFHX4 binds with the promoter regions of genes with crucial roles in embryonic, neuron and axon development. Since loss-of-function variants in ZFHX4 are found with consistent dysmorphic facial features, we investigated whether the disruption of zfhx4 causes craniofacial abnormalities in zebrafish. First-generation (F0) zfhx4 crispant zebrafish, (mosaic) mutant for zfhx4 loss-of-function variants, have significantly shorter Meckel's cartilages and smaller ethmoid plates compared to control zebrafish. Furthermore, behavioral assays show a decreased movement frequency in the zfhx4 crispant zebrafish in comparison with control zebrafish larvae. Although further research is needed, our in vivo work suggests a role for zfhx4 in facial skeleton patterning, palatal development and behavior.
Collapse
Affiliation(s)
- Pérez Baca María Del Rocío
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - María Palomares Bralo
- CIBERER-ISCIII and INGEMM, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ITHACA- European Reference Network, Spain
| | - Michiel Vanhooydonck
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lisa Hamerlinck
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Eva D'haene
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sebastian Leimbacher
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Eva Z Jacobs
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laurenz De Cock
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Erika D'haenens
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Annelies Dheedene
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Zoë Malfait
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lies Vantomme
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ananilia Silva
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Kathleen Rooney
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, Canada
| | - Fernando Santos-Simarro
- Unit of Molecular Diagnostics and Clinical Genetics, Hospital Universitari Son Espases, Health Research Institute of the Balearic Islands (IdiSBa), Palma, Spain
| | - Roser Lleuger-Pujol
- Hereditary Cancer Program, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital; Precision Oncology Group (OncoGIR-Pro), Institut d'Investigació Biomèdica de Girona (IDIGBI), Girona, Spain
| | - Sixto García-Miñaúr
- CIBERER-ISCIII and INGEMM, Institute of Medical and Molecular Genetics, Hospital Universitario La Paz, Madrid, Spain
- ITHACA- European Reference Network, Spain
| | | | - Björn Menten
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Gaia Gestri
- University College London, London, England, Great Britain
| | - Nicola Ragge
- Birmingham Women's and Children's NHS Foundation Trust, Clinical Genetics Unit, Birmingham Womens Hospital, Lavender House, Mindelsohn Way, Edgbaston, Birmingham B15 2TG
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, Canada
| | - Elke Bogaert
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Delfien Syx
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sarah Vergult
- Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Deng Q, Wu C, Parker E, Liu TCY, Duan R, Yang L. Microglia and Astrocytes in Alzheimer's Disease: Significance and Summary of Recent Advances. Aging Dis 2024; 15:1537-1564. [PMID: 37815901 PMCID: PMC11272214 DOI: 10.14336/ad.2023.0907] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Alzheimer's disease, one of the most common forms of dementia, is characterized by a slow progression of cognitive impairment and neuronal loss. Currently, approved treatments for AD are hindered by various side effects and limited efficacy. Despite considerable research, practical treatments for AD have not been developed. Increasing evidence shows that glial cells, especially microglia and astrocytes, are essential in the initiation and progression of AD. During AD progression, activated resident microglia increases the ability of resting astrocytes to transform into reactive astrocytes, promoting neurodegeneration. Extensive clinical and molecular studies show the involvement of microglia and astrocyte-mediated neuroinflammation in AD pathology, indicating that microglia and astrocytes may be potential therapeutic targets for AD. This review will summarize the significant and recent advances of microglia and astrocytes in the pathogenesis of AD in three parts. First, we will review the typical pathological changes of AD and discuss microglia and astrocytes in terms of function and phenotypic changes. Second, we will describe microglia and astrocytes' physiological and pathological role in AD. These roles include the inflammatory response, "eat me" and "don't eat me" signals, Aβ seeding, propagation, clearance, synapse loss, synaptic pruning, remyelination, and demyelination. Last, we will review the pharmacological and non-pharmacological therapies targeting microglia and astrocytes in AD. We conclude that microglia and astrocytes are essential in the initiation and development of AD. Therefore, understanding the new role of microglia and astrocytes in AD progression is critical for future AD studies and clinical trials. Moreover, pharmacological, and non-pharmacological therapies targeting microglia and astrocytes, with specific studies investigating microglia and astrocyte-mediated neuronal damage and repair, may be a promising research direction for future studies regarding AD treatment and prevention.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Emily Parker
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
13
|
Lian P, Cai X, Wang C, Zhai H, Liu K, Yang X, Wu Y, Ma Z, Cao X, Xu Y. Identification and experimental validation of m7G-related molecular subtypes, immune signature, and feature genes in Alzheimer's disease. Heliyon 2024; 10:e33836. [PMID: 39027505 PMCID: PMC11255592 DOI: 10.1016/j.heliyon.2024.e33836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 06/01/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Background Studies has shown that N7-methylguanosine (m7G) modification plays a critical role in neurological diseases. However, the exact role and association of m7G with the immune microenvironment in Alzheimer's disease (AD) remain largely unknown and unexplored. Methods The study datasets comprised 667 AD samples and 503 control samples selected from eight datasets in the Gene Expression Omnibus database; m7G regulator genes were obtained from previous literature. The AD subtypes were identified by consensus clustering analysis according to m7G regulator genes. The clinical characteristics, immune infiltration, and biological functions of the AD subgroups were evaluated. A combination of different types of machine-learning algorithms were used for the identification of AD genes. We also assessed and validated the diagnostic performance of the identified genes via qRT-PCR, immunofluorescence, and immunohistochemical analyses. Results Two AD distinct subgroups, namely cluster A and cluster B, were identified. Cluster A had poor pathological progression and immune infiltration, representing a high-risk subgroup for AD. The differentially expressed genes of cluster A were enriched in immune and synapse-related pathways, suggesting that these genes probably contribute to AD progression by regulating immune-related pathways. Additionally, five feature genes (AEBP1, CARTPT, AK5, NPTX2, and COPG2IT1) were identified, which were used to construct a nomogram model with good ability to predict AD. The animal experiment analyses further confirmed that these feature genes were associated with AD development. Conclusion To the best of our knowledge, this is the first study to reveal close correlations among m7G RNA modification, the immune microenvironment, and the pathogenesis of AD. We also identified five feature genes associated with AD, further contributing to our understanding of the underlying mechanisms and potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Piaopiao Lian
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Cai
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cailin Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhai
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoran Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Baum ML, Wilton DK, Fox RG, Carey A, Hsu YHH, Hu R, Jäntti HJ, Fahey JB, Muthukumar AK, Salla N, Crotty W, Scott-Hewitt N, Bien E, Sabatini DA, Lanser TB, Frouin A, Gergits F, Håvik B, Gialeli C, Nacu E, Lage K, Blom AM, Eggan K, McCarroll SA, Johnson MB, Stevens B. CSMD1 regulates brain complement activity and circuit development. Brain Behav Immun 2024; 119:317-332. [PMID: 38552925 DOI: 10.1016/j.bbi.2024.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Complement proteins facilitate synaptic elimination during neurodevelopmental pruning, but neural complement regulation is not well understood. CUB and Sushi Multiple Domains 1 (CSMD1) can regulate complement activity in vitro, is expressed in the brain, and is associated with increased schizophrenia risk. Beyond this, little is known about CSMD1 including whether it regulates complement activity in the brain or otherwise plays a role in neurodevelopment. We used biochemical, immunohistochemical, and proteomic techniques to examine the regional, cellular, and subcellular distribution as well as protein interactions of CSMD1 in the brain. To evaluate whether CSMD1 is involved in complement-mediated synapse elimination, we examined Csmd1-knockout mice and CSMD1-knockout human stem cell-derived neurons. We interrogated synapse and circuit development of the mouse visual thalamus, a process that involves complement pathway activity. We also quantified complement deposition on synapses in mouse visual thalamus and on cultured human neurons. Finally, we assessed uptake of synaptosomes by cultured microglia. We found that CSMD1 is present at synapses and interacts with complement proteins in the brain. Mice lacking Csmd1 displayed increased levels of complement component C3, an increased colocalization of C3 with presynaptic terminals, fewer retinogeniculate synapses, and aberrant segregation of eye-specific retinal inputs to the visual thalamus during the critical period of complement-dependent refinement of this circuit. Loss of CSMD1 in vivo enhanced synaptosome engulfment by microglia in vitro, and this effect was dependent on activity of the microglial complement receptor, CR3. Finally, human stem cell-derived neurons lacking CSMD1 were more vulnerable to complement deposition. These data suggest that CSMD1 can function as a regulator of complement-mediated synapse elimination in the brain during development.
Collapse
Affiliation(s)
- Matthew L Baum
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; MD-PhD Program of Harvard & MIT, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel K Wilton
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rachel G Fox
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alanna Carey
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Han H Hsu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ruilong Hu
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Henna J Jäntti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jaclyn B Fahey
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Allie K Muthukumar
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nikkita Salla
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - William Crotty
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Nicole Scott-Hewitt
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth Bien
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - David A Sabatini
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Toby B Lanser
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arnaud Frouin
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Frederick Gergits
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Chrysostomi Gialeli
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-214 28 Malmö, Sweden; Cardiovascular Research - Translational Studies Research Group, Department of Clinical Sciences, Lund University, S-214 28 Malmö, Sweden
| | - Eugene Nacu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kasper Lage
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, S-214 28 Malmö, Sweden
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Matthew B Johnson
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, USA.
| |
Collapse
|
15
|
Hadler MD, Alle H, Geiger JRP. Parvalbumin interneuron cell-to-network plasticity: mechanisms and therapeutic avenues. Trends Pharmacol Sci 2024; 45:586-601. [PMID: 38763836 DOI: 10.1016/j.tips.2024.04.003] [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: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) and schizophrenia (SCZ) represent two major neuropathological conditions with a high disease burden. Despite their distinct etiologies, patients suffering from AD or SCZ share a common burden of disrupted memory functions unattended by current therapies. Recent preclinical analyses highlight cell-type-specific contributions of parvalbumin interneurons (PVIs), particularly the plasticity of their cellular excitability, towards intact neuronal network function (cell-to-network plasticity) and memory performance. Here we argue that deficits of PVI cell-to-network plasticity may underlie memory deficits in AD and SCZ, and we explore two therapeutic avenues: the targeting of PVI-specific neuromodulation, including by neuropeptides, and the recruitment of network synchrony in the gamma frequency range (40 Hz) by external stimulation. We finally propose that these approaches be merged under consideration of recent insights into human brain physiology.
Collapse
Affiliation(s)
- Michael D Hadler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R P Geiger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Xing Y, Shi H, Gao X, Zhu X, Zhang D, Fang L, Wang J, Liu C, Wu D, Wang X, Min W. Walnut-Derived Peptides Alleviate Learning and Memory Impairments in a Mice Model via Inhibition of Microglia Phagocytose Synapses. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38853533 DOI: 10.1021/acs.jafc.4c01201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Microglia phagocytose synapses have an important effect on the pathogenesis of neurological disorders. Here, we investigated the neuroprotective effects of the walnut-derived peptide, TWLPLPR(TW-7), against LPS-induced cognitive deficits in mice and explored the underlying C1q-mediated microglia phagocytose synapses mechanisms in LPS-treated HT22 cells. The MWM showed that TW-7 improved the learning and memory capacity of the LPS-injured mice. Both transmission electron microscopy and immunofluorescence analysis illustrated that synaptic density and morphology were increased while associated with the decreased colocalized synapses with C1q. Immunohistochemistry and immunofluorescence demonstrated that TW-7 effectively reduced the microglia phagocytosis of synapses. Subsequently, overexpression of C1q gene plasmid was used to verify the contribution of the TW-7 via the classical complement pathway-regulated mitochondrial function-mediated microglia phagocytose synapses in LPS-treated HT22 cells. These data suggested that TW-7 improved the learning and memory capability of LPS-induced cognitively impaired mice through a mechanism associated with the classical complement pathway-mediated microglia phagocytose synapse.
Collapse
Affiliation(s)
- Yihang Xing
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Haoyuan Shi
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Xi Gao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Xinyu Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Dingwen Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Li Fang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Ji Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Chunlei Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Dan Wu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Xiyan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Weihong Min
- College of Food and Health, Zhejiang A&F University, Hangzhou 311300, P. R. China
| |
Collapse
|
17
|
Xu B, Ling Y, Liu L, Liu Y, Lin Y, Lyu J, Zhang Y. Potential prognostic value of CSF-targeted proteomics across the Alzheimer's disease continuum. BMC Geriatr 2024; 24:501. [PMID: 38844858 PMCID: PMC11157758 DOI: 10.1186/s12877-024-05104-z] [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: 02/04/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Core biomarkers for Alzheimer's disease (AD), such as Aβ42 and tau, have demonstrated high prognostic accuracy but do not fully capture the complex pathophysiology of AD. In this study, our objective was to identify novel cerebrospinal fluid (CSF) biomarkers using proteomics across the entire AD continuum to predict conversion to AD and explore their involvement in AD pathogenesis. METHODS A cohort of 186 cognitively normal (CN), 127 subjective memory complaint (SMC), 79 early mild cognitive impairment (EMCI), 249 late MCI (LMCI), and 132 AD individuals was analyzed, with a follow-up period of over 3 years for non-AD participants. CSF 65 peptides, as well as hippocampal and entorhinal volumes were analyzed, and cognitive function was evaluated using the 13-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cog 13). Cox proportional hazards models and mediation analysis were performed to investigate associations and causal relationships. RESULTS During the follow-up, approximately one-fourth (146/580) of the non-AD participants progressed to AD. After adjusting for baseline diagnosis (CN to LMCI) and other variables, multivariable Cox regression analysis identified three peptides (VAELEDEK, VSFELFADK, and VVSSIEQK) as significant predictors of conversion to AD. Incorporating these three peptides into the initial model significantly improved the C-statistic from 0.82 to 0.85 for predicting AD conversion, surpassing the predictive ability of Aβ42 and P-tau. Moreover, hippocampal and entorhinal volumes mediated 30.3-53.8% of the association between the three peptides and ADAS-Cog 13 scores. CONCLUSIONS These findings underscore the potential of these three peptides as robust prognostic biomarker candidates for AD conversion across the entire AD continuum, with a mechanism involving the mediation of hippocampal and entorhinal volumes.
Collapse
Affiliation(s)
- Bingdong Xu
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Guangzhou, Guangdong, 510632, P.R. China
| | - Yitong Ling
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Guangzhou, Guangdong, 510632, P.R. China
| | - Leiyuan Liu
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Guangzhou, Guangdong, 510632, P.R. China
| | - Yujun Liu
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Guangzhou, Guangdong, 510632, P.R. China
| | - Yingze Lin
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Guangzhou, Guangdong, 510632, P.R. China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China
| | - Yusheng Zhang
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Guangzhou, Guangdong, 510632, P.R. China.
| |
Collapse
|
18
|
Gu D, Xia Y, Ding Z, Qian J, Gu X, Bai H, Jiang M, Yao D. Inflammation in the Peripheral Nervous System after Injury. Biomedicines 2024; 12:1256. [PMID: 38927464 PMCID: PMC11201765 DOI: 10.3390/biomedicines12061256] [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: 04/28/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Nerve injury is a common condition that occurs as a result of trauma, iatrogenic injury, or long-lasting stimulation. Unlike the central nervous system (CNS), the peripheral nervous system (PNS) has a strong capacity for self-repair and regeneration. Peripheral nerve injury results in the degeneration of distal axons and myelin sheaths. Macrophages and Schwann cells (SCs) can phagocytose damaged cells. Wallerian degeneration (WD) makes the whole axon structure degenerate, creating a favorable regenerative environment for new axons. After nerve injury, macrophages, neutrophils and other cells are mobilized and recruited to the injury site to phagocytose necrotic cells and myelin debris. Pro-inflammatory and anti-inflammatory factors involved in the inflammatory response provide a favorable microenvironment for peripheral nerve regeneration and regulate the effects of inflammation on the body through relevant signaling pathways. Previously, inflammation was thought to be detrimental to the body, but further research has shown that appropriate inflammation promotes nerve regeneration, axon regeneration, and myelin formation. On the contrary, excessive inflammation can cause nerve tissue damage and pathological changes, and even lead to neurological diseases. Therefore, after nerve injury, various cells in the body interact with cytokines and chemokines to promote peripheral nerve repair and regeneration by inhibiting the negative effects of inflammation and harnessing the positive effects of inflammation in specific ways and at specific times. Understanding the interaction between neuroinflammation and nerve regeneration provides several therapeutic ideas to improve the inflammatory microenvironment and promote nerve regeneration.
Collapse
Affiliation(s)
- Dandan Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Yiming Xia
- Medical School, Nantong University, Nantong 226001, China
| | - Zihan Ding
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Jiaxi Qian
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Xi Gu
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Huiyuan Bai
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Maorong Jiang
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| | - Dengbing Yao
- School of Life Sciences, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China (H.B.)
| |
Collapse
|
19
|
Huo A, Wang J, Li Q, Li M, Qi Y, Yin Q, Luo W, Shi J, Cong Q. Molecular mechanisms underlying microglial sensing and phagocytosis in synaptic pruning. Neural Regen Res 2024; 19:1284-1290. [PMID: 37905877 PMCID: PMC11467947 DOI: 10.4103/1673-5374.385854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/08/2023] [Accepted: 08/03/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Microglia are the main non-neuronal cells in the central nervous system that have important roles in brain development and functional connectivity of neural circuits. In brain physiology, highly dynamic microglial processes are facilitated to sense the surrounding environment and stimuli. Once the brain switches its functional states, microglia are recruited to specific sites to exert their immune functions, including the release of cytokines and phagocytosis of cellular debris. The crosstalk of microglia between neurons, neural stem cells, endothelial cells, oligodendrocytes, and astrocytes contributes to their functions in synapse pruning, neurogenesis, vascularization, myelination, and blood-brain barrier permeability. In this review, we highlight the neuron-derived "find-me," "eat-me," and "don't eat-me" molecular signals that drive microglia in response to changes in neuronal activity for synapse refinement during brain development. This review reveals the molecular mechanism of neuron-microglia interaction in synaptic pruning and presents novel ideas for the synaptic pruning of microglia in disease, thereby providing important clues for discovery of target drugs and development of nervous system disease treatment methods targeting synaptic dysfunction.
Collapse
Affiliation(s)
- Anran Huo
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiali Wang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Qi Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Mengqi Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Yuwan Qi
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Qiao Yin
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Weifeng Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jijun Shi
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qifei Cong
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
20
|
Beiter RM, Sheehan PW, Schafer DP. Microglia phagocytic mechanisms: Development informing disease. Curr Opin Neurobiol 2024; 86:102877. [PMID: 38631077 PMCID: PMC11162951 DOI: 10.1016/j.conb.2024.102877] [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: 12/15/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
Microglia are tissue-resident macrophages and professional phagocytes of the central nervous system (CNS). In development, microglia-mediated phagocytosis is important for sculpting the cellular architecture. This includes the engulfment of dead/dying cells, pruning extranumerary synapses and axons, and phagocytosing fragments of myelin sheaths. Intriguingly, these developmental phagocytic mechanisms by which microglia sculpt the CNS are now appreciated as important for eliminating synapses, myelin, and proteins during neurodegeneration. Here, we discuss parallels between neurodevelopment and neurodegeneration, which highlights how development is informing disease. We further discuss recent advances and challenges towards therapeutically targeting these phagocytic pathways and how we can leverage development to overcome these challenges.
Collapse
Affiliation(s)
- Rebecca M Beiter
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Patrick W Sheehan
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
21
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.575409. [PMID: 38328248 PMCID: PMC10849664 DOI: 10.1101/2024.01.27.575409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement dysregulation - a prevalent locus of brain disease etiology - in PV cells may drive disease pathogenesis, we have developed a transgenic mouse line that permits cell-type specific overexpression of the schizophrenia-associated complement component 4 (C4) gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific behavioral alterations and concomitant deficits in synaptic connectivity and excitability of PV cells of the prefrontal cortex. Using a computational network, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that C4 perturbations in fast-spiking neurons are more harmful to brain function than pan-neuronal alterations. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, Massachusetts, United States
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts, United States
- Hearing Research Center, Boston University, Boston, Massachusetts, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
- Bioinformatics MS Program, Boston University, Boston, MA, United States
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, United States
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, United States
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, United States
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, Massachusetts, United States
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts, United States
- Hearing Research Center, Boston University, Boston, Massachusetts, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| |
Collapse
|
22
|
Wang S, Xie S, Zheng Q, Zhang Z, Wang T, Zhang G. Biofluid biomarkers for Alzheimer's disease. Front Aging Neurosci 2024; 16:1380237. [PMID: 38659704 PMCID: PMC11039951 DOI: 10.3389/fnagi.2024.1380237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease, with a complex pathogenesis and an irreversible course. Therefore, the early diagnosis of AD is particularly important for the intervention, prevention, and treatment of the disease. Based on the different pathophysiological mechanisms of AD, the research progress of biofluid biomarkers are classified and reviewed. In the end, the challenges and perspectives of future research are proposed.
Collapse
Affiliation(s)
- Sensen Wang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Sitan Xie
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Qinpin Zheng
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Zhihui Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| |
Collapse
|
23
|
Massa F, Martinuzzo C, Gómez de San José N, Pelagotti V, Kreshpa W, Abu-Rumeileh S, Barba L, Mattioli P, Orso B, Brugnolo A, Girtler N, Vigo T, Arnaldi D, Serrati C, Uccelli A, Morbelli S, Chincarini A, Otto M, Pardini M. Cerebrospinal fluid NPTX2 changes and relationship with regional brain metabolism metrics across mild cognitive impairment due to Alzheimer's disease. J Neurol 2024; 271:1999-2009. [PMID: 38157030 DOI: 10.1007/s00415-023-12154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Neuronal pentraxin-2 (NPTX2), crucial for synaptic functioning, declines in cerebrospinal fluid (CSF) as cognition deteriorates. The variations of CSF NPTX2 across mild cognitive impairment (MCI) due to Alzheimer's disease (AD) and its association with brain metabolism remain elusive, albeit relevant for patient stratification and pathophysiological insights. METHODS We retrospectively analyzed 49 MCI-AD patients grouped by time until dementia (EMCI, n = 34 progressing within 2 years; LMCI, n = 15 progressing later/stable at follow-up). We analyzed demographic variables, cognitive status (MMSE score), and CSF NPTX2 levels using a commercial ELISA assay in EMCI, LMCI, and a control group of age-/sex-matched individuals with other non-dementing disorders (OND). Using [18F]FDG PET scans for voxel-based analysis, we explored correlations between regional brain metabolism metrics and CSF NPTX2 levels in MCI-AD patients, accounting for age. RESULTS Baseline and follow-up MMSE scores were lower in LMCI than EMCI (p value = 0.006 and p < 0.001). EMCI exhibited significantly higher CSF NPTX2 values than both LMCI (p = 0.028) and OND (p = 0.006). We found a significant positive correlation between NPTX2 values and metabolism of bilateral precuneus in MCI-AD patients (p < 0.005 at voxel level, p < 0.05 with family-wise error correction at the cluster level). CONCLUSIONS Higher CSF NPTX2 in EMCI compared to controls and LMCI suggests compensatory synaptic responses to initial AD pathology. Disease progression sees these mechanisms overwhelmed, lowering CSF NPTX2 approaching dementia. Positive CSF NPTX2 correlation with precuneus glucose metabolism links to AD-related metabolic changes across MCI course. These findings posit CSF NPTX2 as a promising biomarker for both AD staging and progression risk stratification.
Collapse
Affiliation(s)
- Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Caterina Martinuzzo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | | | - Virginia Pelagotti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Wendy Kreshpa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Lorenzo Barba
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Pietro Mattioli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Beatrice Orso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Andrea Brugnolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicola Girtler
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Dario Arnaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Science (DISSAL), University of Genoa, Genoa, Italy
| | - Andrea Chincarini
- National Institute of Nuclear Physics (INFN), Genoa Section, Genoa, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
24
|
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
|
25
|
Wu M, Chen K, Zhao Y, Jiang M, Bao B, Yu W, Chen Z, Yin X. Normobaric hyperoxia alleviates complement C3-mediated synaptic pruning and brain injury after intracerebral hemorrhage. CNS Neurosci Ther 2024; 30:e14694. [PMID: 38532579 PMCID: PMC10966135 DOI: 10.1111/cns.14694] [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: 11/22/2023] [Revised: 02/02/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a common cerebrovascular disease, and the complement cascade exacerbates brain injury after ICH. As the most abundant component of the complement system, complement component 3 (C3) plays essential roles in all three complement pathways. However, the effects of C3 on neurological impairment and brain injury in ICH patients and the related mechanism have not been fully elucidated. Normobaric hyperoxia (NBO) is regarded as a treatment for ICH patients, and recent clinical studies also have confirmed the neuroprotective role of NBO against acute ICH-mediated brain damage, but the underlying mechanism still remains elusive. AIMS In the present study, we investigated the effects of complement C3 on NBO-treated ICH patients and model mice, and the underlying mechanism of NBO therapy in ICH-mediated brain injury. RESULTS Hemorrhagic injury resulted in the high plasma C3 levels in ICH patients, and the plasma C3 levels were closely related to hemorrhagic severity and clinical outcomes after ICH. BO treatment alleviated neurologic impairments and rescued the hemorrhagic-induced increase in plasma C3 levels in ICH patients and model mice. Moreover, the results indicated that NBO exerted its protective effects of on brain injury after ICH by downregulating the expression of C3 in microglia and alleviating microglia-mediated synaptic pruning. CONCLUSIONS Our results revealed that NBO exerts its neuroprotective effects by reducing C3-mediated synaptic pruning, which suggested that NBO therapy could be used for the clinical treatment of ICH.
Collapse
Affiliation(s)
- Moxin Wu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Kai Chen
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yasong Zhao
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Bing Bao
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangChina
| | - Wenmin Yu
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical SciencesJiujiang UniversityJiujiangJiangxiChina
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangChina
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangChina
| |
Collapse
|
26
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
27
|
Guo Z, Hong X, Wang X, Chen W, Guo Z. Association of reduced cerebrospinal fluid NPTX2 levels with postoperative delirium in patients undergoing knee/hip replacement: a prospective cohort study. Aging Clin Exp Res 2024; 36:42. [PMID: 38367123 PMCID: PMC10874313 DOI: 10.1007/s40520-023-02670-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/21/2023] [Indexed: 02/19/2024]
Abstract
BACKGROUND Postoperative delirium (POD) is a common complication with poor prognosis in the elderly, but its mechanism has not been fully elucidated. There is evidence that the changes in synaptic activity in the brain are closely related to the occurrence of POD. And neuronal pentraxin 2 (NPTX2) can regulate synaptic activity in vivo. AIMS This study aims to explore whether decreased NPTX2 levels affects POD and whether the cerebrospinal fluid (CSF) biomarkers of POD mediate this association. METHODS In this prospective cohort study, we interviewed patients with knee/hip replacement 1 day before surgery to collect patient information and assess their cognitive function. CSF was extracted for measuring the CSF levels of NPTX2 and other POD biomarkers on the day of surgery. And postoperative follow-up visits were performed 1-7 days after surgery. RESULTS Finally, 560 patients were included in the study. The patients were divided into POD group and NPOD (non-POD) group. The POD group had a median age of 80 years, a female proportion of 45%, a median BMI of 24.1 kg/m2, and a median years of education of 9 years. The Mann-Whitney U test showed that CSF NPTX2 levels were significantly lower in POD group, compared with the NPOD group (P < 0.05). Univariate binary logistic regression analysis showed that reduced CSF levels of NPTX2 protected against POD (crude OR = 0.994, 95% CI 0.993-0.995, P < 0.001). The receiver-operating characteristic (ROC) curve indicated that CSF NPTX2 level had high predictive value for POD. Mediation analyses showed that CSF T-tau (mediating proportion = 21%) and P-tau (mediating proportion = 29%) had significant mediating effects on the association between CSF NPTX2 and POD. CONCLUSION CSF NPTX2 levels were associated with the occurrence of POD. Low CSF NPTX2 levels may be an independent protective factor for POD. CSF T-tau and P-tau could mediate the association between CSF NPTX2 and POD occurrence. CLINICAL TRIAL REGISTRATION The trial registration number (TRN): ChiCTR2200064740, Date of Registration: 2022-10-15.
Collapse
Affiliation(s)
- Zongxiao Guo
- Department of Orthopedic Surgery, Hai'an People's Hospital, Haian, China
| | - Xiaoli Hong
- Department of Orthopedic Surgery, Hai'an People's Hospital, Haian, China
| | - Xiang Wang
- Department of Anesthesiology, Hai'an People's Hospital, Haian, China
| | - Weiguo Chen
- Department of Anesthesiology, Hai'an People's Hospital, Haian, China
| | - Zongfeng Guo
- Department of Anesthesiology, Hai'an People's Hospital, Haian, China.
| |
Collapse
|
28
|
Das S, van Engelen MPE, Goossens J, Jacobs D, Bongers B, Fieldhouse JLP, Pijnenburg YAL, Teunissen CE, Vanmechelen E, Verberk IMW. The use of synaptic biomarkers in cerebrospinal fluid to differentiate behavioral variant of frontotemporal dementia from primary psychiatric disorders and Alzheimer's disease. Alzheimers Res Ther 2024; 16:34. [PMID: 38355535 PMCID: PMC10865562 DOI: 10.1186/s13195-024-01409-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/04/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Lack of early molecular biomarkers in sporadic behavioral variants of frontotemporal dementia (bvFTD) and its clinical overlap with primary psychiatric disorders (PPD) hampers its diagnostic distinction. Synaptic dysfunction is an early feature in bvFTD and identification of specific biomarkers might improve its diagnostic accuracy. Our goal was to understand the differential diagnostic potential of cerebrospinal fluid (CSF) synaptic biomarkers in bvFTD versus PPD and their specificity towards bvFTD compared with Alzheimer's disease (AD) and controls. Additionally, we explored the association of CSF synaptic biomarkers with social cognition, cognitive performance, and disease severity in these clinical groups. METHODS Participants with probable bvFTD (n = 57), PPD (n = 71), AD (n = 60), and cognitively normal controls (n = 39) with available CSF, cognitive tests, and disease severity as frontotemporal lobar degeneration-modified clinical dementia rating scale (FTLD-CDR) were included. In a subset of bvFTD and PPD cases, Ekman 60 faces test scores for social cognition were available. CSF synaptosomal-associated protein 25 (SNAP25), neurogranin (Ng), neuronal pentraxin 2 (NPTX2), and glutamate receptor 4 (GluR4) were measured, along with neurofilament light (NfL), and compared between groups using analysis of covariance (ANCOVA) and logistic regression. Diagnostic accuracy was assessed using ROC analyses, and biomarker panels were selected using Wald's backward selection. Correlations with cognitive measures were performed using Pearson's partial correlation analysis. RESULTS NPTX2 concentrations were lower in the bvFTD group compared with PPD (p < 0.001) and controls (p = 0.003) but not compared with AD. Concentrations of SNAP25 (p < 0.001) and Ng (p < 0.001) were elevated in patients with AD versus those with bvFTD and controls. The modeled panel for differential diagnosis of bvFTD versus PPD consisted of NfL and NPTX2 (AUC = 0.96, CI: 0.93-0.99, p < 0.001). In bvFTD versus AD, the modeled panel consisted of NfL, SNAP25, Ng, and GluR4 (AUC = 0.86, CI: 0.79-0.92, p < 0.001). In bvFTD, lower NPTX2 (Pearson's r = 0.29, p = 0.036) and GluR4 (Pearson's r = 0.34, p = 0.014) concentrations were weakly associated with worse performance of total cognitive score. Lower GluR4 concentrations were also associated with worse MMSE scores (Pearson's r = 0.41, p = 0.002) as well as with worse executive functioning (Pearson's r = 0.36, p = 0.011) in bvFTD. There were no associations between synaptic markers and social cognition or disease severity in bvFTD. CONCLUSION Our findings of involvement of NTPX2 in bvFTD but not PPD contribute towards better understanding of bvFTD disease pathology.
Collapse
Affiliation(s)
- Shreyasee Das
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- ADx NeuroSciences, Technologiepark-Zwijnaarde 6, 9052, Gent, Belgium
| | - Marie-Paule E van Engelen
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Julie Goossens
- ADx NeuroSciences, Technologiepark-Zwijnaarde 6, 9052, Gent, Belgium
| | - Dirk Jacobs
- ADx NeuroSciences, Technologiepark-Zwijnaarde 6, 9052, Gent, Belgium
| | - Bram Bongers
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Jay L P Fieldhouse
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Yolande A L Pijnenburg
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Charlotte E Teunissen
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | | | - Inge M W Verberk
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands.
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands.
| |
Collapse
|
29
|
Magisetty J, Gadiraju B, Kondreddy V. Genomic analysis in the colon tissues of omega-3 fatty acid-treated rats identifies novel gene signatures implicated in ulcerative colitis. Int J Biol Macromol 2024; 258:128867. [PMID: 38123036 DOI: 10.1016/j.ijbiomac.2023.128867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/11/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Several long-term intervention trials only studied the ex vivo immunological function to elucidate the beneficial mechanisms of n-3 polyunsaturated fatty acids (PUFA) in the ulcerative colitis (UC). An unbiased whole-transcriptome analysis would be more valuable to obtain a comprehensive understanding of the processes and genes regulated by n-3 PUFA in vivo. In this study, we have performed microarray analysis in the colon tissues of dextran sulfate sodium (DSS)-induced UC in rats supplemented with n-6 PUFA, n-3PUFA and long-chain n-3PUFA (LC-n3PUFA). We have identified the novel gene signatures previously not linked to colitis such as Etv3, Clec4d, CD180, CD72, Megf11, and Angptl4 which are most downregulated in both n-3PUFA and LC-n3PUFA groups compared to the n-6PUFA group. The most upregulated genes were Nr1i3, Nptx2, and Zfp810 in both n-3PUFA and LC-n3PUFA groups. The RT-PCR analysis confirmed similar results. Interestingly, LPS treatment in macrophages upregulated the Megf11, Etv3, CD180, and Angptl4, and correlated with increased secretion of cytokines. Gene silencing of Etv3, Megf11, and CD180 in rats using intravascular delivery of siRNA-lipoparticles attenuated the DSS-induced ulceration and mucosal damage. Thus, our genome-wide microarray analysis identified novel genes regulated by omega-3 PUFA and offers new drug targets that could prevent or reduce UC.
Collapse
Affiliation(s)
- Jhansi Magisetty
- Department of Biochemistry, Central Food Technological Research Institute, Mysore 570020, India
| | - Bhavani Gadiraju
- Center for Lipid Science & Technology, The Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India
| | - Vijay Kondreddy
- Center for Lipid Science & Technology, The Indian Institute of Chemical Technology, Tarnaka, Hyderabad 500007, India.
| |
Collapse
|
30
|
Wen L, Bi D, Shen Y. Complement-mediated synapse loss in Alzheimer's disease: mechanisms and involvement of risk factors. Trends Neurosci 2024; 47:135-149. [PMID: 38129195 DOI: 10.1016/j.tins.2023.11.010] [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: 06/20/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
The complement system is increasingly recognized as a key player in the synapse loss and cognitive impairments observed in Alzheimer's disease (AD). In particular, the process of complement-dependent synaptic pruning through phagocytosis is over-activated in AD brains, driving detrimental excessive synapse elimination and contributing to synapse loss, which is the strongest neurobiological correlate of cognitive impairments in AD. Herein we review recent advances in characterizing complement-mediated synapse loss in AD, summarize the underlying mechanisms, and discuss the possible involvement of AD risk factors such as aging and various risk genes. We conclude with an overview of key questions that remain to be addressed.
Collapse
Affiliation(s)
- Lang Wen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Danlei Bi
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yong Shen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
31
|
Shen Y, Ali M, Timsina J, Wang C, Do A, Western D, Liu M, Gorijala P, Budde J, Liu H, Gordon B, McDade E, Morris JC, Llibre-Guerra JJ, Bateman RJ, Joseph-Mathurin N, Perrin RJ, Maschi D, Wyss-Coray T, Pastor P, Goate A, Renton AE, Surace EI, Johnson ECB, Levey AI, Alvarez I, Levin J, Ringman JM, Allegri RF, Seyfried N, Day GS, Wu Q, Fernández MV, Ibanez L, Sung YJ, Cruchaga C. Systematic proteomics in Autosomal dominant Alzheimer's disease reveals decades-early changes of CSF proteins in neuronal death, and immune pathways. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.12.24301242. [PMID: 38260583 PMCID: PMC10802763 DOI: 10.1101/2024.01.12.24301242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background To date, there is no high throughput proteomic study in the context of Autosomal Dominant Alzheimer's disease (ADAD). Here, we aimed to characterize early CSF proteome changes in ADAD and leverage them as potential biomarkers for disease monitoring and therapeutic strategies. Methods We utilized Somascan® 7K assay to quantify protein levels in the CSF from 291 mutation carriers (MCs) and 185 non-carriers (NCs). We employed a multi-layer regression model to identify proteins with different pseudo-trajectories between MCs and NCs. We replicated the results using publicly available ADAD datasets as well as proteomic data from sporadic Alzheimer's disease (sAD). To biologically contextualize the results, we performed network and pathway enrichment analyses. Machine learning was applied to create and validate predictive models. Findings We identified 125 proteins with significantly different pseudo-trajectories between MCs and NCs. Twelve proteins showed changes even before the traditional AD biomarkers (Aβ42, tau, ptau). These 125 proteins belong to three different modules that are associated with age at onset: 1) early stage module associated with stress response, glutamate metabolism, and mitochondria damage; 2) the middle stage module, enriched in neuronal death and apoptosis; and 3) the presymptomatic stage module was characterized by changes in microglia, and cell-to-cell communication processes, indicating an attempt of rebuilding and establishing new connections to maintain functionality. Machine learning identified a subset of nine proteins that can differentiate MCs from NCs better than traditional AD biomarkers (AUC>0.89). Interpretation Our findings comprehensively described early proteomic changes associated with ADAD and captured specific biological processes that happen in the early phases of the disease, fifteen to five years before clinical onset. We identified a small subset of proteins with the potentials to become therapy-monitoring biomarkers of ADAD MCs. Funding Proteomic data generation was supported by NIH: RF1AG044546.
Collapse
|
32
|
Krishnamurthy K, Pradhan RK. Emerging perspectives of synaptic biomarkers in ALS and FTD. Front Mol Neurosci 2024; 16:1279999. [PMID: 38249293 PMCID: PMC10796791 DOI: 10.3389/fnmol.2023.1279999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/01/2023] [Indexed: 01/23/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) are debilitating neurodegenerative diseases with shared pathological features like transactive response DNA-binding protein of 43 kDa (TDP-43) inclusions and genetic mutations. Both diseases involve synaptic dysfunction, contributing to their clinical features. Synaptic biomarkers, representing proteins associated with synaptic function or structure, offer insights into disease mechanisms, progression, and treatment responses. These biomarkers can detect disease early, track its progression, and evaluate therapeutic efficacy. ALS is characterized by elevated neurofilament light chain (NfL) levels in cerebrospinal fluid (CSF) and blood, correlating with disease progression. TDP-43 is another key ALS biomarker, its mislocalization linked to synaptic dysfunction. In FTD, TDP-43 and tau proteins are studied as biomarkers. Synaptic biomarkers like neuronal pentraxins (NPs), including neuronal pentraxin 2 (NPTX2), and neuronal pentraxin receptor (NPTXR), offer insights into FTD pathology and cognitive decline. Advanced technologies, like machine learning (ML) and artificial intelligence (AI), aid biomarker discovery and drug development. Challenges in this research include technological limitations in detection, variability across patients, and translating findings from animal models. ML/AI can accelerate discovery by analyzing complex data and predicting disease outcomes. Synaptic biomarkers offer early disease detection, personalized treatment strategies, and insights into disease mechanisms. While challenges persist, technological advancements and interdisciplinary efforts promise to revolutionize the understanding and management of ALS and FTD. This review will explore the present comprehension of synaptic biomarkers in ALS and FTD and discuss their significance and emphasize the prospects and obstacles.
Collapse
Affiliation(s)
- Karrthik Krishnamurthy
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | | |
Collapse
|
33
|
Vecchiarelli HA, Lopes LT, Paolicelli RC, Stevens B, Wake H, Tremblay MÈ. Synapse Regulation. ADVANCES IN NEUROBIOLOGY 2024; 37:179-208. [PMID: 39207693 DOI: 10.1007/978-3-031-55529-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are the resident immune cells of the brain. As such, they rapidly detect changes in normal brain homeostasis and accurately respond by fine-tuning in a tightly regulated manner their morphology, gene expression, and functional behavior. Depending on the nature of these changes, microglia can thicken and retract their processes, proliferate and migrate, release numerous signaling factors and compounds influencing neuronal physiology (e.g., cytokines and trophic factors), in addition to secreting proteases able to transform the extracellular matrix, and phagocytosing various types of cellular debris, etc. Because microglia also transform rapidly (on a time scale of minutes) during experimental procedures, studying these very special cells requires methods that are specifically non-invasive. The development of such methods has provided unprecedented insights into the roles of microglia during normal physiological conditions. In particular, transcranial two-photon in vivo imaging revealed that presumably "resting" microglia continuously survey the brain parenchyma with their highly motile processes, in addition to modulating their structural and functional interactions with neuronal circuits along the changes in neuronal activity and behavioral experience occurring throughout the lifespan. In this chapter, we will describe how surveillant microglia interact with synaptic elements and modulate the number, maturation, function, and plasticity of synapses in the healthy developing, mature, and aging brain, with consequences on neuronal activity, learning and memory, and the behavioral outcome.
Collapse
Affiliation(s)
| | | | - Rosa C Paolicelli
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
| | - Beth Stevens
- Department of Neurology, Harvard Medical School, Center for Life Science, Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
| | - Hiroaki Wake
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji-cho, Okazaki, Japan
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
34
|
Dejanovic B, Sheng M, Hanson JE. Targeting synapse function and loss for treatment of neurodegenerative diseases. Nat Rev Drug Discov 2024; 23:23-42. [PMID: 38012296 DOI: 10.1038/s41573-023-00823-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 11/29/2023]
Abstract
Synapse dysfunction and loss are hallmarks of neurodegenerative diseases that correlate with cognitive decline. However, the mechanisms and therapeutic strategies to prevent or reverse synaptic damage remain elusive. In this Review, we discuss recent advances in understanding the molecular and cellular pathways that impair synapses in neurodegenerative diseases, including the effects of protein aggregation and neuroinflammation. We also highlight emerging therapeutic approaches that aim to restore synaptic function and integrity, such as enhancing synaptic plasticity, preventing synaptotoxicity, modulating neuronal network activity and targeting immune signalling. We discuss the preclinical and clinical evidence for each strategy, as well as the challenges and opportunities for developing effective synapse-targeting therapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Morgan Sheng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jesse E Hanson
- Department of Neuroscience, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
35
|
Imrie G, Gray MB, Raghuraman V, Farhy-Tselnicker I. Gene Expression at the Tripartite Synapse: Bridging the Gap Between Neurons and Astrocytes. ADVANCES IN NEUROBIOLOGY 2024; 39:95-136. [PMID: 39190073 DOI: 10.1007/978-3-031-64839-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Astrocytes, a major class of glial cells, are an important element at the synapse where they engage in bidirectional crosstalk with neurons to regulate numerous aspects of neurotransmission, circuit function, and behavior. Mutations in synapse-related genes expressed in both neurons and astrocytes are central factors in a vast number of neurological disorders, making the proteins that they encode prominent targets for therapeutic intervention. Yet, while the roles of many of these synaptic proteins in neurons are well established, the functions of the same proteins in astrocytes are largely unknown. This gap in knowledge must be addressed to refine therapeutic approaches. In this chapter, we integrate multiomic meta-analysis and a comprehensive overview of current literature to show that astrocytes express an astounding number of genes that overlap with the neuronal and synaptic transcriptomes. Further, we highlight recent reports that characterize the expression patterns and potential novel roles of these genes in astrocytes in both physiological and pathological conditions, underscoring the importance of considering both cell types when investigating the function and regulation of synaptic proteins.
Collapse
Affiliation(s)
- Gillian Imrie
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Madison B Gray
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Vishnuvasan Raghuraman
- Department of Biology, Texas A&M University, College Station, TX, USA
- Department of Computer Science and Engineering, Texas A&M University, College Station, TX, USA
| | - Isabella Farhy-Tselnicker
- Department of Biology, Texas A&M University, College Station, TX, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, USA.
- Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
36
|
Fatima H, Rangwala HS, Riaz F, Rangwala BS, Siddiq MA. Breakthroughs in Alzheimer's Research: A Path to a More Promising Future? Ann Neurosci 2024; 31:63-70. [PMID: 38584978 PMCID: PMC10996869 DOI: 10.1177/09727531231187235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/21/2023] [Indexed: 04/09/2024] Open
Abstract
Background Alzheimer's disease (AD) is a widespread neurodegenerative disorder with a significant global impact, affecting approximately 50 million individuals, and projections estimate that up to 152 million people will be affected by 2050. AD is characterized by beta-amyloid plaques and tau tangles in the brain, leading to cognitive decline. Summary Recent research on AD has made significant strides, including the development of an "amyloid clock" biomarker that tracks AD progression through positron emission tomography (PET) scans. Surf4 and other genes have been discovered to play a role in regulating beta-amyloid toxicity, while inhibiting the enzyme hexokinase-2 has shown positive results in preclinical studies. New brain mapping techniques have identified early brain-based causes of cognitive changes in AD, and biomarkers such as neuronal pentraxin protein Nptx2 and astrocytic 7-subunit of the nicotinic acetylcholine receptors (7nAChRs) show potential for early detection. Other approaches, such as replenishing the enzyme Tip60, selectively degrading the modified protein p-p38 with PRZ-18002, and targeting the protein voltage-dependent anion channel-1 (VDAC1), have shown promise in enhancing cognitive function and preventing pathophysiological alterations linked to AD. Baseline blood samples and other biomarkers such as urine formic acid, p-tau 198, microRNAs, and glial fibrillary acidic protein (GFAP) have also been discovered for early detection and intervention of AD. Additionally, recent FDA approvals for medications such as aducanumab and lecanemab provide options for reducing AD symptoms and improving function, while clinical trials for dementia vaccines show promise for the nasal and beta-amyloid 40 vaccines as well as vaccinations targeting tau. Key Messages These advancements in AD research, including biomarker discovery and the development of disease-modifying treatments, are crucial steps towards improving the lives of those affected by AD and finding a cure for this debilitating disease.
Collapse
Affiliation(s)
- Hareer Fatima
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | | | - Faiza Riaz
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | | | | |
Collapse
|
37
|
Li D, Xie Q, Xie J, Ni M, Wang J, Gao Y, Wang Y, Tang Q. Cerebrospinal Fluid Proteomics Identifies Potential Biomarkers for Early-Onset Alzheimer's Disease. J Alzheimers Dis 2024; 100:261-277. [PMID: 38848183 DOI: 10.3233/jad-240022] [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] [Indexed: 06/09/2024]
Abstract
Background Early-onset Alzheimer's disease (EOAD) exhibits a notable degree of heterogeneity as compared to late-onset Alzheimer's disease (LOAD). The proteins and pathways contributing to the pathophysiology of EOAD still need to be completed and elucidated. Objective Using correlation network analysis and machine learning to analyze cerebrospinal fluid (CSF) proteomics data to identify potential biomarkers and pathways associated with EOAD. Methods We employed mass spectrometry to conduct CSF proteomic analysis using the data-independent acquisition method in a Chinese cohort of 139 CSF samples, including 40 individuals with normal cognition (CN), 61 patients with EOAD, and 38 patients with LOAD. Correlation network analysis of differentially expressed proteins was performed to identify EOAD-associated pathways. Machine learning assisted in identifying crucial proteins differentiating EOAD. We validated the results in an Western cohort and examined the proteins expression by enzyme-linked immunosorbent assay (ELISA) in additional 9 EOAD, 9 LOAD, and 9 CN samples from our cohort. Results We quantified 2,168 CSF proteins. Following adjustment for age and sex, EOAD exhibited a significantly greater number of differentially expressed proteins than LOAD compared to CN. Additionally, our data indicates that EOAD may exhibit more pronounced synaptic dysfunction than LOAD. Three potential biomarkers for EOAD were identified: SH3BGRL3, LRP8, and LY6 H, of which SH3BGRL3 also accurately classified EOAD in the Western cohort. LY6 H reduction was confirmed via ELISA, which was consistent with our proteomic results. Conclusions This study provides a comprehensive profile of the CSF proteome in EOAD and identifies three potential EOAD biomarker proteins.
Collapse
Affiliation(s)
- Dazhi Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiang Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jikui Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming Ni
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jinliang Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuru Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yaxin Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiqiang Tang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
38
|
Hou Q, Liu X, Feng M, Zhou Z. WR-PTXF, a novel short pentraxin, protects gut mucosal barrier and enhances the antibacterial activity in Carassius cuvieri × Carassius auratus red var. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105055. [PMID: 37690613 DOI: 10.1016/j.dci.2023.105055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
The pentraxin family is an evolutionarily conserved group that plays an important role in innate immunity. C-reactive protein (CRP) and serum amyloid P component (SAP) are classical members of the short pentraxins and are known to be the major acute phase proteins. In this work, we have cloned a novel pentraxin fusion protein, WR-PTXF, from Carassius cuvieri × Carassius auratus red var. In fish, the biological function of PTXF is essentially unknown. For this purpose, we report the identification and analysis of WR-PTXF and elucidate its role in the antibacterial innate immunity. WR-PTXF contains 224 amino acids and shares 79.8% and 23.0% sequence identities with crucian carp CRP and SAP, respectively. Blast analysis shows that WR-PTXF and goldfish PTXF had the highest similarity (97.3%). WR-PTXF is expressed in multiple tissues and is upregulated by Aeromonas hydrophila infection. WR-PTXF contains a short pentraxin domain and recombinant WR-PTXF protein (rWR-PTXF) can bind the A. hydrophila in a concentration-dependent manner. Further, rWR-PTXF displays apparent bacteriostatic activity against A. hydrophila in vitro by enhancing the uptake of the bound bacteria by host phagocytes. When introduced in vivo, rWR-PTXF not only protects the gut mucosa but also limits the colonization of A. hydrophila in systemic immune organs. Consistently, knockdown of WR-PTXF significantly promotes bacterial dissemination in the tissues of host. These results indicate that WR-PTXF is a classic pattern recognition molecule that exerts a protective effect against bacterial infection.
Collapse
Affiliation(s)
- Qian Hou
- Department of Nutrition, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiaofeng Liu
- Department of Nutrition, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Mengzhe Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Zejun Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
39
|
Danelon V, Garret-Thomson SC, Almo SC, Lee FS, Hempstead BL. Immune activation of the p75 neurotrophin receptor: implications in neuroinflammation. Front Mol Neurosci 2023; 16:1305574. [PMID: 38106879 PMCID: PMC10722190 DOI: 10.3389/fnmol.2023.1305574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
Despite structural similarity with other tumor necrosis factor receptor superfamily (TNFRSF) members, the p75 neurotrophin receptor (p75NTR, TNFR16) mediates pleiotropic biological functions not shared with other TNFRs. The high level of p75NTR expression in the nervous system instead of immune cells, its utilization of co-receptors, and its interaction with soluble dimeric, rather than soluble or cell-tethered trimeric ligands are all characteristics which distinguish it from most other TNFRs. Here, we compare these attributes to other members of the TNFR superfamily. In addition, we describe the recent evolutionary adaptation in B7-1 (CD80), an immunoglobulin (Ig) superfamily member, which allows engagement to neuronally-expressed p75NTR. B7-1-mediated binding to p75NTR occurs in humans and other primates, but not lower mammals due to specific sequence changes that evolved recently in primate B7-1. This discovery highlights an additional mechanism by which p75NTR can respond to inflammatory cues and trigger synaptic elimination in the brain through engagement of B7-1, which was considered to be immune-restricted. These observations suggest p75NTR does share commonality with other immune co-modulatory TNFR family members, by responding to immunoregulatory cues. The evolution of primate B7-1 to bind and elicit p75NTR-mediated effects on neuronal morphology and function are discussed in relationship to immune-driven modulation of synaptic actions during injury or inflammation.
Collapse
Affiliation(s)
- Victor Danelon
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | | | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Francis S. Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, United States
| | - Barbara L. Hempstead
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
40
|
Li Q. Pituitary-immune bidirectional crosstalk under systemic inflammation. PLoS Biol 2023; 21:e3002440. [PMID: 38113236 PMCID: PMC10729977 DOI: 10.1371/journal.pbio.3002440] [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] [Indexed: 12/21/2023] Open
Abstract
The pituitary gland responds to and modulates immune stress through the hypothalamus-pituitary-adrenal axis. A new study in PLOS Biology reveals unconventional bidirectional communication between hormone-producing cells and the immune system upon systemic inflammation.
Collapse
Affiliation(s)
- Qingyun Li
- Department of Neuroscience, Department of Genetics, Hope Center for Neurological Disorders, Center for Brain Immunology and Glia, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
41
|
Yan T, Wang R, Yao J, Luo M. Single-cell transcriptomic analysis reveals rich pituitary-Immune interactions under systemic inflammation. PLoS Biol 2023; 21:e3002403. [PMID: 38109308 PMCID: PMC10727439 DOI: 10.1371/journal.pbio.3002403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/26/2023] [Indexed: 12/20/2023] Open
Abstract
The pituitary represents an essential hub in the hypothalamus-pituitary-adrenal (HPA) axis. Pituitary hormone-producing cells (HPCs) release several hormones to regulate fundamental bodily functions under normal and stressful conditions. It is well established that the pituitary endocrine gland modulates the immune system by releasing adrenocorticotropic hormone (ACTH) in response to neuronal activation in the hypothalamus. However, it remains unclear how systemic inflammation regulates the transcriptomic profiles of pituitary HPCs. Here, we performed single-cell RNA-sequencing (scRNA-seq) of the mouse pituitary and revealed that upon inflammation, all major pituitary HPCs respond robustly in a cell type-specific manner, with corticotropes displaying the strongest reaction. Systemic inflammation also led to the production and release of noncanonical bioactive molecules, including Nptx2 by corticotropes, to modulate immune homeostasis. Meanwhile, HPCs up-regulated the gene expression of chemokines that facilitated the communication between the HPCs and immune cells. Together, our study reveals extensive interactions between the pituitary and immune system, suggesting multifaceted roles of the pituitary in mediating the effects of inflammation on many aspects of body physiology.
Collapse
Affiliation(s)
- Ting Yan
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Ruiyu Wang
- Chinese Institute for Brain Research, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
- PTN Graduate Program, School of Life Sciences, Peking University, Beijing, China
| | - Jingfei Yao
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Minmin Luo
- Chinese Institute for Brain Research, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Beijing, China
- New Cornerstone Science Laboratory, Shenzhen, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Li Y, Zhang S, Liu J, Zhang Y, Zhang N, Cheng Q, Zhang H, Wu X. The pentraxin family in autoimmune disease. Clin Chim Acta 2023; 551:117592. [PMID: 37832905 DOI: 10.1016/j.cca.2023.117592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
The pentraxins represent a family of multifunctional proteins composed of long and short pentamers. The latter includes serum amyloid P component (SAP) and C-reactive protein (CRP) whereas the former includes neuronal PTX1 and PTX2 (NPTX1 and NPTX2, respectively), PTX3 and PTX4. These serve as a bridge between adaptive immunity and innate immunity and a link between inflammation and immunity. Similarities and differences between long and short pentamers are examined and their roles in autoimmune disease are discussed. Increased CRP and PTX3 could indicate the activity of rheumatoid arthritis, systemic lupus erythematosus or other autoimmune diseases. Mechanistically, CRP and PTX3 may predict target organ injury, regulate bone metabolic immunity and maintain homeostasis as well as participate in vascular endothelial remodeling. Interestingly, PTX3 is pleiotropic, being involved in inflammation and tissue repair. Given the therapeutic potential of PTX3 and CRP, targeting these factors to exert a beneficial effect is the focus of research efforts. Unfortunately, studies on NPTX1, NPTX2, PTX4 and SAP are scarce and more research is clearly needed to elaborate their potential roles in autoimmune disease.
Collapse
Affiliation(s)
- Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Shouzan Zhang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, PR China
| | - Jingqi Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yudi Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, PR China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China.
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
43
|
Gomez-Arboledas A, Fonseca MI, Kramar E, Chu SH, Schartz N, Selvan P, Wood MA, Tenner AJ. C5aR1 signaling promotes region and age dependent synaptic pruning in models of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560234. [PMID: 37873302 PMCID: PMC10592845 DOI: 10.1101/2023.09.29.560234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
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 rescues the 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.
Collapse
Affiliation(s)
- Angela Gomez-Arboledas
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Maria I. Fonseca
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Enikö Kramar
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Shu-Hui Chu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Nicole Schartz
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Purnika Selvan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| |
Collapse
|
44
|
Rothschadl MJ, Sathyanesan M, Newton SS. Synergism of Carbamoylated Erythropoietin and Insulin-like Growth Factor-1 in Immediate Early Gene Expression. Life (Basel) 2023; 13:1826. [PMID: 37763230 PMCID: PMC10532867 DOI: 10.3390/life13091826] [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: 07/21/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Trophic factors are secreted proteins that can modulate neuronal integrity, structure, and function. Previous preclinical studies have shown synergistic effects on decreasing apoptosis and improving behavioral performance after stroke when combining two such trophic factors, erythropoietin (EPO) and insulin-like growth factor-1 (IGF-1). However, EPO can elevate the hematocrit level, which can be life-threatening for non-anemic individuals. A chemically engineered derivative of EPO, carbamoylated EPO (CEPO), does not impact hematological parameters but retains neurotrophic effects similar to EPO. To obtain insight into CEPO and IGF-1 combination signaling, we examined immediate early gene (IEG) expression after treatment with CEPO, IGF-1, or CEPO + IGF-1 in rat pheochromocytoma (PC-12) cells and found that combining CEPO and IGF-1 produced a synergistic increase in IEG expression. An in vivo increase in the protein expression of Npas4 and Nptx2 was also observed in the rat hippocampus. We also examined which kinase signaling pathways might be mediating these effects and found that while AKT inhibition did not alter the pattern of IEG expression, both ERK and JAK2 inhibition significantly decreased IEG expression. These results begin to define the molecular effects of combining CEPO and IGF-1 and indicate the potential for these trophic factors to produce positive, synergistic effects.
Collapse
Affiliation(s)
| | | | - Samuel S. Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (M.J.R.); (M.S.)
| |
Collapse
|
45
|
Farsi Z, Sheng M. Molecular mechanisms of schizophrenia: Insights from human genetics. Curr Opin Neurobiol 2023; 81:102731. [PMID: 37245257 DOI: 10.1016/j.conb.2023.102731] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/30/2023]
Abstract
Schizophrenia is a debilitating psychiatric disorder that affects millions of people worldwide; however, its etiology is poorly understood at the molecular and neurobiological levels. A particularly important advance in recent years is the discovery of rare genetic variants associated with a greatly increased risk of developing schizophrenia. These primarily loss-of-function variants are found in genes that overlap with those implicated by common variants and are involved in the regulation of glutamate signaling, synaptic function, DNA transcription, and chromatin remodeling. Animal models harboring mutations in these large-effect schizophrenia risk genes show promise in providing additional insights into the molecular mechanisms of the disease.
Collapse
Affiliation(s)
- Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
46
|
Aryal S, Bonanno K, Song B, Mani DR, Keshishian H, Carr SA, Sheng M, Dejanovic B. Deep proteomics identifies shared molecular pathway alterations in synapses of patients with schizophrenia and bipolar disorder and mouse model. Cell Rep 2023; 42:112497. [PMID: 37171958 DOI: 10.1016/j.celrep.2023.112497] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/10/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
Synaptic dysfunction is implicated in the pathophysiology of schizophrenia (SCZ) and bipolar disorder (BP). We use quantitative mass spectrometry to carry out deep, unbiased proteomic profiling of synapses purified from the dorsolateral prefrontal cortex of 35 cases of SCZ, 35 cases of BP, and 35 controls. Compared with controls, SCZ and BP synapses show substantial and similar proteomic alterations. Network analyses reveal upregulation of proteins associated with autophagy and certain vesicle transport pathways and downregulation of proteins related to synaptic, mitochondrial, and ribosomal function in the synapses of individuals with SCZ or BP. Some of the same pathways are similarly dysregulated in the synaptic proteome of mutant mice deficient in Akap11, a recently discovered shared risk gene for SCZ and BP. Our work provides biological insights into molecular dysfunction at the synapse in SCZ and BP and serves as a resource for understanding the pathophysiology of these disorders.
Collapse
Affiliation(s)
- Sameer Aryal
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin Bonanno
- The Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bryan Song
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - D R Mani
- The Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hasmik Keshishian
- The Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Steven A Carr
- The Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Borislav Dejanovic
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|