1
|
Khan H, Naseem T, Kaushik P, Narang J, Khan R, Panwar S, Parvez S. Decoding paradoxical links of cytokine markers in cognition: Cross talk between physiology, inflammaging, and Alzheimer's disease- related cognitive decline. Ageing Res Rev 2024; 101:102535. [PMID: 39374831 DOI: 10.1016/j.arr.2024.102535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Recent research has revolutionized our understanding of memory consolidation by emphasizing the critical role of astrocytes, microglia, and immune cells in through cytokine signaling. Cytokines, compact proteins, play pivotal roles in neuronal development, synaptic transmission, and normal aging. This review explores the cellular mechanisms contributing to cognitive decline in inflammaging and Alzheimer's disease, highlighting the paradoxical effects of most studied cytokines (IL-1, IL-6, TNF-α) in brain function, which act as a double-edged sword in brain physiology, acting both as facilitators of healthy cognitive function and as a potential contributor to cognitive decline.
Collapse
Affiliation(s)
- Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Talib Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Siddharth Panwar
- School of Computing and Electrical Engineering, Indian Institute of Technology, Mandi, Himachal Pradesh 175075, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
2
|
Yang L, Guttman L, Dawson VL, Dawson TM. Parthanatos: Mechanisms, modulation, and therapeutic prospects in neurodegenerative disease and stroke. Biochem Pharmacol 2024; 228:116174. [PMID: 38552851 PMCID: PMC11410548 DOI: 10.1016/j.bcp.2024.116174] [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: 01/21/2024] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Parthanatos is a cell death signaling pathway that has emerged as a compelling target for pharmaceutical intervention. It plays a pivotal role in the neuron loss and neuroinflammation that occurs in Parkinson's Disease (PD), Alzheimer's Disease (AD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS), and stroke. There are currently no treatments available to humans to prevent cell death in any of these diseases. This review provides an in-depth examination of the current understanding of the Parthanatos mechanism, with a particular focus on its implications in neuroinflammation and various diseases discussed herein. Furthermore, we thoroughly review potential intervention targets within the Parthanatos pathway. We dissect recent progress in inhibitory strategies, complimented by a detailed structural analysis of key Parthanatos executioners, PARP-1, AIF, and MIF, along with an assessment of their established inhibitors. We hope to introduce a new perspective on the feasibility of targeting components within the Parthanatos pathway, emphasizing its potential to bring about transformative outcomes in therapeutic interventions. By delineating therapeutic opportunities and known targets, we seek to emphasize the imperative of blocking Parthanatos as a precursor to developing disease-modifying treatments. This comprehensive exploration aims to catalyze a paradigm shift in our understanding of potential neurodegenerative disease therapeutics, advocating for the pursuit of effective interventions centered around Parthanatos inhibition.
Collapse
Affiliation(s)
- Liu Yang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren Guttman
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
3
|
Hok-A-Hin YS, Vermunt L, Peeters CF, van der Ende EL, de Boer SC, Meeter LH, van Swieten JC, Hu WT, Lleó A, Alcolea D, Engelborghs S, Sieben A, Chen-Plotkin A, Irwin DJ, van der Flier WM, Pijnenburg YA, Teunissen CE, del Campo M. Large-scale CSF proteome profiling identifies biomarkers for accurate diagnosis of Frontotemporal Dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.19.24312100. [PMID: 39228745 PMCID: PMC11370532 DOI: 10.1101/2024.08.19.24312100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Diagnosis of Frontotemporal dementia (FTD) and the specific underlying neuropathologies (frontotemporal lobar degeneration; FTLD- Tau and FTLD-TDP) is challenging, and thus fluid biomarkers are needed to improve diagnostic accuracy. We used proximity extension assays to analyze 665 proteins in cerebrospinal fluid (CSF) samples from a multicenter cohort including patients with FTD (n = 189), Alzheimer's Disease dementia (AD; n = 232), and cognitively unimpaired individuals (n = 196). In a subset, FTLD neuropathology was determined based on phenotype or genotype (FTLD-Tau = 87 and FTLD-TDP = 68). Forty three proteins were differentially regulated in FTD compared to controls and AD, reflecting axon development, regulation of synapse assembly, and cell-cell adhesion mediator activity pathways. Classification analysis identified a 14- and 13-CSF protein panel that discriminated FTD from controls (AUC: 0.96) or AD (AUC: 0.91). Custom multiplex panels confirmed the highly accurate discrimination between FTD and controls (AUCs > 0.96) or AD (AUCs > 0.88) in three validation cohorts, including one with autopsy confirmation (AUCs > 0.90). Six proteins were differentially regulated between FTLD-TDP and FTLD-Tau, but no reproducible classification model could be generated (AUC: 0.80). Overall, this study introduces novel FTD-specific biomarker panels with potential use in diagnostic setting.
Collapse
Affiliation(s)
- Yanaika S. Hok-A-Hin
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Carel F.W. Peeters
- Mathematical & Statistical Methods group – Biometris, Wageningen University & Research, Wageningen, The Netherlands
| | - Emma L. van der Ende
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Sterre C.M. de Boer
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, Australia
| | - Lieke H. Meeter
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John C. van Swieten
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William T. Hu
- Department of Neurology, Center for Neurodegenerative Diseases Research, Emory University School of Medicine, Atlanta, USA
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Neuroprotection and Neuromodulation Research Group (NEUR), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Brussels, Belgium
| | - Anne Sieben
- Lab of neuropathology, Neurobiobank, Institute Born-Bunge, Antwerp University, Edegem, Belgium
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wiesje M. van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Yolande A.L. Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Barcelonaßeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San PabloCEU, CEU Universities, Madrid, Spain
| |
Collapse
|
4
|
Rupar MJ, Hanson H, Rogers S, Botlick B, Trimmer S, Hickman JJ. Modelling the innate immune system in microphysiological systems. LAB ON A CHIP 2024; 24:3604-3625. [PMID: 38957150 PMCID: PMC11264333 DOI: 10.1039/d3lc00812f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/09/2024] [Indexed: 07/04/2024]
Abstract
This critical review aims to highlight how modeling of the immune response has adapted over time to utilize microphysiological systems. Topics covered here will discuss the integral components of the immune system in various human body systems, and how these interactions are modeled using these systems. Through the use of microphysiological systems, we have not only expanded on foundations of basic immune cell information, but have also gleaned insight on how immune cells work both independently and collaboratively within an entire human body system.
Collapse
Affiliation(s)
- Michael J Rupar
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| | - Hannah Hanson
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| | - Stephanie Rogers
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| | - Brianna Botlick
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| | - Steven Trimmer
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| | - James J Hickman
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826, USA.
| |
Collapse
|
5
|
Iannucci J, Dominy R, Bandopadhyay S, Arthur EM, Noarbe B, Jullienne A, Krkasharyan M, Tobin RP, Pereverzev A, Beevers S, Venkatasamy L, Souza KA, Jupiter DC, Dabney A, Obenaus A, Newell-Rogers MK, Shapiro LA. Traumatic brain injury alters the effects of class II invariant peptide (CLIP) antagonism on chronic meningeal CLIP + B cells, neuropathology, and neurobehavioral impairment in 5xFAD mice. J Neuroinflammation 2024; 21:165. [PMID: 38937750 PMCID: PMC11212436 DOI: 10.1186/s12974-024-03146-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: 01/19/2024] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a significant risk factor for Alzheimer's disease (AD), and accumulating evidence supports a role for adaptive immune B and T cells in both TBI and AD pathogenesis. We previously identified B cell and major histocompatibility complex class II (MHCII)-associated invariant chain peptide (CLIP)-positive B cell expansion after TBI. We also showed that antagonizing CLIP binding to the antigen presenting groove of MHCII after TBI acutely reduced CLIP + splenic B cells and was neuroprotective. The current study investigated the chronic effects of antagonizing CLIP in the 5xFAD Alzheimer's mouse model, with and without TBI. METHODS 12-week-old male wild type (WT) and 5xFAD mice were administered either CLIP antagonist peptide (CAP) or vehicle, once at 30 min after either sham or a lateral fluid percussion injury (FPI). Analyses included flow cytometric analysis of immune cells in dural meninges and spleen, histopathological analysis of the brain, magnetic resonance diffusion tensor imaging, cerebrovascular analysis, and assessment of motor and neurobehavioral function over the ensuing 6 months. RESULTS 9-month-old 5xFAD mice had significantly more CLIP + B cells in the meninges compared to age-matched WT mice. A one-time treatment with CAP significantly reduced this population in 5xFAD mice. Importantly, CAP also improved some of the immune, histopathological, and neurobehavioral impairments in 5xFAD mice over the ensuing six months. Although FPI did not further elevate meningeal CLIP + B cells, it did negate the ability of CAP to reduce meningeal CLIP + B cells in the 5xFAD mice. FPI at 3 months of age exacerbated some aspects of AD pathology in 5xFAD mice, including further reducing hippocampal neurogenesis, increasing plaque deposition in CA3, altering microgliosis, and disrupting the cerebrovascular structure. CAP treatment after injury ameliorated some but not all of these FPI effects.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Reagan Dominy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Shreya Bandopadhyay
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - E Madison Arthur
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Brenda Noarbe
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Amandine Jullienne
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Margret Krkasharyan
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Richard P Tobin
- Department of Surgery, Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Aleksandr Pereverzev
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Samantha Beevers
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Lavanya Venkatasamy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Karienn A Souza
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Daniel C Jupiter
- Department of Biostatistics and Data Science, Department of Orthopaedics and Rehabilitation, The University of Texas Medical Branch, Galveston, TX, USA
| | - Alan Dabney
- Department of Statistics, College of Arts & Sciences, Texas A&M University, College Station, TX, USA
| | - Andre Obenaus
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - M Karen Newell-Rogers
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA.
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX, USA.
| | - Lee A Shapiro
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
6
|
Zeng L, Hu P, Zhang Y, Li M, Zhao Y, Li S, Luo A. Macrophage migration inhibitor factor (MIF): Potential role in cognitive impairment disorders. Cytokine Growth Factor Rev 2024; 77:67-75. [PMID: 38548489 DOI: 10.1016/j.cytogfr.2024.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 06/22/2024]
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine in the immune system, participated in both innate and adaptive immune responses. Except from immune cells, MIF is also secreted by a variety of non-immune cells, including hematopoietic cells, endothelial cells (ECs), and neurons. MIF plays a crucial role in various diseases, such as sepsis, rheumatoid arthritis, acute kidney injury, and neurodegenerative diseases. The role of MIF in the neuropathogenesis of cognitive impairment disorders is emphasized, as it recruits multiple inflammatory mediators, leading to activating microglia or astrocyte-derived neuroinflammation. Furthermore, it contributes to the cell death of neurons and ECs with the binding of apoptosis-inducing factor (AIF) through parthanatos-associated apoptosis-inducing factor nuclease (PAAN) / MIF pathway. This review comprehensively delves into the relationship between MIF and the neuropathogenesis of cognitive impairment disorders, providing a series of emerging MIF-targeted pharmaceuticals as potential treatments for cognitive impairment disorders.
Collapse
Affiliation(s)
- Lian Zeng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengchao Hu
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Zhang
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Mingyue Li
- Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Yilin Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
7
|
Patel J, Dawson VL, Dawson TM. Blocking the Self-Destruct Program of Dopamine Neurons through Macrophage Migration Inhibitory Factor Nuclease Inhibition. Mov Disord 2024; 39:644-650. [PMID: 38396375 PMCID: PMC11160583 DOI: 10.1002/mds.29748] [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: 11/13/2023] [Revised: 01/10/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition that pathognomonically involves the death of dopaminergic neurons in the substantia nigra pars compacta, resulting in a myriad of motor and non-motor symptoms. Given the insurmountable burden of this disease on the population and healthcare system, significant efforts have been put forth toward generating disease modifying therapies. This class of treatments characteristically alters disease course, as opposed to current strategies that focus on managing symptoms. Previous literature has implicated the cell death pathway known as parthanatos in PD progression. Inhibition of this pathway by targeting poly (ADP)-ribose polymerase 1 (PARP1) prevents neurodegeneration in a model of idiopathic PD. However, PARP1 has a vast repertoire of functions within the body, increasing the probability of side effects with the long-term treatment likely necessary for clinically significant neuroprotection. Recent work culminated in the development of a novel agent targeting the macrophage migration inhibitory factor (MIF) nuclease domain, also named parthanatos-associated apoptosis-inducing factor nuclease (PAAN). This nuclease activity specifically executes the terminal step in parthanatos. Parthanatos-associated apoptosis-inducing factor nuclease inhibitor-1 was neuroprotective in multiple preclinical mouse models of PD. This piece will focus on contextualizing this discovery, emphasizing its significance, and discussing its potential implications for parthanatos-directed treatment. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jaimin Patel
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
8
|
Pulliam L, Sun B, McCafferty E, Soper SA, Witek MA, Hu M, Ford JM, Song S, Kapogiannis D, Glesby MJ, Merenstein D, Tien PC, Freasier H, French A, McKay H, Diaz MM, Ofotokun I, Lake JE, Margolick JB, Kim EY, Levine SR, Fischl MA, Li W, Martinson J, Tang N. Microfluidic Isolation of Neuronal-Enriched Extracellular Vesicles Shows Distinct and Common Neurological Proteins in Long COVID, HIV Infection and Alzheimer's Disease. Int J Mol Sci 2024; 25:3830. [PMID: 38612641 PMCID: PMC11011771 DOI: 10.3390/ijms25073830] [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: 02/15/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Long COVID (LongC) is associated with a myriad of symptoms including cognitive impairment. We reported at the beginning of the COVID-19 pandemic that neuronal-enriched or L1CAM+ extracellular vesicles (nEVs) from people with LongC contained proteins associated with Alzheimer's disease (AD). Since that time, a subset of people with prior COVID infection continue to report neurological problems more than three months after infection. Blood markers to better characterize LongC are elusive. To further identify neuronal proteins associated with LongC, we maximized the number of nEVs isolated from plasma by developing a hybrid EV Microfluidic Affinity Purification (EV-MAP) technique. We isolated nEVs from people with LongC and neurological complaints, AD, and HIV infection with mild cognitive impairment. Using the OLINK platform that assesses 384 neurological proteins, we identified 11 significant proteins increased in LongC and 2 decreased (BST1, GGT1). Fourteen proteins were increased in AD and forty proteins associated with HIV cognitive impairment were elevated with one decreased (IVD). One common protein (BST1) was decreased in LongC and increased in HIV. Six proteins (MIF, ENO1, MESD, NUDT5, TNFSF14 and FYB1) were expressed in both LongC and AD and no proteins were common to HIV and AD. This study begins to identify differences and similarities in the neuronal response to LongC versus AD and HIV infection.
Collapse
Affiliation(s)
- Lynn Pulliam
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
- Department of Laboratory Medicine, San Francisco VA Health Care System, San Francisco, CA 94121, USA; (B.S.); (E.M.); (N.T.)
| | - Bing Sun
- Department of Laboratory Medicine, San Francisco VA Health Care System, San Francisco, CA 94121, USA; (B.S.); (E.M.); (N.T.)
| | - Erin McCafferty
- Department of Laboratory Medicine, San Francisco VA Health Care System, San Francisco, CA 94121, USA; (B.S.); (E.M.); (N.T.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (S.A.S.); (M.A.W.)
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66103, USA;
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
| | - Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (S.A.S.); (M.A.W.)
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66103, USA;
| | - Mengjia Hu
- Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66103, USA;
| | - Judith M. Ford
- Department of Mental Health, San Francisco VA Health Care System, San Francisco, CA 94121, USA; (J.M.F.); (S.S.)
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sarah Song
- Department of Mental Health, San Francisco VA Health Care System, San Francisco, CA 94121, USA; (J.M.F.); (S.S.)
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA;
| | - Marshall J. Glesby
- Department of Medicine, Weill Cornell Medical College, New York City, NY 10021, USA;
| | - Daniel Merenstein
- Department of Family Medicine, Georgetown University School of Medicine, Washington, DC 20007, USA;
| | - Phyllis C. Tien
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA; (P.C.T.); (H.F.)
- Department of Medicine, San Francisco VA Health Care System, San Francisco, CA 94121, USA
| | - Heather Freasier
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA; (P.C.T.); (H.F.)
| | - Audrey French
- Department of Medicine, Cook County Health, Chicago, IL 60612, USA;
| | - Heather McKay
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Monica M. Diaz
- Department of Neurology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA;
| | - Igho Ofotokun
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Jordan E. Lake
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Joseph B. Margolick
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Eun-Young Kim
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Steven R. Levine
- Department of Neurology, State University of New York College of Medicine and Downstate Medical Sciences University, Brooklyn, NY 11203, USA;
| | | | - Wei Li
- Department of Clinical and Diagnostic Sciences, University of Alabama, Birmingham, AL 35294, USA;
| | - Jeremy Martinson
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Norina Tang
- Department of Laboratory Medicine, San Francisco VA Health Care System, San Francisco, CA 94121, USA; (B.S.); (E.M.); (N.T.)
| |
Collapse
|
9
|
Hjæresen S, Benedikz E, Sejbaek T, Axelsson M, Novakova L, Zhang M, Lycke J, Illes Z, Fex-Svenningsen Å. High temperature requirement A1 and macrophage migration inhibitory factor in the cerebrospinal fluid; a potential marker of conversion from relapsing-remitting to secondary progressive multiple sclerosis. J Neurol Sci 2024; 457:122888. [PMID: 38278096 DOI: 10.1016/j.jns.2024.122888] [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: 05/01/2023] [Revised: 11/30/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Predictive and prognostic biomarkers for multiple sclerosis (MS) remain a significant gap in MS diagnosis and treatment monitoring. Currently, there are no timely markers to diagnose the transition to secondary progressive MS (SPMS). OBJECTIVE This study aims to evaluate the discriminatory potential of the High temperature requirement serine protease (HTRA1)/Macrophage migration inhibitory factor (MIF) cerebrospinal fluid (CSF) ratio in distinguishing relapsing-remitting (RRMS) patients from SPMS patients. METHODS The MIF and HTRA1 CSF levels were determined using ELISA in healthy controls (n = 23), RRMS patients before (n = 22) and after 1 year of dimethyl fumarate treatment (n = 11), as well as in SPMS patients before (n = 11) and after 2 years of mitoxantrone treatment (n = 7). The ability of the HTRA1/MIF ratio to discriminate the different groups was determined using receiver operating curve (ROC) analyses. RESULTS The ratio was significantly increased in treatment naïve RRMS patients while decreased again in SPMS patients at baseline. Systemic administrated disease modifying treatment (DMT) only significantly affected the ratio in RRMS patients. ROC analysis demonstrated that the ratio could discriminate treatment naïve RRMS patients from SPMS patients with 91% sensitivity and 100% specificity. CONCLUSION The HTRA1/MIF ratio is a strong candidate as a MS biomarker for SPMS conversion.
Collapse
Affiliation(s)
- Simone Hjæresen
- University of Southern Denmark, Department of Molecular Medicine, Campusvej 55, 5230 Odense M, Denmark
| | - Eirikur Benedikz
- University of Southern Denmark, Faculty of Health Sciences, Campusvej 55, 5230 Odense M, Denmark
| | - Tobias Sejbaek
- University of Southern Denmark, Department of Regional Health Research, 5000 Odense, Denmark; University of Copenhagen, Department of Neurology, Southwest Jutland University Hospital, 6700 Esbjerg, Denmark
| | - Markus Axelsson
- University of Gothenburg, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lenka Novakova
- University of Gothenburg, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mengliang Zhang
- University of Southern Denmark, Department of Molecular Medicine, Campusvej 55, 5230 Odense M, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark
| | - Jan Lycke
- University of Gothenburg, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Zsolt Illes
- University of Southern Denmark, Department of Molecular Medicine, Campusvej 55, 5230 Odense M, Denmark; Odense University Hospital, Department of Neurology, Odense, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark
| | - Åsa Fex-Svenningsen
- University of Southern Denmark, Department of Molecular Medicine, Campusvej 55, 5230 Odense M, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
10
|
Miao X, Wu Q, Du S, Xiang L, Zhou S, Zhu J, Chen Z, Wang H, Pan X, Fan Y, Zhang L, Qian J, Xing Y, Xie Y, Hu L, Xu H, Wang W, Wang Y, Huang Z. SARM1 Promotes Neurodegeneration and Memory Impairment in Mouse Models of Alzheimer's Disease. Aging Dis 2024; 15:390-407. [PMID: 37307837 PMCID: PMC10796105 DOI: 10.14336/ad.2023.0516-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/16/2023] [Indexed: 06/14/2023] Open
Abstract
Neuroinflammation plays a crucial role in the pathogenesis and progression of Alzheimer's disease (AD). The Sterile Alpha and Toll Interleukin Receptor Motif-containing protein 1 (SARM1) has been shown to promote axonal degeneration and is involved in neuroinflammation. However, the role of SARM1 in AD remains unclear. In this study, we found that SARM1 was reduced in hippocampal neurons of AD model mice. Interestingly, conditional knockout (CKO) of SARM1 in the central nervous system (CNS, SARM1Nestin-CKO mice) delayed the cognitive decline in APP/PS1 AD model mice. Furthermore, SARM1 deletion reduced the Aβ deposition and inflammatory infiltration in the hippocampus and inhibited neurodegeneration in APP/PS1 AD model mice. Further investigation into the underlying mechanisms revealed that the signaling of tumor necrosis factor-α (TNF-α) was downregulated in the hippocampus tissues of APP/PS1;SARM1Nestin-CKO mice, thereby alleviating the cognitive decline, Aβ deposition and inflammatory infiltration. These findings identify unrecognized functions of SARM1 in promoting AD and reveal the SARM1-TNF-α pathway in AD model mice.
Collapse
Affiliation(s)
- Xuemeng Miao
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Qian Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Ludan Xiang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Siyao Zhou
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Junzhe Zhu
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Zirun Chen
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Hui Wang
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Xuyi Pan
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Yiren Fan
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Lihan Zhang
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Jingkang Qian
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Yuxuan Xing
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Yiyang Xie
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Lixin Hu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Haiyun Xu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Ying Wang
- Clinical Research Center, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Zhihui Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
11
|
Matejuk A, Benedek G, Bucala R, Matejuk S, Offner H, Vandenbark AA. MIF contribution to progressive brain diseases. J Neuroinflammation 2024; 21:8. [PMID: 38178143 PMCID: PMC10765708 DOI: 10.1186/s12974-023-02993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Progressive brain diseases create a huge social and economic burden on modern societies as a major cause of disability and death. Incidence of brain diseases has a significantly increasing trend and merits new therapeutic strategies. At the base of many progressive brain malfunctions is a process of unresolved, chronic inflammation. Macrophage migration inhibitory factor, MIF, is an inflammatory mediator that recently gained interest of neuro-researchers due to its varied effects on the CNS such as participation of nervous system development, neuroendocrine functions, and modulation of neuroinflammation. MIF appears to be a candidate as a new biomarker and target of novel therapeutics against numerous neurologic diseases ranging from cancer, autoimmune diseases, vascular diseases, neurodegenerative pathology to psychiatric disorders. In this review, we will focus on MIF's crucial role in neurological diseases such as multiple sclerosis (MS), Alzheimer's disease (AD) and glioblastoma (GBM).
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland.
| | - Gil Benedek
- Tissue Typing and Immunogenetics Unit, Department of Genetics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Richard Bucala
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | | | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| |
Collapse
|
12
|
Ji D, Chen WZ, Zhang L, Zhang ZH, Chen LJ. Gut microbiota, circulating cytokines and dementia: a Mendelian randomization study. J Neuroinflammation 2024; 21:2. [PMID: 38178103 PMCID: PMC10765696 DOI: 10.1186/s12974-023-02999-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Some studies have shown that gut microbiota may be associated with dementia. However, the causal effects between gut microbiota and different types of dementia and whether cytokines act as a mediator remain unclear. METHODS Gut microbiota, cytokines, and five dementia types, including Alzheimer's disease (AD), frontotemporal dementia (FTD), dementia with Lewy body (DLB), vascular dementia (VD), and Parkinson's disease dementia (PDD) were identified from large-scale genome-wide association studies (GWAS) summary data. We used Mendelian randomization (MR) to investigate the causal relationships between gut microbiota, cytokines, and five types of dementia. Inverse variance weighting (IVW) was used as the main statistical method. In addition, we explored whether cytokines act as a mediating factor in the pathway from gut microbiota to dementia. RESULTS There were 20 positive and 16 negative causal effects between genetic liability in the gut microbiota and dementia. Also, there were five positive and four negative causal effects between cytokines and dementias. Cytokines did not act as mediating factors. CONCLUSIONS Gut microbiota and cytokines were causally associated with five types of dementia, and cytokines seemed not to be the mediating factors in the pathway from gut microbiota to dementia.
Collapse
Affiliation(s)
- Dong Ji
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China
| | - Wen-Zhu Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China
| | - Lei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China
| | - Zhi-Hua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Li-Jian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China.
| |
Collapse
|
13
|
Yao J, Wang Z, Song W, Zhang Y. Targeting NLRP3 inflammasome for neurodegenerative disorders. Mol Psychiatry 2023; 28:4512-4527. [PMID: 37670126 DOI: 10.1038/s41380-023-02239-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023]
Abstract
Neuroinflammation is a key pathological feature in neurological diseases, including Alzheimer's disease (AD). The nucleotide-binding domain leucine-rich repeat-containing proteins (NLRs) belong to the pattern recognition receptors (PRRs) family that sense stress signals, which play an important role in inflammation. As a member of NLRs, the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) is predominantly expressed in microglia, the principal innate immune cells in the central nervous system (CNS). Microglia release proinflammatory cytokines to cause pyroptosis through activating NLRP3 inflammasome. The active NLRP3 inflammasome is involved in a variety of neurodegenerative diseases (NDs). Recent studies also indicate the key role of neuronal NLRP3 in the pathogenesis of neurological disorders. In this article, we reviewed the mechanisms of NLRP3 expression and activation and discussed the role of active NLRP3 inflammasome in the pathogenesis of NDs, particularly focusing on AD. The studies suggest that targeting NLRP3 inflammasome could be a novel approach for the disease modification.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325000, Zhejiang, China.
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
| |
Collapse
|
14
|
Jang J, Yeo S, Baek S, Jung HJ, Lee MS, Choi SH, Choe Y. Abnormal accumulation of extracellular vesicles in hippocampal dystrophic axons and regulation by the primary cilia in Alzheimer's disease. Acta Neuropathol Commun 2023; 11:142. [PMID: 37667395 PMCID: PMC10478284 DOI: 10.1186/s40478-023-01637-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Dystrophic neurites (DNs) are abnormal axons and dendrites that are swollen or deformed in various neuropathological conditions. In Alzheimer's disease (AD), DNs play a crucial role in impairing neuronal communication and function, and they may also contribute to the accumulation and spread of amyloid beta (Aβ) in the brain of AD patients. However, it is still a challenge to understand the DNs of specific neurons that are vulnerable to Aβ in the pathogenesis of AD. To shed light on the development of radiating DNs, we examined enriched dystrophic hippocampal axons in a mouse model of AD using a three-dimensional rendering of projecting neurons. We employed the anterograde spread of adeno-associated virus (AAV)1 and conducted proteomic analysis of synaptic compartments obtained from hippocampo-septal regions. Our findings revealed that DNs were formed due to synaptic loss at the axon terminals caused by the accumulation of extracellular vesicle (EV). Abnormal EV-mediated transport and exocytosis were identified in association with primary cilia, indicating their involvement in the accumulation of EVs at presynaptic terminals. To further address the regulation of DNs by primary cilia, we conducted knockdown of the Ift88 gene in hippocampal neurons, which impaired EV-mediated secretion of Aβ and promoted accumulation of axonal spheroids. Using single-cell RNA sequencing, we identified the septal projecting hippocampal somatostatin neurons (SOM) as selectively vulnerable to Aβ with primary cilia dysfunction and vesicle accumulation. Our study suggests that DNs in AD are initiated by the ectopic accumulation of EVs at the neuronal axon terminals, which is affected by neuronal primary cilia.
Collapse
Affiliation(s)
| | - Seungeun Yeo
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | | | - Mi Suk Lee
- Korea Brain Research Institute, Daegu, 41068, Korea
| | | | - Youngshik Choe
- Korea Brain Research Institute, Daegu, 41068, Korea.
- , Daegu, Korea.
| |
Collapse
|
15
|
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther 2023; 8:248. [PMID: 37386015 PMCID: PMC10310781 DOI: 10.1038/s41392-023-01484-7] [Citation(s) in RCA: 150] [Impact Index Per Article: 150.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| |
Collapse
|
16
|
Hok-A-Hin YS, Del Campo M, Boiten WA, Stoops E, Vanhooren M, Lemstra AW, van der Flier WM, Teunissen CE. Neuroinflammatory CSF biomarkers MIF, sTREM1, and sTREM2 show dynamic expression profiles in Alzheimer's disease. J Neuroinflammation 2023; 20:107. [PMID: 37147668 PMCID: PMC10163795 DOI: 10.1186/s12974-023-02796-9] [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: 01/16/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND There is a need for novel fluid biomarkers tracking neuroinflammatory responses in Alzheimer's disease (AD). Our recent cerebrospinal fluid (CSF) proteomics study revealed that migration inhibitory factor (MIF) and soluble triggering receptor expressed on myeloid cells 1 (sTREM1) increased along the AD continuum. We aimed to assess the potential use of these proteins, in addition to sTREM2, as CSF biomarkers to monitor inflammatory processes in AD. METHODS We included cognitively unimpaired controls (n = 67, 63 ± 9 years, 24% females, all amyloid negative), patients with mild cognitive impairment (MCI; n = 92, 65 ± 7 years, 47% females, 65% amyloid positive), AD (n = 38, 67 ± 6 years, 8% females, all amyloid positive), and DLB (n = 50, 67 ± 6 years, 5% females, 54% amyloid positive). MIF, sTREM1, and sTREM2 levels were measured by validated immunoassays. Differences in protein levels between groups were tested with analysis of covariance (corrected for age and sex). Spearman correlation analysis was performed to evaluate the association between these neuroinflammatory markers with AD-CSF biomarkers (Aβ42, tTau, pTau) and mini-mental state examination (MMSE) scores. RESULTS MIF levels were increased in MCI (p < 0.01), AD (p < 0.05), and DLB (p > 0.05) compared to controls. Levels of sTREM1 were specifically increased in AD compared to controls (p < 0.01), MCI (p < 0.05), and DLB patients (p > 0.05), while sTREM2 levels were increased specifically in MCI compared to all other groups (all p < 0.001). Neuroinflammatory proteins were highly correlated with CSF pTau levels (MIF: all groups; sTREM1: MCI, AD and DLB; sTREM2: controls, MCI and DLB). Correlations with MMSE scores were observed in specific clinical groups (MIF in controls, sTREM1 in AD, and sTREM2 in DLB). CONCLUSION Inflammatory-related proteins show diverse expression profiles along different AD stages, with increased protein levels in the MCI stage (MIF and sTREM2) and AD stage (MIF and sTREM1). The associations of these inflammatory markers primarily with CSF pTau levels indicate an intertwined relationship between tau pathology and inflammation. These neuroinflammatory markers might be useful in clinical trials to capture dynamics in inflammatory responses or monitor drug-target engagement of inflammatory modulators.
Collapse
Affiliation(s)
- Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Marta Del Campo
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Walter A Boiten
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands
| | | | | | - Afina W Lemstra
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, VU University Medical Centers, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Xuan W, Xie W, Li F, Huang D, Zhu Z, Lin Y, Lu B, Yu W, Li Y, Li P. Dualistic roles and mechanistic insights of macrophage migration inhibitory factor in brain injury and neurodegenerative diseases. J Cereb Blood Flow Metab 2023; 43:341-356. [PMID: 36369735 PMCID: PMC9941868 DOI: 10.1177/0271678x221138412] [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: 06/11/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/15/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is involved in various immune-mediated pathologies and regulates both innate and adaptive immune reactions, thus being related to several acute and chronic inflammatory diseases such as rheumatoid arthritis, septic shock, and atherosclerosis. Its role in acute and chronic brain pathologies, such as stroke and neurodegenerative diseases, has attracted increasing attention in recent years. In response to stimuli like hypoxia, inflammation or infection, different cell types can rapidly release MIF, including immune cells, endothelial cells, and neuron cells. Notably, clinical data from past decades also suggested a possible link between serum MIF levels and the severity of stroke and the evolving of neurodegenerative diseases. In this review, we summarize the major and recent findings focusing on the mechanisms of MIF modulating functions in brain injury and neurodegenerative diseases, which may provide important therapeutic targets meriting further investigation.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Wanqing Xie
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Fengshi Li
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong
University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yuxuan Lin
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Binwei Lu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| |
Collapse
|
18
|
Zhao J, Wang X, Li Q, Lu C, Li S. The relevance of serum macrophage migratory inhibitory factor and cognitive dysfunction in patients with cerebral small vascular disease. Front Aging Neurosci 2023; 15:1083818. [PMID: 36824264 PMCID: PMC9941340 DOI: 10.3389/fnagi.2023.1083818] [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: 10/29/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Cerebral small vascular disease (CSVD) is a common type of cerebrovascular disease, and an important cause of vascular cognitive impairment (VCI) and stroke. The disease burden is expected to increase further as a result of population aging, an ongoing high prevalence of risk factors (e.g., hypertension), and inadequate management. Due to the poor understanding of pathophysiology in CSVD, there is no effective preventive or therapeutic approach for CSVD. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine that is related to the occurrence and development of vascular dysfunction diseases. Therefore, MIF may contribute to the pathogenesis of CSVD and VCI. Here, reviewed MIF participation in chronic cerebral ischemia-hypoperfusion and neurodegeneration pathology, including new evidence for CSVD, and its potential role in protection against VCI.
Collapse
Affiliation(s)
- Jianhua Zhao
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China,*Correspondence: Jianhua Zhao,
| | - Xiaoting Wang
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qiong Li
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Chengbiao Lu
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Wang X, Wang X, Liu Y, Sun Z, Liu H, Shen J, Zhu HL, Qian Y. Activity-Based Imaging of Macrophage Migration Inhibitory Factor with a Two-Photon Fluorescent Probe. ACS Sens 2023; 8:335-343. [PMID: 36530142 DOI: 10.1021/acssensors.2c02326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Macrophage migration inhibitory factor (MIF), as a cytokine, plays an important role in the pathogenesis of cancer and some other diseases, and it is also one of the potential drug targets for disease treatment. However, due to the lack of simple and effective MIF imaging detection tools, the fluctuation and distribution of MIF in living cells or at lesion sites remain difficult to track precisely and in real time. Here, we report activity-based fluorescent probes, named MIFP1-MIFP3, which are used for real-time imaging and tracking of intracellular MIF, thus establishing a relationship between the fluctuation of MIF and the change of fluorescence signal during the cancer disease process. With the excellent optical properties of two-photon probe imaging, we can easily distinguish multiple cancer cells from normal cells with the representative probe, MIFP3. Moreover, MIFP3 has also been successfully used to directly identify the pathological tissues of patients with clinical liver cancer. These potential MIF probes could provide powerful tools for further study of the physiological function of MIF and will be helpful to promote the accurate diagnosis and therapeutic evaluation of MIF-associated malignancies.
Collapse
Affiliation(s)
- Xueao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Wenyuan Road 1, Nanjing 210046, China
| | - Xueting Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China
| | - Yani Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China
| | - Zhigang Sun
- Central Laboratory, Linyi Central Hospital, No. 17 Jiankang Road, Linyi 276400, China
| | - Huan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China
| | - Jiawen Shen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China
| | - Yong Qian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Xianlin Road 163, Nanjing 210023, China.,Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Wenyuan Road 1, Nanjing 210046, China
| |
Collapse
|
20
|
Koopmans F, Li KW, Klaassen RV, Smit AB. MS-DAP Platform for Downstream Data Analysis of Label-Free Proteomics Uncovers Optimal Workflows in Benchmark Data Sets and Increased Sensitivity in Analysis of Alzheimer's Biomarker Data. J Proteome Res 2022; 22:374-386. [PMID: 36541440 PMCID: PMC9903323 DOI: 10.1021/acs.jproteome.2c00513] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the rapidly moving proteomics field, a diverse patchwork of data analysis pipelines and algorithms for data normalization and differential expression analysis is used by the community. We generated a mass spectrometry downstream analysis pipeline (MS-DAP) that integrates both popular and recently developed algorithms for normalization and statistical analyses. Additional algorithms can be easily added in the future as plugins. MS-DAP is open-source and facilitates transparent and reproducible proteome science by generating extensive data visualizations and quality reporting, provided as standardized PDF reports. Second, we performed a systematic evaluation of methods for normalization and statistical analysis on a large variety of data sets, including additional data generated in this study, which revealed key differences. Commonly used approaches for differential testing based on moderated t-statistics were consistently outperformed by more recent statistical models, all integrated in MS-DAP. Third, we introduced a novel normalization algorithm that rescues deficiencies observed in commonly used normalization methods. Finally, we used the MS-DAP platform to reanalyze a recently published large-scale proteomics data set of CSF from AD patients. This revealed increased sensitivity, resulting in additional significant target proteins which improved overlap with results reported in related studies and includes a large set of new potential AD biomarkers in addition to previously reported.
Collapse
Affiliation(s)
- Frank Koopmans
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands,
| | - Ka Wan Li
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands
| | - Remco V. Klaassen
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands
| | - August B. Smit
- Department
of Molecular and Cellular Neurobiology, Center for Neurogenomics and
Cognitive Research, Amsterdam Neuroscience, VU University, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
21
|
Kaczmarczyk L, Schleif M, Dittrich L, Williams RH, Koderman M, Bansal V, Rajput A, Schulte T, Jonson M, Krost C, Testaquadra FJ, Bonn S, Jackson WS. Distinct translatome changes in specific neural populations precede electroencephalographic changes in prion-infected mice. PLoS Pathog 2022; 18:e1010747. [PMID: 35960762 PMCID: PMC9401167 DOI: 10.1371/journal.ppat.1010747] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 08/24/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Selective vulnerability is an enigmatic feature of neurodegenerative diseases (NDs), whereby a widely expressed protein causes lesions in specific cell types and brain regions. Using the RiboTag method in mice, translational responses of five neural subtypes to acquired prion disease (PrD) were measured. Pre-onset and disease onset timepoints were chosen based on longitudinal electroencephalography (EEG) that revealed a gradual increase in theta power between 10- and 18-weeks after prion injection, resembling a clinical feature of human PrD. At disease onset, marked by significantly increased theta power and histopathological lesions, mice had pronounced translatome changes in all five cell types despite appearing normal. Remarkably, at a pre-onset stage, prior to EEG and neuropathological changes, we found that 1) translatomes of astrocytes indicated reduced synthesis of ribosomal and mitochondrial components, 2) glutamatergic neurons showed increased expression of cytoskeletal genes, and 3) GABAergic neurons revealed reduced expression of circadian rhythm genes. These data demonstrate that early translatome responses to neurodegeneration emerge prior to conventional markers of disease and are cell type-specific. Therapeutic strategies may need to target multiple pathways in specific populations of cells, early in disease. Prions are infectious agents composed of a misfolded protein. When isolated from a mammalian brain and transferred to the same host species, prions will cause the same neurodegenerative disease affecting the same brain regions and cell types. This concept of selective vulnerability is also a feature of more common types of neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, and Huntington’s. To better understand the mechanisms behind selective vulnerability, we studied disease responses of five cell types with different vulnerabilities in prion-infected mice at two different disease stages. Responses were measured as changes to mRNAs undergoing translation, referred to as the translatome. Before prion-infected mice demonstrated typical disease signs, electroencephalography (a method used clinically to characterize neurodegeneration in humans) revealed brain changes resembling those in human prion diseases, and surprisingly, the translatomes of all cells were drastically changed. Furthermore, before electroencephalography changes emerged, three cell types made unique responses while the most vulnerable cell type did not. These results suggests that mechanisms causing selective vulnerability will be difficult to dissect and that therapies will likely need to be provided before clinical signs emerge and individually engage multiple cell types and their distinct molecular pathways.
Collapse
Affiliation(s)
- Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Melvin Schleif
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Lars Dittrich
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Maruša Koderman
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Vikas Bansal
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Ashish Rajput
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
- Maximon AG, Zug, Switzerland
| | | | - Maria Jonson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Clemens Krost
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | | | - Stefan Bonn
- Institute of Medical Systems Biology, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Germany
| | - Walker S. Jackson
- Wallenberg Center for Molecular Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- German Center for Neurodegenerative Diseases, Bonn, Germany
- * E-mail:
| |
Collapse
|
22
|
Hjæresen S, Svenningsen ÅF. The possible function and regulation of macrophage migration inhibitory factor (MIF) in multiple sclerosis (MS). J Neurol Sci 2022; 441:120352. [DOI: 10.1016/j.jns.2022.120352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/15/2022]
|
23
|
Khalil RM, Alaa S, Eissa H, Youssef I. Early Prediction of a Pre-Symptomatic Neurodegeneration Disorder by Measuring Macrophage Inhibitory Factor Level in Diabetic Patients. J Alzheimers Dis 2022; 88:1167-1177. [PMID: 35754265 DOI: 10.3233/jad-215561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The relationship between diabetes mellitus and neurodegenerative disorders has been of great interest. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine in which a variety of signaling cascades are activated through it. MIF has been involved in the pathogenesis of several diseases and can predict early pre-symptomatic stages of neurodegeneration in diabetic patients. OBJECTIVE To investigate whether serum MIF could predict brain neurodegeneration at the early pre-symptomatic stages in diabetic patients. METHODS We examined adults with type 2 diabetes mellitus and compared with normal control adults using a short form of the IQCODE and biochemical examination, including assessment of HA1C, fasting blood glucose, lipid profile, and MIF which was measured by ELISA technique. Correlations between parameters were studied. Computational PathLinker bioinformatic tool was used to search for potential pathway reconstructions for the insulin/amyloid-β/MIF signaling. RESULTS We demonstrated that MIF level was increased in the serum at the early pre-symptomatic stages of neurodegenerative disorder in diabetic patients. In addition, network analysis demonstrates that insulin receptor substrate 1 can ameliorate amyloid-β protein precursor through COP9 signalosome complex subunit 5 that enhances MIF elevation. CONCLUSION Diagnosis processes could not be used as routine examinations for still pre-symptomatic neurodegenerative disorders. This may be due to the time constraints and the heavy dependence on the physician's experience. Therefore, serum MIF level could predict brain neurodegeneration at the early pre-symptomatic stages in diabetic patients which may support its potential utility as a clinically useful biomarker.
Collapse
Affiliation(s)
- Rania M Khalil
- Biochemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Shereen Alaa
- Pharmacology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Hanan Eissa
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | |
Collapse
|
24
|
Hjæresen S, Sejbaek T, Axelsson M, Mortensen SK, Vinsløv-Jensen H, Pihl-Jensen G, Novakova L, Pedersen CB, Halle B, Poulsen FR, Zhang M, Benedikz E, Frederiksen JL, Lycke J, Illes Z, Fex-Svenningsen Å. MIF in the cerebrospinal fluid is decreased during relapsing-remitting while increased in secondary progressive multiple sclerosis. J Neurol Sci 2022; 439:120320. [PMID: 35717879 DOI: 10.1016/j.jns.2022.120320] [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: 11/29/2021] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is involved in the function of both the innate and adaptive immune systems and in neuroprotection and has recently been implicated in multiple sclerosis (MS). OBJECTIVES Determination of MIF levels in the cerebrospinal fluid (CSF) of patients with distinct subtypes of MS and the cellular localization of MIF in human brain tissue. METHODS The levels of MIF were investigated in CSF from patients with clinically isolated syndrome (CIS) (n = 26), relapsing-remitting MS (RRMS) (n = 22), secondary progressive MS (SPMS) (n = 19), and healthy controls (HCs) (n = 24), using ELISA. The effect of disease-modifying therapies in the RRMS and SPMS cohorts were examined. Cellular distribution of MIF in the human brain was studied using immunochemistry and the newly available OligoInternode database. RESULTS MIF was significantly decreased in treatment-naïve CIS and RRMS patients compared to HCs but was elevated in SPMS. Interestingly, MIF levels were sex-dependent and significantly higher in women with CIS and RRMS. MIF expression in the human brain was localized to neurons, astrocytes, pericytes, and oligo5 oligodendrocytes but not in microglia. CONCLUSION The finding that MIF was decreased in newly diagnosed CIS and RRMS patients but was high in patients with SPMS may suggest that MIF levels in CSF are regulated by local MIF receptor expression that affects the overall MIF signaling in the brain and may represent a protective mechanism that eventually fails.
Collapse
Affiliation(s)
- Simone Hjæresen
- University of Southern Denmark, Department of Molecular Medicine, J.B. Winsløws vej 21, 5000 Odense, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| | - Tobias Sejbaek
- University of Southern Denmark, Department of Regional Health Research, 5000 Odense, Denmark; University of Copenhagen, Department of Neurology, Southwest Jutland University Hospital, 6700 Esbjerg, Denmark.
| | - Markus Axelsson
- University of Gothenburg, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Sif Kløvedal Mortensen
- University of Southern Denmark, Department of Molecular Medicine, J.B. Winsløws vej 21, 5000 Odense, Denmark.
| | - Helle Vinsløv-Jensen
- University of Southern Denmark, Department of Molecular Medicine, J.B. Winsløws vej 21, 5000 Odense, Denmark
| | - Gorm Pihl-Jensen
- University of Copenhagen, Department of Neurology, Rigshospitalet Glostrup, Copenhagen, Denmark
| | - Lenka Novakova
- University of Gothenburg, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Christian Bonde Pedersen
- Odense University Hospital, Department of Neurosurgery, Clinical Institute and University of Southern Denmark, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| | - Bo Halle
- Odense University Hospital, Department of Neurosurgery, Clinical Institute and University of Southern Denmark, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| | - Frantz Rom Poulsen
- Odense University Hospital, Department of Neurosurgery, Clinical Institute and University of Southern Denmark, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| | - Mengliang Zhang
- University of Southern Denmark, Department of Molecular Medicine, J.B. Winsløws vej 21, 5000 Odense, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| | - Eirikur Benedikz
- University of Southern Denmark, Faculty of Health Sciences, J.B. Winsløws vej 19., 5000 Odense, Denmark.
| | | | - Jan Lycke
- University of Gothenburg, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Zsolt Illes
- University of Southern Denmark, Department of Molecular Medicine, J.B. Winsløws vej 21, 5000 Odense, Denmark; Odense University Hospital, Department of Neurology, Odense, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| | - Åsa Fex-Svenningsen
- University of Southern Denmark, Department of Molecular Medicine, J.B. Winsløws vej 21, 5000 Odense, Denmark; BRIDGE - Brain Research InterDisciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
25
|
Wang Q, Fang C, Huang X, Xue L. Research progress of the CXCR4 mechanism in Alzheimer's disease. IBRAIN 2022; 8:3-14. [PMID: 37786419 PMCID: PMC10528775 DOI: 10.1002/ibra.12026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 10/04/2023]
Abstract
Alzheimer's disease (AD) is a degenerative brain disease with complex clinical manifestations and pathogeneses such as abnormal deposition of beta-amyloid protein and inflammation caused by the excessive activation of microglia. CXC motif chemokine receptor type 4 (CXCR4) is a type of G protein-coupled receptor that binds to CXC motif ligand 12 (CXCL12) to activate downstream signaling pathways, such as the Janus kinase/signal transducer and activator of transcription and the renin-angiotensin system (Ras)/RAF proto-oncogene serine (Raf)/mitogen-activated protein kinase/extracellular-regulated protein kinase; most of these signaling pathways are involved in inflammatory responses. CXCR4 is highly expressed in the microglia and astrocytes; this might be one of the important causes of inflammation caused by microglia and astrocytes. In this review, we summarize the mechanism and therapeutics of AD, the structures of CXCR4 and the CXCL12 ligand, and the mechanisms of CXCR4/CXCL12 that are involved in the occurrence and development of AD. The possible treatment of AD through microglia and astrocytes is also discussed, with the aim of providing a new method for the treatment of AD.
Collapse
Affiliation(s)
- Qiu‐Lin Wang
- Department of Clinical MedicineChongqing Medical UniversityChongqingChina
| | - Chang‐Le Fang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Xue‐Yan Huang
- Department of AnesthesiologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Lu‐Lu Xue
- State Key Laboratory of Biotherapy of Sichuan UniversityChengduSichuanChina
| |
Collapse
|
26
|
Yu JJ, Zhao Q, Li HN, Song JQ, Chen DC. Macrophage migration inhibitory factor as a potential novel biomarker for cognitive function in patients with first-episode schizophrenia. Aust N Z J Psychiatry 2022; 56:292-300. [PMID: 33985351 DOI: 10.1177/00048674211013086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Cognitive impairment is prevalent in schizophrenia. Macrophage migration inhibitory factor which is released into the circulation under stress or inflammation, is associated with cognition and also plays an important role in immunity. However, no study has investigated the relationship between macrophage migration inhibitory factor and cognitive function in first-episode schizophrenia patients at baseline or after treatment. This study investigated the pre- and post-risperidone treatment correlations between serum macrophage migration inhibitory factor levels and cognitive function in first-episode schizophrenia patients. METHODS A total of 83 first-episode schizophrenia patients who received risperidone monotherapy and 57 healthy controls - matched for sex, age, smoking status, education (years), marital status and waist-to-hip ratio - were included. Macrophage migration inhibitory factor levels were measured before and 10 weeks after treatment in the patient group and at baseline in the controls. Pre- and post-treatment cognitive functions in patients were assessed using the MATRICS Consensus Cognitive Battery. RESULTS At baseline, macrophage migration inhibitory factor levels were significantly higher in first-episode schizophrenia patients than those in healthy controls (p < 0.01) and decreased in patients after 10 weeks of risperidone treatment compared with baseline (p < 0.05). The MATRICS Consensus Cognitive Battery total score and the sub-scores for the Trail Making Test, Symbol Coding, Letter Number Sequence, Maze and Brief Visuospatial Memory Test-Revised improved significantly after risperidone treatment. After controlling for age, sex, education, waist-to-hip ratio and smoking status, partial correlation analysis showed a positive correlation between baseline macrophage migration inhibitory factor levels and patients' baseline MATRICS Consensus Cognitive Battery verbal memory scores (r = 0.29, p = 0.01). Macrophage migration inhibitory factor changes correlated negatively with verbal memory changes (r = -0.26, p = 0.04). Multiple linear regression analysis identified a definite correlation between the changes in word memory test score and macrophage migration inhibitory factor level (β = -0.09, p = 0.04). CONCLUSION Macrophage migration inhibitory factor may be involved in the process of cognitive impairment in first-episode schizophrenia and repair mechanisms following risperidone treatment.
Collapse
Affiliation(s)
- Jian-Jin Yu
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Changping district, Beijing 100096, China
| | - Qing Zhao
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Changping district, Beijing 100096, China
| | - Hong-Na Li
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Changping district, Beijing 100096, China
| | - Jia-Qi Song
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Changping district, Beijing 100096, China
| | - Da-Chun Chen
- Peking University Huilongguan Clinical Medical School, Beijing Huilongguan Hospital, Changping district, Beijing 100096, China
| |
Collapse
|
27
|
Gao L, Zhang Y, Sterling K, Song W. Brain-derived neurotrophic factor in Alzheimer's disease and its pharmaceutical potential. Transl Neurodegener 2022; 11:4. [PMID: 35090576 PMCID: PMC8796548 DOI: 10.1186/s40035-022-00279-0] [Citation(s) in RCA: 169] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/01/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic abnormalities are a cardinal feature of Alzheimer's disease (AD) that are known to arise as the disease progresses. A growing body of evidence suggests that pathological alterations to neuronal circuits and synapses may provide a mechanistic link between amyloid β (Aβ) and tau pathology and thus may serve as an obligatory relay of the cognitive impairment in AD. Brain-derived neurotrophic factors (BDNFs) play an important role in maintaining synaptic plasticity in learning and memory. Considering AD as a synaptic disorder, BDNF has attracted increasing attention as a potential diagnostic biomarker and a therapeutical molecule for AD. Although depletion of BDNF has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation and neuronal apoptosis, the exact mechanisms underlying the effect of impaired BDNF signaling on AD are still unknown. Here, we present an overview of how BDNF genomic structure is connected to factors that regulate BDNF signaling. We then discuss the role of BDNF in AD and the potential of BDNF-targeting therapeutics for AD.
Collapse
Affiliation(s)
- Lina Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, College of Pharmacy, Jining Medical University, Jining, 272067, Shandong, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
28
|
Chen X, Chen Y, Qi D, Cui D. Multifaceted interconnections between macrophage migration inhibitory factor and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110422. [PMID: 34358623 DOI: 10.1016/j.pnpbp.2021.110422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 01/02/2023]
Abstract
Inflammation is involved in the pathogenesis of psychiatric disorders. Many previous studies have defined the important roles of inflammatory factors in the pathogenesis, diagnosis, and treatment outcomes of psychiatric disorders. Macrophage migration inhibitory factor (MIF), a pro-inflammatory factor, has been gradually recognized to be involved in the development of neurological diseases in recent years. Our current review focuses on discussing the potential beneficial and detrimental roles of MIF in psychiatric disorders. We will provide new mechanistic insights for the development of potential diagnostic and therapeutic biomarkers based on MIF for psychiatric diseases.
Collapse
Affiliation(s)
- Xi Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
| | - Yifan Chen
- Department of Psychology, Tufts University, Medford, MA, USA.
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China; Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
29
|
Hu J, Lin SL, Schachner M. A fragment of cell adhesion molecule L1 reduces amyloid-β plaques in a mouse model of Alzheimer's disease. Cell Death Dis 2022; 13:48. [PMID: 35013124 PMCID: PMC8748658 DOI: 10.1038/s41419-021-04348-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 10/02/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023]
Abstract
Deposition of amyloid-β (Aβ) in the brain is one of the important histopathological features of Alzheimer's disease (AD). Previously, we reported a correlation between cell adhesion molecule L1 (L1) expression and the occurrence of AD, but its relationship was unclear. Here, we report that the expression of L1 and a 70 kDa cleavage product of L1 (L1-70) was reduced in the hippocampus of AD (APPswe) mice. Interestingly, upregulation of L1-70 expression in the hippocampus of 18-month-old APPswe mice, by parabiosis involving the joining of the circulatory system of an 18-month-old APPswe mouse with a 2-month-old wild-type C57BL/6 mouse, reduced amyloid plaque deposition. Furthermore, the reduction was accompanied by the appearance of a high number of activated microglia. Mechanistically, we observed that L1-70 could combine with topoisomerase 1 (Top1) to form a complex, L1-70/Top1, that was able to regulate expression of macrophage migration inhibitory factor (MIF), resulting in the activation of microglia and reduction of Aβ plaques. Also, transforming growth factor β1 (TGFβ-1) transferred from the blood of young wild-type C57BL/6 mice to the aged AD mice, was identified as a circulating factor that induces full-length L1 and L1-70 expression. All together, these findings suggest that L1-70 contributes to the clearance of Aβ in AD, thereby adding a novel perspective in understanding AD pathogenesis.
Collapse
Affiliation(s)
- Junkai Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Stanley Li Lin
- Deaprtment of Cell Biology, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| |
Collapse
|
30
|
Zarbock KR, Han JH, Singh AP, Thomas SP, Bendlin BB, Denu JM, Yu JPJ, Rey FE, Ulland TK. Trimethylamine N-Oxide Reduces Neurite Density and Plaque Intensity in a Murine Model of Alzheimer's Disease. J Alzheimers Dis 2022; 90:585-597. [PMID: 36155509 PMCID: PMC9881463 DOI: 10.3233/jad-220413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common aging-associated neurodegenerative disease; nevertheless, the etiology and progression of the disease is still incompletely understood. We have previously shown that the microbially-derived metabolite trimethylamine N-oxide (TMAO) is elevated in the cerebrospinal fluid (CSF) of individuals with cognitive impairment due to AD and positively correlates with increases in CSF biomarkers for tangle, plaque, and neuronal pathology. OBJECTIVE We assessed the direct impact of TMAO on AD progression. METHODS To do so, transgenic 5XFAD mice were supplemented with TMAO for 12 weeks. Neurite density was assessed through quantitative brain microstructure imaging with neurite orientation dispersion and density imaging magnetic resonance imaging (MRI). Label-free, quantitative proteomics was performed on cortex lysates from TMAO-treated and untreated animals. Amyloid-β plaques, astrocytes, and microglia were assessed by fluorescent immunohistochemistry and synaptic protein expression was quantified via western blot. RESULTS Oral TMAO administration resulted in significantly reduced neurite density in several regions of the brain. Amyloid-β plaque mean intensity was reduced, while plaque count and size remained unaltered. Proteomics analysis revealed that TMAO treatment impacted the expression of 30 proteins (1.5-fold cut-off) in 5XFAD mice, including proteins known to influence neuronal health and amyloid-β precursor protein processing. TMAO treatment did not alter astrocyte and microglial response nor cortical synaptic protein expression. CONCLUSION These data suggest that elevated plasma TMAO impacts AD pathology via reductions in neurite density.
Collapse
Affiliation(s)
- Katie R. Zarbock
- University of Wisconsin-Madison, Department of Bacteriology
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine
| | - Jessica H. Han
- University of Wisconsin-Madison, Department of Bacteriology
- University of Wisconsin-Madison, Department of Biomolecular Chemistry
| | - Ajay P. Singh
- University of Wisconsin-Madison, Department of Radiology, Division of Neuroradiology
| | - Sydney P. Thomas
- University of Wisconsin-Madison, Department of Biomolecular Chemistry
| | - Barbara B. Bendlin
- University of Wisconsin-Madison, Department of Medicine, Division of Geriatrics and Gerontology
- Wisconsin Alzheimer’s Disease Research Center
| | - John M. Denu
- University of Wisconsin-Madison, Department of Biomolecular Chemistry
| | - John-Paul J. Yu
- University of Wisconsin-Madison, Department of Radiology, Division of Neuroradiology
| | | | - Tyler K. Ulland
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine
| |
Collapse
|
31
|
Ruan Z, Lu Q, Wang JE, Zhou M, Liu S, Zhang H, Durvasula A, Wang Y, Wang Y, Luo W, Wang Y. MIF promotes neurodegeneration and cell death via its nuclease activity following traumatic brain injury. Cell Mol Life Sci 2021; 79:39. [DOI: 10.1007/s00018-021-04037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 10/19/2022]
|
32
|
Sumaiya K, Langford D, Natarajaseenivasan K, Shanmughapriya S. Macrophage migration inhibitory factor (MIF): A multifaceted cytokine regulated by genetic and physiological strategies. Pharmacol Ther 2021; 233:108024. [PMID: 34673115 DOI: 10.1016/j.pharmthera.2021.108024] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine encoded within a functionally polymorphic genetic locus. MIF was initially recognized as a cytokine generated by activated T cells, but in recent days it has been identified as a multipotent key cytokine secreted by many other cell types involved in immune response and physiological processes. MIF is a highly conserved 12.5 kDa secretory protein that is involved in numerous biological processes. The expression and secretion profile of MIF suggests that MIF to be ubiquitously and constitutively expressed in almost all mammalian cells and is vital for numerous physiological processes. MIF is a critical upstream mediator of host innate and adaptive immunity and survival pathways resulting in the clearance of pathogens thus playing a protective role during infectious diseases. On the other hand, MIF being an immune modulator accelerates detrimental inflammation, promotes cancer metastasis and progression, thus worsening disease conditions. Several reports demonstrated that genetic and physiological factors, including MIF gene polymorphisms, posttranslational regulations, and receptor binding control the functional activities of MIF. Taking into consideration the multi-faceted role of MIF both in physiology and pathology, we thought it is timely to review and summarize the expressional and functional regulation of MIF, its functional mechanisms associated with its beneficial and pathological roles, and MIF-targeting therapies. Thus, our review will provide an overview on how MIF is regulated, its response, and the potency of the therapies that target MIF.
Collapse
Affiliation(s)
- Krishnamoorthi Sumaiya
- Medical Microbiology Laboratory, Department of Microbiology, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Dianne Langford
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kalimuthusamy Natarajaseenivasan
- Medical Microbiology Laboratory, Department of Microbiology, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA..
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey PA-17033, USA.
| |
Collapse
|
33
|
Fei X, Zhang Y, Mei Y, Yue X, Jiang W, Ai L, Yu Y, Luo H, Li H, Luo W, Yang X, Lyv J, He R, Song W, Tong Z. Degradation of FA reduces Aβ neurotoxicity and Alzheimer-related phenotypes. Mol Psychiatry 2021; 26:5578-5591. [PMID: 33328587 DOI: 10.1038/s41380-020-00929-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 02/05/2023]
Abstract
Dysregulation of formaldehyde (FA) has been implicated in the development of Alzheimer's Disease (AD). Elevated FA levels in Alzheimer's patients and animal models are associated with impaired cognitive functions. However, the exact role of FA in AD remains unknown. We now identified that oxidative demethylation at serine8/26 of amyloid-beta protein (Aβ) induced FA generation and FA cross-linked with the lysine28 residue in the β-turn of Aβ monomer to form Aβ dimers, and then accelerated Aβ oligomerization and fibrillogenesis in vitro. However, Aβ42 mutation in serine8/26, lysine28 abolished Aβ self-aggregation. Furthermore, Aβ inhibited the activity of formaldehyde dehydrogenase (FDH), the enzyme for FA degradation, resulting in FA accumulation. In turn, excess of FA stimulated Aβ aggregation both in vitro and in vivo by increasing the formation of Aβ oligomers and fibrils. We found that degradation of FA by formaldehyde scavenger-NaHSO3 or coenzyme Q10 reduced Aβ aggregation and ameliorated the neurotoxicity, and improved the cognitive performance in APP/PS1 mice. Our study provides evidence that endogenous FA is essential for Aβ self-aggregation and scavenging FA could be an effective strategy for treating AD.
Collapse
Affiliation(s)
- Xuechao Fei
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Yun Zhang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yufei Mei
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiangpei Yue
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Wenjing Jiang
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
- Center for Cognitive Disorders, Beijing Geriatric Hospital, 100095, Beijing, China
| | - Li Ai
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Yan Yu
- Chinese institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, 100068, Beijing, China
| | - Hongjun Luo
- Central Laboratory, Shantou University Medical College, Guangdong, 515041, China
| | - Hui Li
- Central Laboratory, Shantou University Medical College, Guangdong, 515041, China
| | - Wenhong Luo
- Central Laboratory, Shantou University Medical College, Guangdong, 515041, China
| | - Xu Yang
- Section of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Jihui Lyv
- Center for Cognitive Disorders, Beijing Geriatric Hospital, 100095, Beijing, China
| | - Rongqiao He
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
- State Key Lab of Brain and Cognitive Science and Key Lab of Mental Health, IBP, UCAS, Beijing, China
| | - Weihong Song
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Zhiqian Tong
- Alzheimer's disease Center, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
34
|
Stem cells from human exfoliated deciduous teeth affect mitochondria and reverse cognitive decline in a senescence-accelerated mouse prone 8 model. Cytotherapy 2021; 24:59-71. [PMID: 34598900 DOI: 10.1016/j.jcyt.2021.07.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/31/2021] [Accepted: 07/31/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND AIMS Stem cell therapy is a novel therapy being explored for AD. The molecular mechanism of its effect is still unclear. The authors investigated the effects and mechanism by injection of SHEDs into an AD mouse model. METHODS SHEDs were cultured in vitro and injected into AD SAMP8 mice by caudal vein, and SHEDs labeled via synthetic dye showed in vivo migration to the head. The cognitive ability of SAMP8 mice was evaluated via Barnes maze and new object recognition. The pathological indicators of AD, including Tau, amyloid plaques and inflammatory factors, were examined at the protein or RNA level. Next, macro-proteomics analysis and weighted gene co-expression network analysis (WGCNA) based on protein groups and behavioral data were applied to discover the important gene cluster involved in the improvement of AD by SHEDs, which was further confirmed in an AD model in both mouse and cell lines. RESULTS SHED treatment improved the cognitive ability and pathological symptoms of SAMP8 mice. Proteomics analysis indicated that these improvements were tightly related to the mitochondria, which was proved through examination of the shape and function of mitochondria both in vivo (SAMP8 brain) and in vitro (SH-SY5Y cells). Finally, the core targets of SHEDs in the mitochondrial pathway, Hook3, Mic13 and MIF, were screened out and confirmed in vivo. CONCLUSIONS SHED treatment significantly relieved AD symptoms, improved cognitive ability and reversed memory loss in an AD mouse model, possibly through the recovery of dysfunctional mitochondria. These results raise the possibility that SHED may ease the symptoms of AD by targeting the mitochondria.
Collapse
|
35
|
Yeung SSH, Ho YS, Chang RCC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med 2021; 53:1251-1267. [PMID: 34489558 PMCID: PMC8492689 DOI: 10.1038/s12276-021-00660-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.
Collapse
Affiliation(s)
- Sherry Sin-Hang Yeung
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Yuen-Shan Ho
- grid.16890.360000 0004 1764 6123School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR China
| | - Raymond Chuen-Chung Chang
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| |
Collapse
|
36
|
van der Linden RJ, De Witte W, Poelmans G. Shared Genetic Etiology between Alzheimer's Disease and Blood Levels of Specific Cytokines and Growth Factors. Genes (Basel) 2021; 12:genes12060865. [PMID: 34198788 PMCID: PMC8226721 DOI: 10.3390/genes12060865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 01/24/2023] Open
Abstract
Late-onset Alzheimer’s disease (AD) has a significant genetic and immunological component, but the molecular mechanisms through which genetic and immunity-related risk factors and their interplay contribute to AD pathogenesis are unclear. Therefore, we screened for genetic sharing between AD and the blood levels of a set of cytokines and growth factors to elucidate how the polygenic architecture of AD affects immune marker profiles. For this, we retrieved summary statistics from Finnish genome-wide association studies of AD and 41 immune marker blood levels and assessed for shared genetic etiology, using a polygenic risk score-based approach. For the blood levels of 15 cytokines and growth factors, we identified genetic sharing with AD. We also found positive and negative genetic concordances—implying that genetic risk factors for AD are associated with higher and lower blood levels—for several immune markers and were able to relate some of these results to the literature. Our results imply that genetic risk factors for AD also affect specific immune marker levels, which may be leveraged to develop novel treatment strategies for AD.
Collapse
|
37
|
Delaney C, Farrell M, Doherty CP, Brennan K, O’Keeffe E, Greene C, Byrne K, Kelly E, Birmingham N, Hickey P, Cronin S, Savvides SN, Doyle SL, Campbell M. Attenuated CSF-1R signalling drives cerebrovascular pathology. EMBO Mol Med 2021; 13:e12889. [PMID: 33350588 PMCID: PMC7863388 DOI: 10.15252/emmm.202012889] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Cerebrovascular pathologies occur in up to 80% of cases of Alzheimer's disease; however, the underlying mechanisms that lead to perivascular pathology and accompanying blood-brain barrier (BBB) disruption are still not fully understood. We have identified previously unreported mutations in colony stimulating factor-1 receptor (CSF-1R) in an ultra-rare autosomal dominant condition termed adult-onset leucoencephalopathy with axonal spheroids and pigmented glia (ALSP). Cerebrovascular pathologies such as cerebral amyloid angiopathy (CAA) and perivascular p-Tau were some of the primary neuropathological features of this condition. We have identified two families with different dominant acting alleles with variants located in the kinase region of the CSF-1R gene, which confer a lack of kinase activity and signalling. The protein product of this gene acts as the receptor for 2 cognate ligands, namely colony stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). Here, we show that depletion in CSF-1R signalling induces BBB disruption and decreases the phagocytic capacity of peripheral macrophages but not microglia. CSF-1R signalling appears to be critical for macrophage and microglial activation, and macrophage localisation to amyloid appears reduced following the induction of Csf-1r heterozygosity in macrophages. Finally, we show that endothelial/microglial crosstalk and concomitant attenuation of CSF-1R signalling causes re-modelling of BBB-associated tight junctions and suggest that regulating BBB integrity and systemic macrophage recruitment to the brain may be therapeutically relevant in ALSP and other Alzheimer's-like dementias.
Collapse
Affiliation(s)
- Conor Delaney
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Michael Farrell
- Department of NeuropathologyBeaumont HospitalDublin 9Ireland
| | - Colin P Doherty
- Department of NeurologyHealth Care CentreSt James's HospitalDublin 8Ireland
- Academic Unit of NeurologyBiomedical Sciences InstituteTrinity College DublinDublin 2Ireland
- FutureNeuro SFI Research CentreRoyal College of Surgeons in IrelandDublinIreland
| | - Kiva Brennan
- Trinity College Institute of NeuroscienceTrinity College Dublin 2Dublin 2Ireland
| | - Eoin O’Keeffe
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Chris Greene
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Kieva Byrne
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Eoin Kelly
- Department of NeurologyHealth Care CentreSt James's HospitalDublin 8Ireland
| | | | | | - Simon Cronin
- Department of MedicineUniversity College CorkCorkIreland
| | - Savvas N Savvides
- Unit for Structural BiologyDepartment of Biochemistry and MicrobiologyGhent UniversityGhentBelgium
- VIB‐UGent Center for Inflammation ResearchGhentBelgium
| | - Sarah L Doyle
- Trinity College Institute of NeuroscienceTrinity College Dublin 2Dublin 2Ireland
| | - Matthew Campbell
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
- FutureNeuro SFI Research CentreRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
38
|
McQuade A, Kang YJ, Hasselmann J, Jairaman A, Sotelo A, Coburn M, Shabestari SK, Chadarevian JP, Fote G, Tu CH, Danhash E, Silva J, Martinez E, Cotman C, Prieto GA, Thompson LM, Steffan JS, Smith I, Davtyan H, Cahalan M, Cho H, Blurton-Jones M. Gene expression and functional deficits underlie TREM2-knockout microglia responses in human models of Alzheimer's disease. Nat Commun 2020; 11:5370. [PMID: 33097708 PMCID: PMC7584603 DOI: 10.1038/s41467-020-19227-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
The discovery of TREM2 as a myeloid-specific Alzheimer's disease (AD) risk gene has accelerated research into the role of microglia in AD. While TREM2 mouse models have provided critical insight, the normal and disease-associated functions of TREM2 in human microglia remain unclear. To examine this question, we profile microglia differentiated from isogenic, CRISPR-modified TREM2-knockout induced pluripotent stem cell (iPSC) lines. By combining transcriptomic and functional analyses with a chimeric AD mouse model, we find that TREM2 deletion reduces microglial survival, impairs phagocytosis of key substrates including APOE, and inhibits SDF-1α/CXCR4-mediated chemotaxis, culminating in an impaired response to beta-amyloid plaques in vivo. Single-cell sequencing of xenotransplanted human microglia further highlights a loss of disease-associated microglial (DAM) responses in human TREM2 knockout microglia that we validate by flow cytometry and immunohistochemistry. Taken together, these studies reveal both conserved and novel aspects of human TREM2 biology that likely play critical roles in the development and progression of AD.
Collapse
Affiliation(s)
- Amanda McQuade
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - You Jung Kang
- Department of Mechanical Engineering and Engineering Science, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Department of Biological Sciences, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Center for Biomedical Engineering and Science, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
| | - Jonathan Hasselmann
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Amit Jairaman
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, 92697, USA
| | - Alexandra Sotelo
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Morgan Coburn
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Sepideh Kiani Shabestari
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Jean Paul Chadarevian
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Gianna Fote
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Christina H Tu
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Emma Danhash
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Jorge Silva
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Eric Martinez
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Carl Cotman
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - G Aleph Prieto
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
- Institute of Neurobiology, National Autonomous University of Mexico, Queretaro, Mexico
| | - Leslie M Thompson
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
- Department of Biological Sciences, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
| | - Joan S Steffan
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
- Department of Psychology and Human Behavior, University of California Irvine, Irvine, CA, 92697, USA
| | - Ian Smith
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA
| | - Hayk Davtyan
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA
| | - Michael Cahalan
- Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, 92697, USA
| | - Hansang Cho
- Department of Mechanical Engineering and Engineering Science, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Department of Biological Sciences, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Center for Biomedical Engineering and Science, University of North Carolina Charlotte, Charlotte, NC, 28223, USA
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, 92697, USA.
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
39
|
Inhibition of Macrophage Migration Inhibitory Factor Protects against Inflammation through a Toll-like Receptor-Related Pathway after Diffuse Axonal Injury in Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5946205. [PMID: 32964038 PMCID: PMC7492939 DOI: 10.1155/2020/5946205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/19/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
Objective We have previously demonstrated that inflammation induced by toll-like receptors (TLRs) 2/4 exert cerebral deleterious effects after diffuse axonal injury (DAI); however, the underlying mechanisms are not fully understood. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine involved in inflammatory responses. The purpose of this study was to investigate the role of MIF in inflammation induced by TLRs in the cortices of DAI rats. Methods The rat DAI model was established by head rotational acceleration and confirmed by β-APP, HE, and silver staining. MIF protein expression at 3 h, 6 h, 12 h, 1 d, and 3 d after DAI was measured by western blot. The localization of MIF was measured by immunofluorescence. MIF antagonist ISO-1 was intracerebroventricularly injected to inhibit MIF. Neuronal and axonal injury and glial responses were assessed by TUNEL, immunohistochemistry, and TEM. Expression of TLR2, TLR4, ERK, phospho-ERK, NF-κB, and phospho-NF-κB was examined by western blot. The level of IL-1β, IL-6, and TNF-α was measured by ELISA. Results MIF expression was significantly increased, peaking at 1 day after DAI, and MIF was mainly localized in microglial cells and neurons. ISO-1 suppressed neuronal apoptosis, axonal injury, and glial responses and decreased the expression of downstream signaling molecules related to TLR2/4, including ERK, phospho-ERK, NF-κB, phospho-NF-κB, IL-1β, IL-6, and TNF-α. Conclusion MIF was involved in the neuronal and axonal damage through a TLR-related pathway following DAI.
Collapse
|
40
|
Chai X, Zhang W, Li L, Wu Y, Zhu X, Zhao S. Profile of MIF in Developing Hippocampus: Association With Cell Proliferation and Neurite Outgrowth. Front Mol Neurosci 2020; 13:147. [PMID: 32903462 PMCID: PMC7434973 DOI: 10.3389/fnmol.2020.00147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/15/2020] [Indexed: 11/13/2022] Open
Abstract
Proinflammatory cytokine macrophage migration inhibitory factor (MIF) is a multifunctional cytokine and has been found involved in many neurological diseases such as Alzheimer disease (AD), epilepsy, and multiple sclerosis. Previous studies have shown that MIF is expressed in neocortex, hippocampus, hypothalamus, cerebellum, and spinal cord in adult mice. It is expressed by astrocytes and activates microglias in neuroinflammation. Further studies have shown that MIF is detected in moss fibers of dentate granule cells and in apical dendrites of pyramidal neurons in adult hippocampus. Only NeuroD-positive immature granule neurons but not NeuN-positive mature neurons express MIF. These findings led us eager to know the exact role of MIF in the development of hippocampus. Therefore, we systematically checked the spatial and temporal expression pattern of MIF and characterized MIF-positive cells in hippocampus from mice aged from postnatal day 0 (P0) to 3 months. Our results showed that the lowest level of MIF protein occurred at P7 and mif mRNA increased from P0, reached a peak at P7, and stably expressed until P30 before declining dramatically at 3 months. MIF was localized in fibers of GFAP- and BLBP-positive radial glial precursor cells in dentate gyrus (DG). DCX-expressing newly generated neurons were MIF-negative. Inhibition of MIF by MIF antagonist S, R-3-(4-hydroxyphenyl)-4, 5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) reduced BrdU-positive cells. Interestingly, MIF was expressed by NeuN-positive GABAergic interneurons including parvalbumin-and Reelin-expressing cells in the DG. Neither NeuN-positive granule cells nor NeuN-positive pyramidal neurons expressed MIF. In transgenic mice, POMC-EGFP–positive immature dentate granule cells and Thy1-EGFP–positive mature granule cells were MIF-negative. Treatment of neuronal cultures with ISO-1 inhibited neurite outgrowth. Therefore, we conclude that MIF might be important for feature maintenance of neural stem cells and neurite outgrowth during hippocampal development.
Collapse
Affiliation(s)
- Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, China
| | - Wei Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lingling Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
41
|
The Role of Macrophage Migration Inhibitory Factor in Alzheimer's Disease: Conventionally Pathogenetic or Unconventionally Protective? Molecules 2020; 25:molecules25020291. [PMID: 31936865 PMCID: PMC7024279 DOI: 10.3390/molecules25020291] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Recent preclinical and clinical observations have offered relevant insights on the etiopathogenesis of late onset Alzheimer′s disease (AD) and upregulated immunoinflammatory events have been described as underlying mechanisms involved in the development of AD. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine produced by several cells of the innate and adaptive immune system, as well as non-immune cells. In the present review, we highlight experimental, genetic, and clinical studies on MIF in rodent models of AD and AD patients, and we discuss emerging therapeutic opportunities for tailored modulation of the activity of MIF, that may potentially be applied to AD patients. Dismantling the exact role of MIF and its receptors in AD may offer novel diagnostic and therapeutic opportunities in AD.
Collapse
|
42
|
Macrophage Migration Inhibitory Factor Alters Functional Properties of CA1 Hippocampal Neurons in Mouse Brain Slices. Int J Mol Sci 2019; 21:ijms21010276. [PMID: 31906137 PMCID: PMC6981710 DOI: 10.3390/ijms21010276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is implicated in a host of neurological insults, such as traumatic brain injury (TBI), ischemic stroke, Alzheimer's disease, Parkinson's disease, and epilepsy. The immune response to central nervous system (CNS) injury involves sequelae including the release of numerous cytokines and chemokines. Macrophage migration inhibitory factor (MIF), is one such cytokine that is elevated following CNS injury, and is associated with the prognosis of TBI, and ischemic stroke. MIF has been identified in astrocytes and neurons, and some of the trophic actions of MIF have been related to its direct and indirect actions on astrocytes. However, the potential modulation of CNS neuronal function by MIF has not yet been explored. This study tests the hypothesis that MIF can directly influence hippocampal neuronal function. MIF was microinjected into the hippocampus and the genetically encoded calcium indicator, GCaMP6f, was used to measure Ca2+ events in acute adult mouse brain hippocampal slices. Results demonstrated that a single injection of 200 ng MIF into the hippocampus significantly increased baseline calcium signals in CA1 pyramidal neuron somata, and altered calcium responses to N-methyl-d-aspartate (NMDA) + D-serine in pyramidal cell apical dendrites located in the stratum radiatum. These data are the first to show direct effects of MIF on hippocampal neurons and on NMDA receptor function. Considering that MIF is elevated after brain insults such as TBI, the data suggest that, in addition to the previously described role of MIF in astrocyte reactivity, elevated MIF can have significant effects on neuronal function in the hippocampus.
Collapse
|
43
|
Belmonte KCD, Harman JC, Lanson NA, Gidday JM. Intra- and intergenerational changes in the cortical DNA methylome in response to therapeutic intermittent hypoxia in mice. Physiol Genomics 2019; 52:20-34. [PMID: 31762411 DOI: 10.1152/physiolgenomics.00094.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent evidence from our laboratory documents functional resilience to retinal ischemic injury in untreated mice derived from parents exposed to repetitive hypoxic conditioning (RHC) before breeding. To begin to understand the epigenetic basis of this intergenerational protection, we used methylated DNA immunoprecipitation and sequencing to identify genes with differentially methylated promoters (DMGPs) in the prefrontal cortex of mice treated directly with the same RHC stimulus (F0-RHC) and in the prefrontal cortex of their untreated F1-generation offspring (F1-*RHC). Subsequent bioinformatic analyses provided key mechanistic insights into how changes in gene expression secondary to promoter hypo- and hypermethylation might afford such protection within and across generations. We found extensive changes in DNA methylation in both generations consistent with the expression of many survival-promoting genes, with twice the number of DMGPs in the cortex of F1*RHC mice relative to their F0 parents that were directly exposed to RHC. In contrast to our hypothesis that similar epigenetic modifications would be realized in the cortices of both F0-RHC and F1-*RHC mice, we instead found relatively few DMGPs common to both generations; in fact, each generation manifested expected injury resilience via distinctly unique gene expression profiles. Whereas in the cortex of F0-RHC mice, predicted protein-protein interactions reflected activation of an anti-ischemic phenotype, networks activated in F1-*RHC cortex comprised networks indicative of a much broader cytoprotective phenotype. Altogether, our results suggest that the intergenerational transfer of an acquired phenotype to offspring does not necessarily require the faithful recapitulation of the conditioning-modified DNA methylome of the parent.
Collapse
Affiliation(s)
- Krystal Courtney D Belmonte
- Department of Ophthalmology, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana.,Department of Physiology, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana
| | - Jarrod C Harman
- Department of Ophthalmology, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana.,Neuroscience Center of Excellence, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana
| | - Nicholas A Lanson
- Department of Ophthalmology, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana
| | - Jeffrey M Gidday
- Department of Ophthalmology, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana.,Department of Physiology, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana.,Neuroscience Center of Excellence, Louisiana State University School of Medicine, Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|