1
|
Medeiros-Furquim T, Miedema A, Schilder E, Brouwer N, Holtman IR, Kooistra SM, Eggen BJL. Microglia endotoxin tolerance is retained after enforced repopulation. Brain Behav Immun 2025; 128:512-528. [PMID: 40274001 DOI: 10.1016/j.bbi.2025.04.014] [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] [Received: 07/24/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Microglia are crucial for CNS homeostasis and are involved in a wide range of neurodegenerative and neuroinflammatory diseases. Systemic inflammation and infections can contribute to neurodegeneration later in life by affecting microglia. Like other innate immune cells, microglia can develop innate immune memory (IIM) in response to an inflammatory challenge, altering their response to subsequent stimuli. IIM can ameliorate or worsen CNS pathology, but it is unclear if IIM can be reversed to restore microglia functions. Here, we investigated whether microglia depletion-repopulation by inhibition of the colony-stimulating factor 1 receptor with BLZ945 reversed LPS-induced microglia endotoxin tolerance in mice. Repopulated microglia displayed a reduced expression of homeostatic genes and genes related to mitochondrial respiration and TCA cycle metabolism and an increased expression of immune effector and activation genes. Nonetheless, the blunted inflammatory gene response after LPS-preconditioning was retained after a depletion-repopulation cycle. Our study highlights the persistence of endotoxin tolerance in microglia after a depletion-repopulation cycle, which might impact the potential effectiveness of strategies targeted at microglia depletion for clinical applications.
Collapse
Affiliation(s)
- Tiago Medeiros-Furquim
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anneke Miedema
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Edwin Schilder
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge R Holtman
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Susanne M Kooistra
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Bart J L Eggen
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
2
|
Tenner AJ, Petrisko TJ. Knowing the enemy: strategic targeting of complement to treat Alzheimer disease. Nat Rev Neurol 2025:10.1038/s41582-025-01073-y. [PMID: 40128350 DOI: 10.1038/s41582-025-01073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/26/2025]
Abstract
The complement system protects against infection, positively responds to tissue damage, clears cell debris, directs and modulates the adaptive immune system, and functions in neuronal development, normal synapse elimination and intracellular metabolism. However, complement also has a role in aberrant synaptic pruning and neuroinflammation - processes that lead to a feedforward loop of inflammation, injury and neuronal death that can contribute to neurodegenerative and neurological disorders, including Alzheimer disease. This Review provides justification, largely from preclinical mouse models but also from correlates with human tissue and biomarkers, for targeting specific complement components for therapeutic intervention in Alzheimer disease. We discuss promising strategies to slow the progression of cognitive loss with minimal undesired effects. The diverse interactions and functions of complement system components can influence biological processes in the healthy and diseased brain; here, these functions are described as a prerequisite to selecting appropriate, safe and effective therapeutic targets for translation to the clinic.
Collapse
Affiliation(s)
- Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA.
| | - Tiffany J Petrisko
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
3
|
Tao W, Zhang Y, Wang B, Nie S, Fang L, Xiao J, Wu Y. Advances in molecular mechanisms and therapeutic strategies for central nervous system diseases based on gut microbiota imbalance. J Adv Res 2025; 69:261-278. [PMID: 38579985 PMCID: PMC11954836 DOI: 10.1016/j.jare.2024.03.023] [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: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUD Central nervous system (CNS) diseases pose a serious threat to human health, but the regulatory mechanisms and therapeutic strategies of CNS diseases need to be further explored. It has been demonstrated that the gut microbiota (GM) is closely related to CNS disease. GM structure disorders, abnormal microbial metabolites, intestinal barrier destruction and elevated inflammation exist in patients with CNS diseases and promote the development of CNS diseases. More importantly, GM remodeling alleviates CNS pathology to some extent. AIM OF REVIEW Here, we have summarized the regulatory mechanism of the GM in CNS diseases and the potential treatment strategies for CNS repair based on GM regulation, aiming to provide safer and more effective strategies for CNS repair from the perspective of GM regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The abundance and composition of GM is closely associated with the CNS diseases. On the basis of in-depth analysis of GM changes in mice with CNS disease, as well as the changes in its metabolites, therapeutic strategies, such as probiotics, prebiotics, and FMT, may be used to regulate GM balance and affect its microbial metabolites, thereby promoting the recovery of CNS diseases.
Collapse
Affiliation(s)
- Wei Tao
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Yanren Zhang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Bingbin Wang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Saiqun Nie
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Li Fang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
4
|
Liu X, Tan X, Yu Y, Niu J, Zhao B, Wang Q. Short chain fatty acids mediates complement C1q pathway alleviation of perioperative neurocognitive disorders. Neuropharmacology 2025; 265:110266. [PMID: 39681213 DOI: 10.1016/j.neuropharm.2024.110266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/18/2024]
Abstract
Perioperative neurocognitive disorders (PND) is one of the most common postoperative complications, which can lead to a harmful impact on self-dependence, longer hospital stays, increased medical costs, morbidity, and mortality amongst older adults. Microglia can modulate synapse elimination involved in the complement component protein 1q (C1q) pathway to induce cognitive dysfunction, which is significantly improved by short chain fatty acids (SCFAs) treatment. Here we investigate the effects of SCFAs treatment on PND via mediating C1q complement pathway. High-throughput sequencing of 16S rDNA from fecal samples of male SD rats was applied to assess the changes in gut microbiota. Fecal microbiota transplantation (FMT) was performed to investigate whether gut microbiota from PND rats could alter cognitive impairment. The blood from the rat tail vein was collected to measure the SCFAs concentrations. Hippocampal and brain tissue samples were obtained to perform Western blots, Golgi and immunofluorescence staining. Primary microglia treated with SCFAs or Histone deacetylase inhibitor were cultured to measure microglial activation states and the expression of acetylated histone. The 16S rDNA sequencing results showed that PND rats had the significant changes in the species diversity of the gut microbiota and the metabolite of specifc species. Gut microbiota from PND rats could alter spatial learning and memory, and meanwhile, the changed SCFAs concentrations in plasma were involved. The synapse elimination in PND rats was strikingly reversed by SCFAs treatment involved in modulation complement C1q via suppressing neuroinflammation. This suggests that a link between gut microbiota dysbiosis and cognitive function impairment is involved in synapse elimination via mediating complement C1q pathway. SCFAs treatment can alleviate PND, the mechanisms of which may be associated with regulating complement C1q pathway.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Anesthesiology, Hebei Medical University Third Hospital, Shijiazhuang, 050051, Hebei Province, PR China; Department of Anesthesiology, Hebei Children's Hospital, Shi Jiazhuang, 050031, Hebei Province, PR China
| | - Xiaona Tan
- Department of Neurological Rehabilitation, Hebei Children's Hospital, Shi Jiazhuang, 050031, Hebei Province, PR China
| | - Yaozong Yu
- Department of Anesthesiology, Hebei Medical University Third Hospital, Shijiazhuang, 050051, Hebei Province, PR China
| | - Junfang Niu
- Department of Anesthesiology, Hebei Medical University Third Hospital, Shijiazhuang, 050051, Hebei Province, PR China
| | - Bo Zhao
- Experimental Centre for Teaching, Hebei Medical University, Shi Jiazhuang, 050000, Hebei Province, PR China
| | - Qiujun Wang
- Department of Anesthesiology, Hebei Medical University Third Hospital, Shijiazhuang, 050051, Hebei Province, PR China.
| |
Collapse
|
5
|
Sona C, Yeh YT, Li Y, Liu X, Ghosh A, Hinte LC, Ku MC, Rathjen T, Niendorf T, Yu G, Jia S, Kononenko NL, Hermann A, Luo J, Lin J, von Meyenn F, Yan X, Poy MN. Glutamatergic argonaute2 promotes the formation of the neurovascular unit in mice. Sci Signal 2025; 18:eadl6745. [PMID: 39999211 DOI: 10.1126/scisignal.adl6745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Proper formation of the complex neurovascular unit (NVU) along with the blood-brain barrier is critical for building and sustaining a healthy, functioning central nervous system. The RNA binding protein argonaute2 (Ago2) mediates microRNA (miRNA)-mediated gene silencing, which is critical for many facets of brain development, including NVU development. Here, we found that Ago2 in glutamatergic neurons was critical for NVU formation in the developing cortices of mice. Glutamatergic neuron-specific loss of Ago2 diminished synaptic formation, neuronal-to-endothelial cell contacts, and morphogenesis of the brain vasculature, ultimately compromising the integrity of the blood-brain barrier. Ago2 facilitated miRNA targeting of phosphatase and tensin homolog (Pten) mRNA, which encodes a phosphatase that modulates reelin-dependent phosphatidylinositol 3-kinase (PI3K)-Akt signaling within the glutamatergic subpopulation. Conditionally deleting Pten in Ago2-deficient neurons restored Akt2 phosphorylation as well as postnatal development and survival. Several mutations in AGO2 impair small RNA silencing and are associated with Lessel-Kreienkamp syndrome, a neurodevelopmental disorder. When expressed in a neuronal cell line, these human AGO2 loss-of-function variants failed to suppress PTEN, resulting in attenuated PI3K-Akt signaling, further indicating that dysregulation of Ago2 function may contribute to both impaired development and neurological disorders. Together, these results identify Ago2 as central to the engagement of neurons with blood vessels in the developing brain.
Collapse
Affiliation(s)
- Chandan Sona
- All Children's Hospital, Johns Hopkins University, St. Petersburg, FL 33701, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Yu-Te Yeh
- All Children's Hospital, Johns Hopkins University, St. Petersburg, FL 33701, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Yunxiao Li
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
| | - Xiaoxuan Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Adhideb Ghosh
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Laura C Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Min-Chi Ku
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Thomas Rathjen
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Thoralf Niendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Guoxing Yu
- Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Shiqi Jia
- Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Natalia L Kononenko
- CECAD Excellence Center & Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne 50931, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock 17489, Germany
| | - Jiankai Luo
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach 8603, Switzerland
| | - Xin Yan
- Translational Neurodegeneration Section, "Albrecht Kossel," University Medical Center Rostock, University of Rostock, Rostock 18147, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| | - Matthew N Poy
- All Children's Hospital, Johns Hopkins University, St. Petersburg, FL 33701, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21287, USA
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, Berlin 13125, Germany
| |
Collapse
|
6
|
Rao B, Liu X, Xiao J, Wu X, He F, Yang Q, Zhao W, Lin X, Zhang J. Microglia heterogeneity during neuroinflammation and neurodegeneration in the mouse retina. Brain Struct Funct 2024; 230:19. [PMID: 39720969 DOI: 10.1007/s00429-024-02875-4] [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: 06/13/2024] [Accepted: 10/15/2024] [Indexed: 12/26/2024]
Abstract
Microglia play important roles in maintaining homeostasis and immunoreactive defense in the central nervous system including retina. To accomplish such a wide range of functions, microglia are highly heterogeneous. Dark microglia (DM) were recently identified by electron microscopy (EM). However, the specific correlation between microglial morphological phenotypes, including DM, and physiological or pathological conditions remains poorly understood. We established acute and chronic neuroinflammatory models by Lipopolysaccharide (LPS) and light-induced photoreceptor neurodegeneration model to explore these questions in the mouse retina. Immunofluorescence and EM were used to detect microglia in these models. Our light microscopy (LM) results reveal that the withdrawal phenotype is predominant in acute neuroinflammation models, both in vitro and in vivo, while the dystrophic microglia are the major phenotype in chronic neuroinflammation and neurodegeneration models in vivo. Ultrastructurally, acute models exhibit high electron dense processes, but not somas, while chronic models show high electron dense somas and processes. Given the consistency between LM and EM, we propose that DM-like somas and processes likely indicate a dystrophic population. It's important to note, however, that DM may not represent a single specific microglia phenotype, but rather a dynamic transformation of gradually activated microglia. Finally, we provide evidence for the presence of DM in mouse retinas in the neuroinflammatory model and the neurodegenerative model. This research provides valuable insights into investigating microglia phenotypes through both LM and EM.
Collapse
Affiliation(s)
- Bilin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoqing Liu
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiayi Xiao
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaotian Wu
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Fang He
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingwen Yang
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenna Zhao
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jun Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
7
|
Batey L, Baumberger B, Khoshbouei H, Hashemi P. Lipopolysaccharide Effects on Neurotransmission: Understanding Implications for Depression. ACS Chem Neurosci 2024; 15:4339-4347. [PMID: 39601433 PMCID: PMC11660149 DOI: 10.1021/acschemneuro.4c00591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Immune activation in the body is well studied; however, much less is known about how peripheral inflammation changes brain chemistry. Because depression and inflammation are close comorbidities, investigating how inflammation affects the brain's chemicals will help us to better understand depression. The levels of the monoamines dopamine, serotonin and norepinephrine are thought to be affected by both inflammation and depression. In this Perspective, we review studies that find chemical changes in the brain after administration of the endotoxin LPS, which is a robust method to induce rapid inflammation. From these studies, we interpreted LPS to reduce dopamine and serotonin and increase norepinephrine levels in various regions in the brain. These changes are not a sign of "dysfunction" but serve an important evolutionary purpose that encourages the body to recover from an immune insult by altering mood.
Collapse
Affiliation(s)
- L. Batey
- Department
of Bioengineering, Imperial College, South Kensington, London, SW7 2AZ, U.K.
| | - B. Baumberger
- Department
of Bioengineering, Imperial College, South Kensington, London, SW7 2AZ, U.K.
| | - H. Khoshbouei
- Department
of Neuroscience, University of Florida College
of Medicine, Gainesville, Florida 32610, United States
| | - P. Hashemi
- Department
of Bioengineering, Imperial College, South Kensington, London, SW7 2AZ, U.K.
| |
Collapse
|
8
|
Massaro Cenere M, Tiberi M, Paldino E, D'Addario SL, Federici M, Giacomet C, Cutuli D, Matteocci A, Cossa F, Zarrilli B, Casadei N, Ledonne A, Petrosini L, Berretta N, Fusco FR, Chiurchiù V, Mercuri NB. Systemic inflammation accelerates neurodegeneration in a rat model of Parkinson's disease overexpressing human alpha synuclein. NPJ Parkinsons Dis 2024; 10:213. [PMID: 39500895 PMCID: PMC11538257 DOI: 10.1038/s41531-024-00824-w] [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: 02/05/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Increasing efforts have been made to elucidate how genetic and environmental factors interact in Parkinson's disease (PD). In the present study, we assessed the development of symptoms on a genetic PD rat model that overexpresses human α-synuclein (Snca+/+) at a presymptomatic age, exposed to a pro-inflammatory insult by intraperitoneal injection of lipopolysaccharide (LPS), using immunohistology, high-dimensional flow cytometry, constant potential amperometry, and behavioral analyses. A single injection of LPS into WT and Snca+/+ rats triggered long-lasting increase in the activation of pro-inflammatory microglial markers, monocytes, and T lymphocytes. However, only LPS Snca+/+ rats showed dopaminergic neuronal loss in the substantia nigra pars compacta (SNpc), associated with a reduction in the release of evoked dopamine in the striatum. No significant changes were observed in the behavioral domain. We propose our double-hit animal as a reliable model to investigate the mechanisms whereby α-synuclein and inflammation interact to promote neurodegeneration in PD.
Collapse
Affiliation(s)
- Mariangela Massaro Cenere
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Marta Tiberi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Emanuela Paldino
- Laboratory of Neuroanatomy, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Sebastian Luca D'Addario
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Mauro Federici
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Cecilia Giacomet
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Debora Cutuli
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
- PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Francesca Cossa
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatrice Zarrilli
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Petrosini
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Nicola Berretta
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Nicola B Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
9
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
10
|
Lin YY, Chang WH, Hsieh SL, Cheng IHJ. The deficient CLEC5A ameliorates the behavioral and pathological deficits via the microglial Aβ clearance in Alzheimer's disease mouse model. J Neuroinflammation 2024; 21:273. [PMID: 39443966 PMCID: PMC11515658 DOI: 10.1186/s12974-024-03253-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease that causes cognitive dysfunction in older adults. One of the AD pathological factors, β-Amyloid (Aβ), triggers inflammatory responses and phagocytosis of microglia. C-type lectin domain family 5 member A (CLEC5A) induces over-reactive inflammatory responses in several virus infections. Yet, the role of CLEC5A in AD progression remains unknown. This study aimed to elucidate the contribution of CLEC5A to Aβ-induced microglial activation and behavioral deficits. METHODS The AD mouse model was crossed with Clec5a knockout mice for subsequent behavioral and pathological tests. The memory deficit was revealed by the Morris water maze, while the nociception abnormalities were examined by the von Frey filament and hotplate test. The Aβ deposition and microglia recruitment were identified by ELISA and immunohistochemistry. The inflammatory signals were identified by ELISA and western blotting. In the Clec5a knockdown microglial cell model and Clec5a knockout primary microglia, the microglial phagocytosis was revealed using the fluorescent-labeled Aβ. RESULTS The AD mice with Clec5a knockout improved Aβ-induced memory deficit and abnormal nociception. These mice have reduced Aβ deposition and increased microglia coverage surrounding the amyloid plaque, suggesting the involvement of CLEC5A in AD progression and Aβ clearance. Moreover, the phagocytosis was also increased in the Aβ-stressed Clec5a knockdown microglial cell lines and Clec5a knockout primary microglia. CONCLUSION The Clec5a knockout ameliorates AD-like deficits by modulating microglial Aβ clearance. This study implies that targeting microglial Clec5a could offer a promising approach to mitigate AD progression.
Collapse
MESH Headings
- Animals
- Lectins, C-Type/metabolism
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Microglia/metabolism
- Microglia/pathology
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Alzheimer Disease/genetics
- Mice
- Amyloid beta-Peptides/metabolism
- Disease Models, Animal
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Mice, Transgenic
- Maze Learning/physiology
- Phagocytosis
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
Collapse
Affiliation(s)
- Yu-Yi Lin
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Han Chang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.
- Institute of Clinical Medicine, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Irene Han-Juo Cheng
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
11
|
Młynarska E, Jakubowska P, Frąk W, Gajewska A, Sornowska J, Skwira S, Wasiak J, Rysz J, Franczyk B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients 2024; 16:3570. [PMID: 39458564 PMCID: PMC11510709 DOI: 10.3390/nu16203570] [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: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Recent research highlights the growing interest in the impact of nutrition on cognitive health and function in disease, as dietary habits are increasingly recognized as crucial factors in relation to brain function. This focus is especially important given the rising prevalence of neurodegenerative diseases and the cognitive decline associated with poor dietary choices. Links are now being sought between brain function and the microbiota and gut-brain axis. Mechanisms are proposed that include low-grade chronic neuroinflammation, the influence of short-chain fatty acids, or the disruption of glial cells and transmitters in the brain. METHODS We reviewed the articles on pubmed. This is not a systematic review, but of the narrative type. We wanted to outline the issue and summarise the latest information. RESULTS The axis in question has its foundation in nutrition. It has been reported that diet, particularly the components and the timing of food intake, has an impact on cognitive processes. The Mediterranean diet is most often cited in the literature as being beneficial to health. In order to obtain a more complete view, it is worth considering other dietary patterns, even those that impair our health. CONCLUSIONS Determining what is beneficial and what is not will allow us to develop a speronized strategy for the prevention of, and fight against, cognitive impairment. Appropriately selected supplements, the functions of which we have also discussed, may prove supportive.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Paulina Jakubowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Sornowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Sylwia Skwira
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
12
|
Mado H, Stasiniewicz A, Adamczyk-Sowa M, Sowa P. Selected Interleukins Relevant to Multiple Sclerosis: New Directions, Potential Targets and Therapeutic Perspectives. Int J Mol Sci 2024; 25:10931. [PMID: 39456713 PMCID: PMC11506881 DOI: 10.3390/ijms252010931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that progresses with demyelination and neurodegeneration. To date, many studies have revealed the key role of interleukins in the pathogenesis of MS, but their impact has not been fully explained. The aim of the present study was to collect and review the results obtained so far regarding the influence of interleukins on the development and course of MS and to assess the potential for their further use. Through the platform "PubMed", terms related to interleukins and MS were searched. The following interval was set as the time criterion: 2014-2024. A total of 12,731 articles were found, and 100 papers were subsequently used. Cells that produce IL-10 have a neuroprotective effect, whereas those that synthesize IL-6 most likely exacerbate neuroinflammation. IL-12, IL-23 and IL-18 represent pro-inflammatory cytokines. It was found that treatment with an anti-IL-12p40 monoclonal antibody in a study group of MS patients showed a beneficial effect. IL-4 is a pleiotropic cytokine that plays a significant role in type 2 immune responses and inhibits MS progression. IL-13 is an anti-inflammatory cytokine through which the processes of oligodendrogenesis and remyelination occur more efficiently. The group of interleukins discussed in our paper may represent a promising starting point for further research aimed at finding new therapies and prognostic markers for MS.
Collapse
Affiliation(s)
- Hubert Mado
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland
| | - Artur Stasiniewicz
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| |
Collapse
|
13
|
Yao H, Tong W, Song Y, Li R, Xiang X, Cheng W, Zhou Y, He Y, Yang Y, Liu Y, Li S, Jin L. Exercise training upregulates CD55 to suppress complement-mediated synaptic phagocytosis in Parkinson's disease. J Neuroinflammation 2024; 21:246. [PMID: 39342308 PMCID: PMC11439226 DOI: 10.1186/s12974-024-03234-0] [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: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
The primary pathological change in Parkinson's disease (PD) is the progressive degeneration of dopaminergic neurons in the substantia nigra. Additionally, excessive microglial activation and synaptic loss are also typical features observed in PD samples. Exercise trainings have been proven to improve PD symptoms, delay the disease progression as well as affect excessive microglial synaptic phagocytosis. In this study, we established a mouse model of PD by injecting mouse-derived α-synuclein preformed fibrils (M-α-syn PFFs) into the substantia nigra, and demonstrated that treadmill exercise inhibits microglial activation and synaptic phagocytosis in striatum. Using RNA-Seq and proteomics, we also found that PD involves excessive activation of the complement pathway which is closely related to over-activation of microglia and abnormal synaptic function. More importantly, exercise training can inhibit complement levels and complement-mediated microglial phagocytosis of synapses. It is probably triggered by CD55, as we observed that CD55 in the striatum significantly increased after exercise training and up-regulation of that molecule rescued motor deficits of PD mice, accompanied with reduced microglial synaptic phagocytosis in the striatum. This research elucidated the interplay among microglia, complement, and synapses, and analyzed the effects of exercise training on these factors. Our work also suggested CD55 as a complement-relevant candidate molecule for developing therapeutic strategies of PD.
Collapse
Affiliation(s)
- Hongkai Yao
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weifang Tong
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yunping Song
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Ruoyu Li
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Xuerui Xiang
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Wen Cheng
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yunjiao Zhou
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yijing He
- Neurotoxin Research Center, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Yang
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yunxi Liu
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Siguang Li
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Lingjing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Disabled Persons' Federation Key Laboratory of Intelligent Rehabilitation Assistive Devices and Technologies, Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
14
|
Dundee JM, Brown GC. The microglial P2Y 6 receptor as a therapeutic target for neurodegenerative diseases. Transl Neurodegener 2024; 13:47. [PMID: 39243044 PMCID: PMC11380353 DOI: 10.1186/s40035-024-00438-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
Neurodegenerative diseases are associated with chronic neuroinflammation in the brain, which can result in microglial phagocytosis of live synapses and neurons that may contribute to cognitive deficits and neuronal loss. The microglial P2Y6 receptor (P2Y6R) is a G-protein coupled receptor, which stimulates microglial phagocytosis when activated by extracellular uridine diphosphate, released by stressed neurons. Knockout or inhibition of P2Y6R can prevent neuronal loss in mouse models of Alzheimer's disease (AD), Parkinson's disease, epilepsy, neuroinflammation and aging, and prevent cognitive deficits in models of AD, epilepsy and aging. This review summarises the known roles of P2Y6R in the physiology and pathology of the brain, and its potential as a therapeutic target to prevent neurodegeneration and other brain pathologies.
Collapse
Affiliation(s)
- Jacob M Dundee
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
Lind-Holm Mogensen F, Sousa C, Ameli C, Badanjak K, Pereira SL, Muller A, Kaoma T, Coowar D, Scafidi A, Poovathingal SK, Tziortziou M, Antony PMA, Nicot N, Ginolhac A, Vogt Weisenhorn DM, Wurst W, Poli A, Nazarov PV, Skupin A, Grünewald A, Michelucci A. PARK7/DJ-1 deficiency impairs microglial activation in response to LPS-induced inflammation. J Neuroinflammation 2024; 21:174. [PMID: 39014482 PMCID: PMC11253405 DOI: 10.1186/s12974-024-03164-x] [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: 02/05/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Specific microglia responses are thought to contribute to the development and progression of neurodegenerative diseases, including Parkinson's disease (PD). However, the phenotypic acquisition of microglial cells and their role during the underlying neuroinflammatory processes remain largely elusive. Here, according to the multiple-hit hypothesis, which stipulates that PD etiology is determined by a combination of genetics and various environmental risk factors, we investigate microglial transcriptional programs and morphological adaptations under PARK7/DJ-1 deficiency, a genetic cause of PD, during lipopolysaccharide (LPS)-induced inflammation. METHODS Using a combination of single-cell RNA-sequencing, bulk RNA-sequencing, multicolor flow cytometry and immunofluorescence analyses, we comprehensively compared microglial cell phenotypic characteristics in PARK7/DJ-1 knock-out (KO) with wildtype littermate mice following 6- or 24-h intraperitoneal injection with LPS. For translational perspectives, we conducted corresponding analyses in human PARK7/DJ-1 mutant induced pluripotent stem cell (iPSC)-derived microglia and murine bone marrow-derived macrophages (BMDMs). RESULTS By excluding the contribution of other immune brain resident and peripheral cells, we show that microglia acutely isolated from PARK7/DJ-1 KO mice display a distinct phenotype, specially related to type II interferon and DNA damage response signaling, when compared with wildtype microglia, in response to LPS. We also detected discrete signatures in human PARK7/DJ-1 mutant iPSC-derived microglia and BMDMs from PARK7/DJ-1 KO mice. These specific transcriptional signatures were reflected at the morphological level, with microglia in LPS-treated PARK7/DJ-1 KO mice showing a less amoeboid cell shape compared to wildtype mice, both at 6 and 24 h after acute inflammation, as also observed in BMDMs. CONCLUSIONS Taken together, our results show that, under inflammatory conditions, PARK7/DJ-1 deficiency skews microglia towards a distinct phenotype characterized by downregulation of genes involved in type II interferon signaling and a less prominent amoeboid morphology compared to wildtype microglia. These findings suggest that the underlying oxidative stress associated with the lack of PARK7/DJ-1 affects microglia neuroinflammatory responses, which may play a causative role in PD onset and progression.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Carole Sousa
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- International Iberian Nanotechnology Laboratory, 4715-330, Braga, Portugal
| | - Corrado Ameli
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Katja Badanjak
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Sandro L Pereira
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Arnaud Muller
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- LuxGen Genome Center, Luxembourg Institute of Health and Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Djalil Coowar
- Rodent Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Andrea Scafidi
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Suresh K Poovathingal
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Single Cell Analytics and Microfluidics Core, Vlaams Instituut Voor Biotechnologie-KU Leuven, 3000, Louvain, Belgium
| | - Maria Tziortziou
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Paul M A Antony
- Bioimaging Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Nathalie Nicot
- LuxGen Genome Center, Luxembourg Institute of Health and Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Daniela M Vogt Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Technische Universität München-Weihenstephan, 85354, Freising, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Technische Universität München-Weihenstephan, 85354, Freising, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
- Deutsche Zentrum für Psychische Gesundheit (DZPG), 80336, Munich, Germany
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Petr V Nazarov
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Multiomics Data Science Group, Department of Cancer Research, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Alexander Skupin
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Neuroscience, University of California San Diego, La Jolla, CA, 92093, USA
- Integrative Biophysics, Department of Physics and Material Science, University of Luxembourg, L-1511, Luxembourg, Luxembourg
| | - Anne Grünewald
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg.
| |
Collapse
|
16
|
Chmielarz M, Sobieszczańska B, Środa-Pomianek K. Metabolic Endotoxemia: From the Gut to Neurodegeneration. Int J Mol Sci 2024; 25:7006. [PMID: 39000116 PMCID: PMC11241432 DOI: 10.3390/ijms25137006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Metabolic endotoxemia is a severe health problem for residents in developed countries who follow a Western diet, disrupting intestinal microbiota and the whole organism's homeostasis. Although the effect of endotoxin on the human immune system is well known, its long-term impact on the human body, lasting many months or even years, is unknown. This is due to the difficulty of conducting in vitro and in vivo studies on the prolonged effect of endotoxin on the central nervous system. In this article, based on the available literature, we traced the path of endotoxin from the intestines to the blood through the intestinal epithelium and factors promoting the development of metabolic endotoxemia. The presence of endotoxin in the bloodstream and the inflammation it induces may contribute to lowering the blood-brain barrier, potentially allowing its penetration into the central nervous system; although, the theory is still controversial. Microglia, guarding the central nervous system, are the first line of defense and respond to endotoxin with activation, which may contribute to the development of neurodegenerative diseases. We traced the pro-inflammatory role of endotoxin in neurodegenerative diseases and its impact on the epigenetic regulation of microglial phenotypes.
Collapse
Affiliation(s)
- Mateusz Chmielarz
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Beata Sobieszczańska
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Wroclaw University of Medicine, Chalubinskiego 3a, 50-368 Wroclaw, Poland
| |
Collapse
|
17
|
Ryabushkina YA, Ayriyants KA, Sapronova AA, Mutovina AS, Kolesnikova MM, Mezhlumyan EV, Bondar NP, Reshetnikov VV. Effects of different types of induced neonatal inflammation on development and behavior of C57BL/6 and BTBR mice. Physiol Behav 2024; 280:114550. [PMID: 38614416 DOI: 10.1016/j.physbeh.2024.114550] [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: 09/05/2023] [Revised: 03/21/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024]
Abstract
Neuroinflammation in the early postnatal period can disturb trajectories of the completion of normal brain development and can lead to mental illnesses, such as depression, anxiety disorders, and personality disorders later in life. In our study, we focused on evaluating short- and long-term effects of neonatal inflammation induced by lipopolysaccharide, poly(I:C), or their combination in female and male C57BL/6 and BTBR mice. We chose the BTBR strain as potentially more susceptible to neonatal inflammation because these mice have behavioral, neuroanatomical, and physiological features of autism spectrum disorders, an abnormal immune response, and several structural aberrations in the brain. Our results indicated that BTBR mice are more sensitive to the influence of the neonatal immune activation (NIA) on the formation of neonatal reflexes than C57BL/6 mice are. In these experiments, the injection of lipopolysaccharide had an effect on the formation of the cliff aversion reflex in female BTBR mice. Nonetheless, NIA had no delayed effects on either social behavior or anxiety-like behavior in juvenile and adolescent BTBR and C57BL/6 mice. Altogether, our data show that NIA has mimetic-, age-, and strain-dependent effects on the development of neonatal reflexes and on exploratory activity in BTBR and C57BL/6 mice.
Collapse
Affiliation(s)
- Yuliya A Ryabushkina
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia
| | - Kseniya A Ayriyants
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia
| | - Anna A Sapronova
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia
| | - Anastasia S Mutovina
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia; Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia
| | - Maria M Kolesnikova
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia; Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia
| | - Eva V Mezhlumyan
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia; Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia
| | - Natalya P Bondar
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia; Novosibirsk State University, Pirogova Street 2, Novosibirsk 630090, Russia.
| | - Vasiliy V Reshetnikov
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), Prospekt Akad. Lavrentyeva 10, Novosibirsk 630090, Russia; Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Avenue, Sochi 354340, Russia.
| |
Collapse
|
18
|
Liu X, Yang W, Zhu C, Sun S, Yang B, Wu S, Wang L, Liu Z, Ge Z. TLR2 Mediates Microglial Activation and Contributes to Central Sensitization in a Recurrent Nitroglycerin-induced Chronic Migraine Model. Mol Neurobiol 2024; 61:3697-3714. [PMID: 38008889 DOI: 10.1007/s12035-023-03781-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/06/2023] [Indexed: 11/28/2023]
Abstract
Central sensitization is an important pathophysiological mechanism underlying chronic migraine (CM). Previous studies have shown that microglial activation and subsequent inflammation in the trigeminal nucleus caudalis (TNC) contribute to central sensitization. Toll-like receptor 2 (TLR2) is a receptor expressed on the membrane of microglia and participates in central sensitization in inflammatory and chronic pain; however, its role in CM is unclear. Therefore, this study investigated TLR2 involvement in CM in detail. Mice treated with recurrent nitroglycerin (NTG) were used as a CM model. Hyperalgesia was assessed using a 50% paw mechanical threshold and a 50% periorbital threshold on a Von Frey filament pain meter. Western blotting and immunofluorescence analyses were used to detect the expression of TLR2, microglia, c-fos and CGRP in TNC. The expression of inflammatory factors (IL-6, IL-1β、 IL-10、TNF-α and IFN-β1) was detected using quantitative real-time polymerase chain reaction (qRT-PCR). A selective TLR2 antagonist (C29) was systematically administered to observe its effect on hyperalgesia, microglia activation and the expression of c-fos, CGRP and inflammatory factors. Recurrent administration of NTG resulted in acute and chronic hypersensitivity, accompanied by upregulation of TLR2 expression and microglial activation in TNC. C29 partially inhibited pain hypersensitivity. C29 suppressed microglial activation induced by NTG administration. Inhibition of TLR2 reduced the expression of c-fos and CGRP in TNC after NTG treatment. C29 inhibited the expression of inflammatory mediators in TNC. These data showed that microglial TLR2 plays a critical role in the pathogenesis of CM by regulating microglial activation in TNC.
Collapse
Affiliation(s)
- Xuejiao Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Wenping Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chenlu Zhu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Songtang Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Bin Yang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Shouyi Wu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Longde Wang
- Expert Workstation of Academician Wang Longde, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Zhiyan Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| | - Zhaoming Ge
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
19
|
Kong W, Frouard J, Xie G, Corley MJ, Helmy E, Zhang G, Schwarzer R, Montano M, Sohn P, Roan NR, Ndhlovu LC, Gan L, Greene WC. Neuroinflammation generated by HIV-infected microglia promotes dysfunction and death of neurons in human brain organoids. PNAS NEXUS 2024; 3:pgae179. [PMID: 38737767 PMCID: PMC11086946 DOI: 10.1093/pnasnexus/pgae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024]
Abstract
Despite the success of combination antiretroviral therapy (ART) for individuals living with HIV, mild forms of HIV-associated neurocognitive disorder (HAND) continue to occur. Brain microglia form the principal target for HIV infection in the brain. It remains unknown how infection of these cells leads to neuroinflammation, neuronal dysfunction, and/or death observed in HAND. Utilizing two different inducible pluripotent stem cell-derived brain organoid models (cerebral and choroid plexus [ChP] organoids) containing microglia, we investigated the pathogenic changes associated with HIV infection. Infection of microglia was associated with a sharp increase in CCL2 and CXCL10 chemokine gene expression and the activation of many type I interferon stimulated genes (MX1, ISG15, ISG20, IFI27, IFITM3 and others). Production of the proinflammatory chemokines persisted at low levels after treatment of the cell cultures with ART, consistent with the persistence of mild HAND following clinical introduction of ART. Expression of multiple members of the S100 family of inflammatory genes sharply increased following HIV infection of microglia measured by single-cell RNA-seq. However, S100 gene expression was not limited to microglia but was also detected more broadly in uninfected stromal cells, mature and immature ChP cells, neural progenitor cells and importantly in bystander neurons suggesting propagation of the inflammatory response to bystander cells. Neurotransmitter transporter expression declined in uninfected neurons, accompanied by increased expression of genes promoting cellular senescence and cell death. Together, these studies underscore how an inflammatory response generated in HIV-infected microglia is propagated to multiple uninfected bystander cells ultimately resulting in the dysfunction and death of bystander neurons.
Collapse
Affiliation(s)
- Weili Kong
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Julie Frouard
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Guorui Xie
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael J Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ekram Helmy
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Gang Zhang
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Roland Schwarzer
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Mauricio Montano
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
| | - Peter Sohn
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Nadia R Roan
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Li Gan
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Warner C Greene
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA 94158, USA
- Gladstone Institute of Virology, San Francisco, CA 94158, USA
- Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
20
|
Brown GC, Heneka MT. The endotoxin hypothesis of Alzheimer's disease. Mol Neurodegener 2024; 19:30. [PMID: 38561809 PMCID: PMC10983749 DOI: 10.1186/s13024-024-00722-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Lipopolysaccharide (LPS) constitutes much of the surface of Gram-negative bacteria, and if LPS enters the human body or brain can induce inflammation and act as an endotoxin. We outline the hypothesis here that LPS may contribute to the pathophysiology of Alzheimer's disease (AD) via peripheral infections or gut dysfunction elevating LPS levels in blood and brain, which promotes: amyloid pathology, tau pathology and microglial activation, contributing to the neurodegeneration of AD. The evidence supporting this hypothesis includes: i) blood and brain levels of LPS are elevated in AD patients, ii) AD risk factors increase LPS levels or response, iii) LPS induces Aβ expression, aggregation, inflammation and neurotoxicity, iv) LPS induces TAU phosphorylation, aggregation and spreading, v) LPS induces microglial priming, activation and neurotoxicity, and vi) blood LPS induces loss of synapses, neurons and memory in AD mouse models, and cognitive dysfunction in humans. However, to test the hypothesis, it is necessary to test whether reducing blood LPS reduces AD risk or progression. If the LPS endotoxin hypothesis is correct, then treatments might include: reducing infections, changing gut microbiome, reducing leaky gut, decreasing blood LPS, or blocking LPS response.
Collapse
Affiliation(s)
- Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
21
|
Kou L, Chi X, Sun Y, Yin S, Wu J, Zou W, Wang Y, Jin Z, Huang J, Xiong N, Xia Y, Wang T. Circadian regulation of microglia function: Potential targets for treatment of Parkinson's Disease. Ageing Res Rev 2024; 95:102232. [PMID: 38364915 DOI: 10.1016/j.arr.2024.102232] [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: 07/17/2023] [Revised: 02/11/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Circadian rhythms are involved in the regulation of many aspects of the body, including cell function, physical activity and disease. Circadian disturbance often predates the typical symptoms of neurodegenerative diseases and is not only a non-motor symptom, but also one of the causes of their occurrence and progression. Glial cells possess circadian clocks that regulate their function to maintain brain development and homeostasis. Emerging evidence suggests that the microglial circadian clock is involved in the regulation of many physiological processes, such as cytokine release, phagocytosis, and nutritional and metabolic support, and that disruption of the microglia clock may affect multiple aspects of Parkinson's disease, especially neuroinflammation and α-synuclein processes. Herein, we review recent advances in the circadian control of microglia function in health and disease, and discuss novel pharmacological interventions for microglial clocks in neurodegenerative disorders.
Collapse
Affiliation(s)
- Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
22
|
Hao W, Ma Q, Wang L, Yuan N, Gan H, He L, Li X, Huang J, Chen J. Gut dysbiosis induces the development of depression-like behavior through abnormal synapse pruning in microglia-mediated by complement C3. MICROBIOME 2024; 12:34. [PMID: 38378622 PMCID: PMC10877840 DOI: 10.1186/s40168-024-01756-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/04/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Remodeling eubiosis of the gut microenvironment may contribute to preventing the occurrence and development of depression. Mounting experimental evidence has shown that complement C3 signaling is associated with the pathogenesis of depression, and disruption of the gut microbiota may be an underlying cause of complement system activation. However, the mechanism by which complement C3 participates in gut-brain crosstalk in the pathogenesis of depression remains unknown. RESULTS In the present study, we found that chronic unpredictable mild stress (CUMS)-induced mice exhibited obvious depression-like behavior as well as cognitive impairment, which was associated with significant gut dysbiosis, especially enrichment of Proteobacteria and elevation of microbiota-derived lipopolysaccharides (LPS). In addition, peripheral and central complement C3 activation and central C3/CR3-mediated aberrant synaptic pruning in microglia have also been observed. Transplantation of gut microbiota from CUMS-induced depression model mice into specific pathogen-free and germ-free mice induced depression-like behavior and concomitant cognitive impairment in the recipient mice, accompanied by increased activation of the complement C3/CR3 pathway in the prefrontal cortex and abnormalities in microglia-mediated synaptic pruning. Conversely, antidepressants and fecal microbiota transplantation from antidepressant-treated donors improved depression-like behaviors and restored gut microbiome disturbances in depressed mice. Concurrently, inhibition of the complement C3/CR3 pathway, amelioration of abnormal microglia-mediated synaptic pruning, and increased expression of the synapsin and postsynaptic density protein 95 were observed. Collectively, our results revealed that gut dysbiosis induces the development of depression-like behaviors through abnormal synapse pruning in microglia-mediated by complement C3, and the inhibition of abnormal synaptic pruning is the key to targeting microbes to treat depression. CONCLUSIONS Our findings provide novel insights into the involvement of complement C3/CR3 signaling and aberrant synaptic pruning of chemotactic microglia in gut-brain crosstalk in the pathogenesis of depression. Video Abstract.
Collapse
Affiliation(s)
- Wenzhi Hao
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lu Wang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Hua Gan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Liangliang He
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| | - Junqing Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
23
|
Liu H, Liu X, Luo S, Ma R, Ge W, Meng S, Gao Y. Lamin A/C mediates microglial activation by modulating cell proliferation and immune response. J Neurosci Res 2024; 102:e25263. [PMID: 38284866 DOI: 10.1002/jnr.25263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/01/2023] [Indexed: 01/30/2024]
Abstract
Lamin A/C is involved in macrophage activation and premature aging, also known as progeria. As the resident macrophage in brain, overactivation of microglia causes brain inflammation, promoting aging and brain disease. In this study, we investigated the role of Lamin A/C in microglial activation and its impact on progeria using Lmna-/- mice, primary microglia, Lmna knockout (Lmna-KO) and Lmna-knockdown (Lmna-KD) BV2 cell lines. We found that the microglial activation signatures, including cell proliferation, morphology changes, and proinflammatory cytokine secretion (IL-1β, IL-6, and TNF-α), were significantly suppressed in all Lamin A/C-deficient models when stimulated with LPS. TMT-based quantitative proteomic and bioinformatic analysis were further applied to explore the mechanism of Lamin A/C-regulated microglia activation from the proteome level. The results revealed that immune response and phagocytosis were impaired in Lmna-/- microglia. Stat1 was identified as the hub protein in the mechanism by which Lamin A/C regulates microglial activation. Additionally, DNA replication, chromatin organization, and mRNA processing were also altered by Lamin A/C, with Ki67 fulfilling the main hub function. Lamin A/C is a mechanosensitive protein and, the immune- and proliferation-related biological processes are also regulated by mechanotransduction. We speculate that Lamin A/C-mediated mechanotransduction is required for microglial activation. Our study proposes a novel mechanism for microglial activation mediated by Lamin A/C.
Collapse
Affiliation(s)
- Haotian Liu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xinnan Liu
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shiqi Luo
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Rayna Ma
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wei Ge
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shu Meng
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yanpan Gao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Xu J, Huang P, Bie B, Dai Y, Ben-Salem S, Borjini N, Zhang L, Chen J, Olman M, Cheng J, Lin F. Complement Receptor C3aR1 Contributes to Paclitaxel-Induced Peripheral Neuropathic Pain in Mice and Rats. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1736-1746. [PMID: 37861348 PMCID: PMC10841827 DOI: 10.4049/jimmunol.2300252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023]
Abstract
Cancer chemotherapy-induced neuropathic pain is a devastating pain syndrome without effective therapies. We previously reported that rats deficient in complement C3, the central component of complement activation cascade, showed a reduced degree of paclitaxel-induced mechanical allodynia (PIMA), suggesting that complement is integrally involved in the pathogenesis of this model. However, the underlying mechanism was unclear. Complement activation leads to the production of C3a, which mediates inflammation through its receptor C3aR1. In this article, we report that the administration of paclitaxel induced a significantly higher expression level of C3aR1 on dorsal root ganglion (DRG) macrophages and expansion of these macrophages in DRGs in wild-type (WT) compared with in C3aR1 knockout (KO) mice. We also found that paclitaxel induced less severe PIMA, along with a reduced DRG expression of transient receptor potential channels of the vanilloid subtype 4 (TRPV4), an essential mediator for PIMA, in C3aR1 KO than in WT mice. Treating WT mice or rats with a C3aR1 antagonist markedly attenuated PIMA in association with downregulated DRG TRPV4 expression, reduced DRG macrophages expansion, suppressed DRG neuron hyperexcitability, and alleviated peripheral intraepidermal nerve fiber loss. Administration of C3aR1 antagonist to TRPV4 KO mice further protected them from PIMA. These results suggest that complement regulates PIMA development through C3aR1 to upregulate TRPV4 on DRG neurons and promote DRG macrophage expansion. Targeting C3aR1 could be a novel therapeutic approach to alleviate this debilitating pain syndrome.
Collapse
Affiliation(s)
- Jijun Xu
- Department of Pain Management, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ping Huang
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Bihua Bie
- Department of Pain Management, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Yang Dai
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Salma Ben-Salem
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Nozha Borjini
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lingjun Zhang
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jin Chen
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mitchell Olman
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jianguo Cheng
- Department of Pain Management, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
- Department of Neurosciences, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| | - Feng Lin
- Department of Inflammation and Immunity, 9500 Euclid Ave., Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
25
|
Santana-Coelho D, Lugo JN. Hippocampal Upregulation of Complement Component C3 in Response to Lipopolysaccharide Stimuli in a Model of Fragile-X Syndrome. Curr Issues Mol Biol 2023; 45:9306-9315. [PMID: 37998759 PMCID: PMC10669955 DOI: 10.3390/cimb45110582] [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: 10/23/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023] Open
Abstract
The complement system is part of the innate immune system and has been shown to be altered in autism spectrum disorder (ASD). Fragile-X syndrome (FXS) is the main genetic cause of ASD and studies suggest a dysregulation in the immune system in patients with the disorder. To assess if an animal model of FXS presents with altered complement signaling, we treated male Fmr1 knockout (KO) mice with lipopolysaccharide (LPS) and collected the hippocampus 24 h later. Assessment of the expression of the complement genes C1q, C3, and C4 identified the upregulation of C3 in both wild-type (WT) and knockout mice. Levels of C3 also increased in both genotypes. Analysis of the correlation between the expression of C3 and the cytokines IL-6, IL-1β, and TNF-α identified a different relationship between the expression of the genes in Fmr1 KO when compared to WT mice. Our findings did not support our initial hypotheses that the lack of the FMR1 gene would alter complement system signaling, and that the induction of the complement system in response to LPS in Fmr1 KO mice differed from wild-type conspecifics.
Collapse
Affiliation(s)
| | - Joaquin N. Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, TX 76798, USA;
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA
- Department of Biology, Baylor University, Waco, TX 76798, USA
| |
Collapse
|
26
|
Yeo S, Jang J, Jung HJ, Lee H, Choe Y. Primary cilia-mediated regulation of microglial secretion in Alzheimer's disease. Front Mol Biosci 2023; 10:1250335. [PMID: 37942288 PMCID: PMC10627801 DOI: 10.3389/fmolb.2023.1250335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023] Open
Abstract
Alzheimer's disease (AD) is a brain disorder manifested by a gradual decline in cognitive function due to the accumulation of extracellular amyloid plaques, disruptions in neuronal substance transport, and the degeneration of neurons. In affected neurons, incomplete clearance of toxic proteins by neighboring microglia leads to irreversible brain inflammation, for which cellular signaling is poorly understood. Through single-cell transcriptomic analysis, we discovered distinct regional differences in the ability of microglia to clear damaged neurites. Specifically, microglia in the septal region of wild type mice exhibited a transcriptomic signature resembling disease-associated microglia (DAM). These lateral septum (LS)-enriched microglia were associated with dense axonal bundles originating from the hippocampus. Further transcriptomic and proteomic approaches revealed that primary cilia, small hair-like structures found on cells, played a role in the regulation of microglial secretory function. Notably, primary cilia were transiently observed in microglia, and their presence was significantly reduced in microglia from AD mice. We observed significant changes in the secretion and proteomic profiles of the secretome after inhibiting the primary cilia gene intraflagellar transport particle 88 (Ift88) in microglia. Intriguingly, inhibiting primary cilia in the septal microglia of AD mice resulted in the expansion of extracellular amyloid plaques and damage to adjacent neurites. These results indicate that DAM-like microglia are present in the LS, a critical target region for hippocampal nerve bundles, and that the primary ciliary signaling system regulates microglial secretion, affecting extracellular proteostasis. Age-related primary ciliopathy probably contributes to the selective sensitivity of microglia, thereby exacerbating AD. Targeting the primary ciliary signaling system could therefore be a viable strategy for modulating neuroimmune responses in AD treatments.
Collapse
Affiliation(s)
- Seungeun Yeo
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jaemyung Jang
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyeyoung Lee
- Division of Applied Bioengineering, Dong-eui University, Busan, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
27
|
Yang Y, García-Cruzado M, Zeng H, Camprubí-Ferrer L, Bahatyrevich-Kharitonik B, Bachiller S, Deierborg T. LPS priming before plaque deposition impedes microglial activation and restrains Aβ pathology in the 5xFAD mouse model of Alzheimer's disease. Brain Behav Immun 2023; 113:228-247. [PMID: 37437821 DOI: 10.1016/j.bbi.2023.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023] Open
Abstract
Microglia have an innate immunity memory (IIM) with divergent functions in different animal models of neurodegenerative diseases, including Alzheimer's disease (AD). AD is characterized by chronic neuroinflammation, neurodegeneration, tau tangles and β-amyloid (Aβ) deposition. Systemic inflammation has been implicated in contributing to the progression of AD. Multiple reports have demonstrated unique microglial signatures in AD mouse models and patients. However, the proteomic profiles of microglia modified by IIM have not been well-documented in an AD model. Therefore, in the present study, we investigate whether lipopolysaccharide (LPS)-induced IIM in the pre-clinical stage of AD alters the microglial responses and shapes the neuropathology. We accomplished this by priming 5xFAD and wild-type (WT) mice with an LPS injection at 6 weeks (before the robust development of plaques). 140 days later, we evaluated microglial morphology, activation, the microglial barrier around Aβ, and Aβ deposition in both 5xFAD primed and unprimed mice. Priming induced decreased soma size of microglia and reduced colocalization of PSD95 and Synaptophysin in the retrosplenial cortex. Priming appeared to increase phagocytosis of Aβ, resulting in fewer Thioflavin S+ Aβ fibrils in the dentate gyrus. RIPA-soluble Aβ 40 and 42 were significantly reduced in Primed-5xFAD mice leading to a smaller size of MOAB2+ Aβ plaques in the prefrontal cortex. We also found that Aβ-associated microglia in the Primed-5xFAD mice were less activated and fewer in number. After priming, we also observed improved memory performance in 5xFAD. To further elucidate the molecular mechanism underlying these changes, we performed quantitative proteomic analysis of microglia and bone marrow monocytes. A specific pattern in the microglial proteome was revealed in primed 5xFAD mice. These results suggest that the imprint signatures of primed microglia display a distinctive phenotype and highlight the potential for a beneficial adaption of microglia when intervention occurs in the pre-clinical stage of AD.
Collapse
Affiliation(s)
- Yiyi Yang
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden.
| | - Marta García-Cruzado
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden
| | - Hairuo Zeng
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden
| | - Lluís Camprubí-Ferrer
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden
| | - Bazhena Bahatyrevich-Kharitonik
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden; Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, CSIC, Spain; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Sara Bachiller
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden; Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, CSIC, Spain; Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Sweden.
| |
Collapse
|
28
|
Abstract
Because the central nervous system is largely nonrenewing, neurons and their synapses must be maintained over the lifetime of an individual to ensure circuit function. Age is a dominant risk factor for neural diseases, and declines in nervous system function are a common feature of aging even in the absence of disease. These alterations extend to the visual system and, in particular, to the retina. The retina is a site of clinically relevant age-related alterations but has also proven to be a uniquely approachable system for discovering principles that govern neural aging because it is well mapped, contains diverse neuron types, and is experimentally accessible. In this article, we review the structural and molecular impacts of aging on neurons within the inner and outer retina circuits. We further discuss the contribution of non-neuronal cell types and systems to retinal aging outcomes. Understanding how and why the retina ages is critical to efforts aimed at preventing age-related neural decline and restoring neural function.
Collapse
Affiliation(s)
- Jeffrey D Zhu
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Sharma Pooja Tarachand
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Qudrat Abdulwahab
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA;
| |
Collapse
|
29
|
Zhou R, Chen SH, Zhao Z, Tu D, Song S, Wang Y, Wang Q, Feng J, Hong JS. Complement C3 Enhances LPS-Elicited Neuroinflammation and Neurodegeneration Via the Mac1/NOX2 Pathway. Mol Neurobiol 2023; 60:5167-5183. [PMID: 37268807 PMCID: PMC10415527 DOI: 10.1007/s12035-023-03393-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/18/2023] [Indexed: 06/04/2023]
Abstract
Recent studies showed increased expression of complements in various neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. However, the mechanism regulating the expression of complements and their roles in the pathogenesis of neurodegeneration are unclear. We hypothesized that acute neuroinflammation increases the expression and activation of brain complements, which, in turn, participate in chronic neuroinflammation and progressive neurodegeneration. We initially focused on the complement component C3, because C3 can activate microglia by binding to C3 receptors and attaching to damaged neurons destined to be phagocytosed by microglia. We found that complement C3 is upregulated in lipopolysaccharide (LPS)-stimulated neuron/glial cultures. Mechanistic studies revealed that microglia-released proinflammatory factors initiated the enhanced expression of C3 in astroglia during acute neuroinflammation. On the other hand, the sustained C3 expression during chronic neuroinflammation requires releasing damage-associated molecule patterns (DAMPs) from damaged/degenerating brain cells. Our results suggested that DAMPs might act on microglial integrin receptor Mac1 to trigger the activation of NADPH oxidase (NOX2). Activated microglial NOX2 increases the production of extracellular reactive oxygen species (ROS), elevating the levels of intracellular ROS of astroglia and sustaining the astroglial C3 expression. This was supported by the findings showing reduced C3 expression and attenuated neurodegeneration in LPS-treated neuron/glial cultures prepared from mice deficient in Mac1 or NOX2. LPS-induced neurodegeneration and oxidative stress are significantly reduced in C3 KO neuron/glial cultures and mouse brains. Together, this study provides the first evidence demonstrating the role of C3 in regulating chronic neuroinflammation and in driving progressive neurodegeneration.
Collapse
Affiliation(s)
- Ran Zhou
- Respiratory Department, The First People's Hospital of Yunnan Provience, Kunming, 650032, People's Republic of China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
| | - Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA.
| | - Zhan Zhao
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Dezhen Tu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
| | - Sheng Song
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yubao Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Qingshan Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
- School of Public Health, Dalian Medical University, Dalian, Liaoning Province, China
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Dr., Research Triangle Park, Durham, NC, 27709, USA
| |
Collapse
|
30
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
31
|
Brown GC, Camacho M, Williams‐Gray CH. The Endotoxin Hypothesis of Parkinson's Disease. Mov Disord 2023; 38:1143-1155. [PMID: 37157885 PMCID: PMC10947365 DOI: 10.1002/mds.29432] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
The endotoxin hypothesis of Parkinson's disease (PD) is the idea that lipopolysaccharide (LPS) endotoxins contribute to the pathogenesis of this disorder. LPS endotoxins are found in, and released from, the outer membrane of Gram-negative bacteria, for example in the gut. It is proposed that gut dysfunction in early PD leads to elevated LPS levels in the gut wall and blood, which promotes both α-synuclein aggregation in the enteric neurons and a peripheral inflammatory response. Communication to the brain via circulating LPS and cytokines in the blood and/or the gut-brain axis leads to neuroinflammation and spreading of α-synuclein pathology, exacerbating neurodegeneration in brainstem nuclei and loss of dopaminergic neurons in the substantia nigra, and manifesting in the clinical symptoms of PD. The evidence supporting this hypothesis includes: (1) gut dysfunction, permeability, and bacterial changes occur early in PD, (2) serum levels of LPS are increased in a proportion of PD patients, (3) LPS induces α-synuclein expression, aggregation, and neurotoxicity, (4) LPS causes activation of peripheral monocytes leading to inflammatory cytokine production, and (5) blood LPS causes brain inflammation and specific loss of midbrain dopaminergic neurons, mediated by microglia. If the hypothesis is correct, then treatment options might include: (1) changing the gut microbiome, (2) reducing gut permeability, (3) reducing circulating LPS levels, or (4) blocking the response of immune cells and microglia to LPS. However, the hypothesis has a number of limitations and requires further testing, in particular whether reducing LPS levels can reduce PD incidence, progression, or severity. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Guy C. Brown
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Marta Camacho
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
32
|
Chung HY, Wickel J, Hahn N, Mein N, Schwarzbrunn M, Koch P, Ceanga M, Haselmann H, Baade-Büttner C, von Stackelberg N, Hempel N, Schmidl L, Groth M, Andreas N, Götze J, Coldewey SM, Bauer M, Mawrin C, Dargvainiene J, Leypoldt F, Steinke S, Wang ZQ, Hust M, Geis C. Microglia mediate neurocognitive deficits by eliminating C1q-tagged synapses in sepsis-associated encephalopathy. SCIENCE ADVANCES 2023; 9:eabq7806. [PMID: 37235660 DOI: 10.1126/sciadv.abq7806] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 04/20/2023] [Indexed: 05/28/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is a severe and frequent complication of sepsis causing delirium, coma, and long-term cognitive dysfunction. We identified microglia and C1q complement activation in hippocampal autopsy tissue of patients with sepsis and increased C1q-mediated synaptic pruning in a murine polymicrobial sepsis model. Unbiased transcriptomics of hippocampal tissue and isolated microglia derived from septic mice revealed an involvement of the innate immune system, complement activation, and up-regulation of lysosomal pathways during SAE in parallel to neuronal and synaptic damage. Microglial engulfment of C1q-tagged synapses could be prevented by stereotactic intrahippocampal injection of a specific C1q-blocking antibody. Pharmacologically targeting microglia by PLX5622, a CSF1-R inhibitor, reduced C1q levels and the number of C1q-tagged synapses, protected from neuronal damage and synapse loss, and improved neurocognitive outcome. Thus, we identified complement-dependent synaptic pruning by microglia as a crucial pathomechanism for the development of neuronal defects during SAE.
Collapse
Affiliation(s)
- Ha-Yeun Chung
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| | - Jonathan Wickel
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| | - Nina Hahn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| | - Nils Mein
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Meike Schwarzbrunn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena 07745, Germany
| | - Mihai Ceanga
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Holger Haselmann
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Carolin Baade-Büttner
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Nikolai von Stackelberg
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Nina Hempel
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Lars Schmidl
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena 07745, Germany
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital, Jena 07743, Germany
| | - Juliane Götze
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
- Septomics Research Center, Jena University Hospital, Jena 07745, Germany
| | - Sina M Coldewey
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
- Septomics Research Center, Jena University Hospital, Jena 07745, Germany
| | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena 07747, Germany
| | - Christian Mawrin
- Department of Neuropathology, University of Magdeburg, Magdeburg, Germany
- Section of Pathology, Institute of Forensic Medicine, Jena University Hospital, Jena 07749, Germany
| | | | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, UKSH, Kiel/Lübeck, Germany
- Department of Neurology, Christian-Albrechts University, Kiel 24105, Germany
| | - Stephan Steinke
- Department Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena 07745, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena 07745, Germany
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Michael Hust
- Department Medical Biotechnology, Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena 07747, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena 07747, Germany
| |
Collapse
|
33
|
Gretenkort L, Thiesler H, Hildebrandt H. Neuroimmunomodulatory properties of polysialic acid. Glycoconj J 2023; 40:277-294. [PMID: 37171513 DOI: 10.1007/s10719-023-10120-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/16/2022] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Polymeric sialic acid (polysialic acid, polySia) is a remarkable posttranslational modification of only few select proteins. The major, and most prominent polySia protein carrier is the neural cell adhesion molecule NCAM. Here, the key functions of polySia are to regulate interactions of NCAM and to balance cellular interactions in brain development and plasticity. During recent years, however, increasing evidence points towards a role of polySia in the modulation of immune responses. These immunomodulatory functions can be mediated by polySia on proteins other than NCAM, presented either on the cell surface or released into the extracellular space. This perspective review summarizes our current knowledge and addresses major open questions on polySia and polySia receptors in modulating innate immune responses in the brain.
Collapse
Affiliation(s)
- Lina Gretenkort
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
34
|
Wasiak J, Gawlik-Kotelnicka O. Intestinal permeability and its significance in psychiatric disorders - a narrative review and future perspectives. Behav Brain Res 2023; 448:114459. [PMID: 37121278 DOI: 10.1016/j.bbr.2023.114459] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/02/2023]
Abstract
The topic of increased intestinal permeability and its impact on the human body is. increasingly being addressed by researchers. It is associated with disruption of the. intestinal barrier, leading to the "leaky gut" syndrome. This can be assessed by. classical methods, determining the concentration of orally administered tracer. molecules in urine or by using biomarkers such as LPS, LBP or zonulin in blood. plasma. The presence of bacterial endotoxins in the body causes inflammation. In this. article, we review research on increased intestinal permeability in psychiatric illness. mood disorders, schizophrenia, alcohol dependence, anxiety disorders,. neurodegenerative and neurodevelopmental disorders. The results of the studies used. to assess intestinal permeability in different disease entities are presented. Possible. mechanisms for these interactions are the effects of chronic, low-grade inflammation. on the human brain, causing interruption of the brain blood barrier and dysfunction of. astrocytes and microglia. This affects brain function by reducing the number of. dopaminergic neurons, disrupting tryptophan metabolism and altering the amount of. GABA and glutamate. The links and mechanisms found may, in the future, allow earlier. detection of diseases and their targeted treatment.
Collapse
Affiliation(s)
- Jakub Wasiak
- Medical University of Lodz, Kosciuszki 4, 90-419 Lodz, Poland
| | - Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland.
| |
Collapse
|
35
|
Ghosh D, Singh G, Mishra P, Singh A, Kumar A, Sinha N. Alteration in mitochondrial dynamics promotes the proinflammatory response of microglia and is involved in cerebellar dysfunction of young and aged mice following LPS exposure. Neurosci Lett 2023; 807:137262. [PMID: 37116576 DOI: 10.1016/j.neulet.2023.137262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023]
Abstract
Cerebellar dysfunction is implicated in impaired motor coordination and balance, thus disturbing the dynamics of sensorimotor integration. Neuroinflammation and aging could be prominent contributors to cerebellar aberration. Additionally, changes in mitochondrial dynamics may precede microglia activation in several chronic neurodegenerative diseases; however, the underlying mechanism remains largely unknown.Here using LPS (1 mg/kg i.p. for four consecutive days) stimulation in both young (3 months old) and aged (12 months old) mice, followed by molecular analysis on the 21st day, we have explored the correlation between aging and mitochondrial dynamic alteration in the backdrop of chronic neuroinflammation. Following LPS stimulation, we observed microglia activation and subsequent elevation in proinflammatory cytokines (M1; TNF-α, IFN-γ) with NLRP3 activationand a concomitant reduction in the expression of anti-inflammatory markers (M2; YM1, TGF-β1) in the cerebellar tissue of aged mice compared with the young LPS and aged controls. Remarkably, senescence (p21, p27, p53) and epigenetic (HDAC2) markers were found upregulated in the cerebellum tissue of the aged LPS group, suggesting their crucial role in LPS-induced cerebellar deficit. Further, we demonstrated alteration in the antagonistic forces of mitochondrial fusion and fission with increased expression of the mitochondrial fission-related gene [FIS1] and decreased fusion-related genes [MFN1 and MFN2]. We noted increased mtDNA copy number, microglia activation, and inflammatory response of IL1β and IFN-γ post-chronic neuroinflammation in aged LPS group. Our results suggest that the crosstalk between mitochondrial dynamics and altered microglial activation paradigm in chronic neuroinflammatory conditions may be the key to understanding the cerebellar molecular mechanism.
Collapse
Affiliation(s)
- Devlina Ghosh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomti Nagar Extension, Lucknow 226028, India; Centre of Biomedical Research, SGPGIMS-Campus, Raibareli Road, Lucknow 226014, India.
| | - Gajendra Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Prabhaker Mishra
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow 226 014, Uttar Pradesh, India
| | - Aditi Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomti Nagar Extension, Lucknow 226028, India
| | - Alok Kumar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Neeraj Sinha
- Centre of Biomedical Research, SGPGIMS-Campus, Raibareli Road, Lucknow 226014, India.
| |
Collapse
|
36
|
Brsikyan LA, Poluektova EA, Poluektov MG. The gut microbiome as a factor in the development of Parkinson's disease. NEUROLOGY, NEUROPSYCHIATRY, PSYCHOSOMATICS 2023. [DOI: 10.14412/2074-2711-2023-1-90-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- L. A. Brsikyan
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| | - E. A. Poluektova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| | - M. G. Poluektov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russia (Sechenov University)
| |
Collapse
|
37
|
Botulinum neurotoxin A ameliorates depressive-like behavior in a reserpine-induced Parkinson's disease mouse model via suppressing hippocampal microglial engulfment and neuroinflammation. Acta Pharmacol Sin 2023:10.1038/s41401-023-01058-x. [PMID: 36765267 DOI: 10.1038/s41401-023-01058-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 01/16/2023] [Indexed: 02/12/2023]
Abstract
Depression is one of the common non-motor symptoms of Parkinson's disease (PD). In the clinic, botulinum neurotoxin A (BoNT/A) has been used to treat depression. In this study, we investigated the mechanisms underlying the anti-depressive effect of BoNT/A in a PD mouse model. Mice were administered reserpine (3 μg/mL in the drinking water) for 10 weeks. From the 10th week, BoNT/A (10 U·kg-1·d-1) was injected into the cheek for 3 consecutive days. We showed that chronic administration of reserpine produced the behavioral phenotypes of depression and neurochemical changes in the substantia nigra pars compacta (SNpc) and striatum. BoNT/A treatment significantly ameliorated the depressive-like behaviors, but did not improve TH activity in SNpc of reserpine-treated mice. We demonstrated that BoNT/A treatment reversed reserpine-induced complement and microglia activation in the hippocampal CA1 region. Furthermore, BoNT/A treatment significantly attenuated the microglial engulfment of presynaptic synapses, thus ameliorating the apparent synapse and spine loss in the hippocampus in the reserpine-treated mice. Moreover, BoNT/A treatment suppressed microglia-mediated expression of pro-inflammatory cytokines TNF-α and IL-1β in reserpine-treated mice. In addition, we showed that BoNT/A (0.1 U/mL) ameliorated reserpine-induced complement and microglia activation in mouse BV2 microglial cells in vitro. We conclude that BoNT/A ameliorates depressive-like behavior in a reserpine-induced PD mouse model through reversing the synapse loss mediated by classical complement induced-microglial engulfment as well as alleviating microglia-mediated proinflammatory responses. BoNT/A ameliorates depressive-like behavior, and reverses synapse loss mediated by classical complement pathway-initiated microglia engulfment as well as alleviates microglia-mediated proinflammatory response in the reserpine-induced Parkinson's disease mouse model.
Collapse
|
38
|
Oun A, Hoeksema E, Soliman A, Brouwer F, García-Reyes F, Pots H, Trombetta-Lima M, Kortholt A, Dolga AM. Characterization of Lipopolysaccharide Effects on LRRK2 Signaling in RAW Macrophages. Int J Mol Sci 2023; 24:ijms24021644. [PMID: 36675159 PMCID: PMC9865464 DOI: 10.3390/ijms24021644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/17/2023] Open
Abstract
Dysfunction of the immune system and mitochondrial metabolism has been associated with Parkinson's disease (PD) pathology. Mutations and increased kinase activity of leucine-rich repeat kinase 2 (LRRK2) are linked to both idiopathic and familial PD. However, the function of LRRK2 in the immune cells under inflammatory conditions is contradictory. Our results showed that lipopolysaccharide (LPS) stimulation increased the kinase activity of LRRK2 in parental RAW 264.7 (WT) cells. In addition to this, LRRK2 deletion in LRRK2 KO RAW 264.7 (KO) cells altered cell morphology following LPS stimulation compared to the WT cells, as shown by an increase in the cell impedance as observed by the xCELLigence measurements. LPS stimulation caused an increase in the cellular reactive oxygen species (ROS) levels in both WT and KO cells. However, WT cells displayed a higher ROS level compared to the KO cells. Moreover, LRRK2 deletion led to a reduction in interleukin-6 (IL-6) inflammatory cytokine and cyclooxygenase-2 (COX-2) expression and an increase in lactate production after LPS stimulation compared to the WT cells. These data illustrate that LRRK2 has an effect on inflammatory processes in RAW macrophages upon LPS stimulation.
Collapse
Affiliation(s)
- Asmaa Oun
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
- Department of Cell Biochemistry, Groningen Institute of Biomolecular Sciences & Biotechnology (GBB), University of Groningen, 9747 AG Groningen, The Netherlands
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt
| | - Emmy Hoeksema
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Ahmed Soliman
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
- Department of Cell Biochemistry, Groningen Institute of Biomolecular Sciences & Biotechnology (GBB), University of Groningen, 9747 AG Groningen, The Netherlands
| | - Famke Brouwer
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Fabiola García-Reyes
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Henderikus Pots
- Department of Cell Biochemistry, Groningen Institute of Biomolecular Sciences & Biotechnology (GBB), University of Groningen, 9747 AG Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Arjan Kortholt
- Department of Cell Biochemistry, Groningen Institute of Biomolecular Sciences & Biotechnology (GBB), University of Groningen, 9747 AG Groningen, The Netherlands
- YETEM-Innovative Technologies Application and Research Centre, Suleyman Demirel University, 32260 Isparta, Turkey
- Correspondence: (A.K.); (A.M.D.); Tel.: +31-50363-4206 (A.K.); +31-50363-6372 (A.M.D.)
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
- Correspondence: (A.K.); (A.M.D.); Tel.: +31-50363-4206 (A.K.); +31-50363-6372 (A.M.D.)
| |
Collapse
|
39
|
Prolonged anesthesia induces neuroinflammation and complement-mediated microglial synaptic elimination involved in neurocognitive dysfunction and anxiety-like behaviors. BMC Med 2023; 21:7. [PMID: 36600274 PMCID: PMC9814183 DOI: 10.1186/s12916-022-02705-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND) with a high incidence frequently occur in elderly surgical patients closely associated with prolonged anesthesia-induced neurotoxicity. The neuromorphopathological underpinnings of anesthesia-induced neurotoxicity have remained elusive. METHODS Prolonged anesthesia with sevoflurane was used to establish the sevoflurane-induced neurotoxicity (SIN) animal model. Morris water maze, elevated plus maze, and open field test were employed to track SIN rats' cognitive behavior and anxiety-like behaviors. We investigated the neuropathological basis of SIN through techniques such as transcriptomic, electrophysiology, molecular biology, scanning electron microscope, Golgi staining, TUNEL assay, and morphological analysis. Our work further clarifies the pathological mechanism of SIN by depleting microglia, inhibiting neuroinflammation, and C1q neutralization. RESULTS This study shows that prolonged anesthesia triggers activation of the NF-κB inflammatory pathway, neuroinflammation, inhibition of neuronal excitability, cognitive dysfunction, and anxiety-like behaviors. RNA sequencing found that genes of different types of synapses were downregulated after prolonged anesthesia. Microglial migration, activation, and phagocytosis were enhanced. Microglial morphological alterations were also observed. C1qa, the initiator of the complement cascade, and C3 were increased, and C1qa tagging synapses were also elevated. Then, we found that the "Eat Me" complement pathway mediated microglial synaptic engulfment in the hippocampus after prolonged anesthesia. Afterward, synapses were remarkably lost in the hippocampus. Furthermore, dendritic spines were reduced, and their genes were also downregulated. Depleting microglia ameliorated the activation of neuroinflammation and complement and rescued synaptic loss, cognitive dysfunction, and anxiety-like behaviors. When neuroinflammatory inhibition or C1q neutralization occurred, complement was also decreased, and synaptic elimination was interrupted. CONCLUSIONS These findings illustrated that prolonged anesthesia triggered neuroinflammation and complement-mediated microglial synaptic engulfment that pathologically caused synaptic elimination in SIN. We have demonstrated the neuromorphopathological underpinnings of SIN, which have direct therapeutic relevance for PND patients.
Collapse
|
40
|
Leinenga G, Bodea L, Schröder J, Sun G, Zhou Y, Song J, Grubman A, Polo JM, Götz J. Transcriptional signature in microglia isolated from an Alzheimer's disease mouse model treated with scanning ultrasound. Bioeng Transl Med 2023; 8:e10329. [PMID: 36684089 PMCID: PMC9842024 DOI: 10.1002/btm2.10329] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/18/2022] [Accepted: 04/04/2022] [Indexed: 01/25/2023] Open
Abstract
Transcranial scanning ultrasound combined with intravenously injected microbubbles (SUS+MB) has been shown to transiently open the blood-brain barrier and reduce the amyloid-β (Aβ) pathology in the APP23 mouse model of Alzheimer's disease (AD). This has been accomplished through the activation of microglial cells; however, their response to the SUS treatment is incompletely understood. Here, wild-type (WT) and APP23 mice were subjected to SUS+MB, using nonsonicated mice as sham controls. After 48 h, the APP23 mice were injected with methoxy-XO4 to label Aβ aggregates, followed by microglial isolation into XO4+ and XO4- populations using flow cytometry. Both XO4+ and XO4- cells were subjected to RNA sequencing and transcriptome profiling. The analysis of the microglial cells revealed a clear segregation depending on genotype (AD model vs. WT mice) and Aβ internalization (XO4+ vs. XO4- microglia), but interestingly, no differences were found between SUS+MB and sham in WT mice. Differential gene expression analysis in APP23 mice detected 278 genes that were significantly changed by SUS+MB in the XO4+ cells (248 up/30 down) and 242 in XO- cells (225 up/17 down). Pathway analysis highlighted differential expression of genes related to the phagosome pathway and marked upregulation of cell cycle-related transcripts in XO4+ and XO4- microglia isolated from SUS+MB-treated APP23 mice. Together, this highlights the complexity of the microglial response to transcranial ultrasound, with potential applications for the treatment of AD.
Collapse
Affiliation(s)
- Gerhard Leinenga
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbane (St Lucia Campus)QueenslandAustralia
| | - Liviu‐Gabriel Bodea
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbane (St Lucia Campus)QueenslandAustralia
| | - Jan Schröder
- Department of Anatomy & Developmental Biology and the Australian Regenerative Medicine InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Giuzhi Sun
- Department of Anatomy & Developmental Biology and the Australian Regenerative Medicine InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Yichen Zhou
- Department of Anatomy & Developmental Biology and the Australian Regenerative Medicine InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Jae Song
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbane (St Lucia Campus)QueenslandAustralia
| | - Alexandra Grubman
- Department of Anatomy & Developmental Biology and the Australian Regenerative Medicine InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Jose M. Polo
- Department of Anatomy & Developmental Biology and the Australian Regenerative Medicine InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandBrisbane (St Lucia Campus)QueenslandAustralia
| |
Collapse
|
41
|
Mayer E, Horn J, Mayer E, Chen S. Role of the gut microbiome in the pathophysiology of brain disorders. NEUROBIOLOGY OF BRAIN DISORDERS 2023:913-928. [DOI: 10.1016/b978-0-323-85654-6.00058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Regensburger M, Rasul Chaudhry S, Yasin H, Zhao Y, Stadlbauer A, Buchfelder M, Kinfe T. Emerging roles of leptin in Parkinson's disease: Chronic inflammation, neuroprotection and more? Brain Behav Immun 2023; 107:53-61. [PMID: 36150585 DOI: 10.1016/j.bbi.2022.09.010] [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: 03/16/2022] [Revised: 08/22/2022] [Accepted: 09/16/2022] [Indexed: 12/13/2022] Open
Abstract
An increasing body of experimental evidence implicates a relationship between immunometabolic deterioration and the progression of Parkinson's disease (PD) with a dysregulation of central and peripheral neuroinflammatory networks mediated by circulating adipokines, in particular leptin. We screened the current literature on the role of adipokines in PD. Hence, we searched known databases (PubMed, MEDLINE/OVID) and reviewed original and review articles using the following terms: "leptin/ObR", "Parkinson's disease", "immune-metabolism", "biomarkers" and "neuroinflammation". Focusing on leptin, we summarize and discuss the existing in vivo and in vitro evidence on how adipokines may be protective against neurodegeneration, but at the same time contribute to the progression of PD. These components of the adipose brain axis represent a hitherto underestimated pathway to study systemic influences on dopaminergic degeneration. In addition, we give a comprehensive update on the potential of adjunctive therapeutics in PD targeting leptin, leptin-receptors, and associated pathways. Further experimental and clinical trials are needed to elucidate the mechanisms of action and the value of central and peripheral adipose-immune-metabolism molecular phenotyping in order to develop and validate the differential roles of different adipokines as potential therapeutic target for PD patients.
Collapse
Affiliation(s)
- Martin Regensburger
- Department of Molecular Neurology, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany; Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Shafqat Rasul Chaudhry
- Obaid Noor Institute of Medical Sciences (ONIMS), Mianwali, Pakistan; Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, 44000 Islamabad, Pakistan
| | - Hammad Yasin
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, 44000 Islamabad, Pakistan
| | - Yining Zhao
- Department of Neurosurgery, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andreas Stadlbauer
- Department of Neurosurgery, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Thomas Kinfe
- Division of Functional Neurosurgery and Stereotaxy, Friedrich-Alexander University (FAU), Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
43
|
Dingwall CB, Strickland A, Yum SW, Yim AK, Zhu J, Wang PL, Yamada Y, Schmidt RE, Sasaki Y, Bloom AJ, DiAntonio A, Milbrandt J. Macrophage depletion blocks congenital SARM1-dependent neuropathy. J Clin Invest 2022; 132:e159800. [PMID: 36287209 PMCID: PMC9711884 DOI: 10.1172/jci159800] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Axon loss contributes to many common neurodegenerative disorders. In healthy axons, the axon survival factor NMNAT2 inhibits SARM1, the central executioner of programmed axon degeneration. We identified 2 rare NMNAT2 missense variants in 2 brothers afflicted with a progressive neuropathy syndrome. The polymorphisms resulted in amino acid substitutions V98M and R232Q, which reduced NMNAT2 NAD+-synthetase activity. We generated a mouse model to mirror the human syndrome and found that Nmnat2V98M/R232Q compound-heterozygous CRISPR mice survived to adulthood but developed progressive motor dysfunction, peripheral axon loss, and macrophage infiltration. These disease phenotypes were all SARM1-dependent. Remarkably, macrophage depletion therapy blocked and reversed neuropathic phenotypes in Nmnat2V98M/R232Q mice, identifying a SARM1-dependent neuroimmune mechanism as a key driver of disease pathogenesis. These findings demonstrate that SARM1 induced inflammatory neuropathy and highlight the potential of immune therapy as a treatment for this rare syndrome and other neurodegenerative conditions associated with NMNAT2 loss and SARM1 activation.
Collapse
Affiliation(s)
- Caitlin B. Dingwall
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sabrina W. Yum
- Division of Neurology, Children’s Hospital of Philadelphia, Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Aldrin K.Y. Yim
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jian Zhu
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter L. Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yurie Yamada
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert E. Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - A. Joseph Bloom
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
| | - Aaron DiAntonio
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
| |
Collapse
|
44
|
Chiapparini L, Zorzi G. Early Neuroimaging Markers in β Propeller Protein-Associated Neurodegeneration. AJNR Am J Neuroradiol 2022; 43:1815-1816. [PMID: 36396333 DOI: 10.3174/ajnr.a7723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
| | - G Zorzi
- Department of Paediatric Neuroscience Fondazione Institute for Hospitalization and Healthcare Istituto Neurologico Carlo Besta Milan, Italy
| |
Collapse
|
45
|
Deng I, Bobrovskaya L. Lipopolysaccharide mouse models for Parkinson's disease research: a critical appraisal. Neural Regen Res 2022; 17:2413-2417. [PMID: 35535880 PMCID: PMC9120679 DOI: 10.4103/1673-5374.331866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/25/2022] Open
Abstract
Parkinson's disease, the most common movement disorder, has a strong neuroinflammatory aspect. This is evident by increased pro-inflammatory cytokines in the serum, and the presence of activated microglial cells, and inflammatory cytokines in the substantia nigra of post-mortem brains as well as cerebrospinal fluid of Parkinson's disease patients. The central and peripheral neuroinflammatory aspects of Parkinson's disease can be investigated in vivo via administration of the inflammagen lipopolysaccharide, a component of the cell wall of gram-negative bacteria. In this mini-review, we will critically evaluate different routes of lipopolysaccharide administration (including intranasal systemic and stereotasic), their relevance to clinical Parkinson's disease as well as the recent findings in lipopolysaccharide mouse models. We will also share our own experiences with systemic and intrastriatal lipopolysaccharide models in C57BL/6 mice and will discuss the usefulness of lipopolysaccharide mouse models for future research in the field.
Collapse
Affiliation(s)
- Isaac Deng
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
46
|
Ayeni EA, Aldossary AM, Ayejoto DA, Gbadegesin LA, Alshehri AA, Alfassam HA, Afewerky HK, Almughem FA, Bello SM, Tawfik EA. Neurodegenerative Diseases: Implications of Environmental and Climatic Influences on Neurotransmitters and Neuronal Hormones Activities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph191912495. [PMID: 36231792 PMCID: PMC9564880 DOI: 10.3390/ijerph191912495] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 05/23/2023]
Abstract
Neurodegenerative and neuronal-related diseases are major public health concerns. Human vulnerability to neurodegenerative diseases (NDDs) increases with age. Neuronal hormones and neurotransmitters are major determinant factors regulating brain structure and functions. The implications of environmental and climatic changes emerged recently as influence factors on numerous diseases. However, the complex interaction of neurotransmitters and neuronal hormones and their depletion under environmental and climatic influences on NDDs are not well established in the literature. In this review, we aim to explore the connection between the environmental and climatic factors to NDDs and to highlight the available and potential therapeutic interventions that could use to improve the quality of life and reduce susceptibility to NDDs.
Collapse
Affiliation(s)
- Emmanuel A. Ayeni
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ahmad M. Aldossary
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Daniel A. Ayejoto
- Department of Industrial Chemistry, University of Ilorin, Ilorin 240003, Nigeria
| | - Lanre A. Gbadegesin
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Mountain Hazards and Environment, Chinese Academy of Sciences, Chengdu 610041, China
| | - Abdullah A. Alshehri
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Haya A. Alfassam
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Henok K. Afewerky
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- School of Allied Health Professions, Asmara College of Health Sciences, Asmara P.O. Box 1220, Eritrea
| | - Fahad A. Almughem
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Saidu M. Bello
- Institute of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary
| | - Essam A. Tawfik
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| |
Collapse
|
47
|
García-Revilla J, Herrera AJ, de Pablos RM, Venero JL. Inflammatory Animal Models of Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S165-S182. [PMID: 35662128 PMCID: PMC9535574 DOI: 10.3233/jpd-213138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Accumulating evidence suggests that microglia and peripheral immune cells may play determinant roles in the pathogenesis of Parkinson’s disease (PD). Consequently, there is a need to take advantage of immune-related models of PD to study the potential contribution of microglia and peripheral immune cells to the degeneration of the nigrostriatal system and help develop potential therapies for PD. In this review, we have summarised the main PD immune models. From a historical perspective, we highlight first the main features of intranigral injections of different pro-inflammogens, including lipopolysaccharide (LPS), thrombin, neuromelanin, etc. The use of adenoviral vectors to promote microglia-specific overexpression of different molecules in the ventral mesencephalon, including α-synuclein, IL-1β, and TNF, are also presented and briefly discussed. Finally, we summarise different models associated with peripheral inflammation whose contribution to the pathogenesis of neurodegenerative diseases is now an outstanding question. Illustrative examples included systemic LPS administration and dextran sulfate sodium-induced colitis in rodents.
Collapse
Affiliation(s)
- Juan García-Revilla
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Antonio J. Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rocío M. de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luis Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| |
Collapse
|
48
|
Choi HR, Ha JS, Kim EA, Cho SW, Yang SJ. MiR-30a-5p and miR-153-3p regulate LPS-induced neuroinflammatory response and neuronal apoptosis by targeting NeuroD1. BMB Rep 2022; 55:447-452. [PMID: 35651331 PMCID: PMC9537026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 03/08/2024] Open
Abstract
Neurogenic differentiation 1 (NeuroD1) is an essential transcription factor for neuronal differentiation, maturation, and survival, and is associated with inflammation in lipopolysaccharide (LPS)- induced glial cells; however, the concrete mechanisms are still ambiguous. Therefore, we investigated whether NeuroD1-targeting miRNAs affect inflammation and neuronal apoptosis, as well as the underlying mechanism. First, we confirmed that miR-30a-5p and miR-153-3p, which target NeuroD1, reduced NeuroD1 expression in microglia and astrocytes. In LPS-induced microglia, miR-30a-5p and miR-153-3p suppressed pro-inflammatory cytokines, reactive oxygen species, the phosphorylation of c-Jun N-terminal kinase, extracellular-signal-regulated kinase (ERK), and p38, and the expression of cyclooxygenase and inducible nitric oxide synthase (iNOS) via the NF-κB pathway. Moreover, miR-30a-5p and miR-153-3p inhibited the expression of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes, NLRP3, cleaved caspase-1, and IL-1β, which are involved in the innate immune response. In LPS-induced astrocytes, miR-30a-5p and miR-153-3p reduced ERK phosphorylation and iNOS expression via the STAT-3 pathway. Notably, miR-30a-5p exerted greater anti-inflammatory effects than miR-153-3p. Together, these results indicate that miR-30a-5p and miR-153-3p inhibit MAPK/NF-κB pathway in microglia as well as ERK/STAT-3 pathway in astrocytes to reduce LPS-induced neuronal apoptosis. This study highlights the importance of NeuroD1 in microglia and astrocytes neuroinflammation and suggests that it can be regulated by miR-30a-5p and miR-153-3p. [BMB Reports 2022; 55(9): 447-452].
Collapse
Affiliation(s)
- Hye-Rim Choi
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Ji Sun Ha
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| | - Eun-A Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea
| |
Collapse
|
49
|
Shi Q, Chang C, Saliba A, Bhat MA. Microglial mTOR Activation Upregulates Trem2 and Enhances β-Amyloid Plaque Clearance in the 5XFAD Alzheimer's Disease Model. J Neurosci 2022; 42:5294-5313. [PMID: 35672148 PMCID: PMC9270922 DOI: 10.1523/jneurosci.2427-21.2022] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway plays a major role in key cellular processes including metabolism and differentiation; however, the role of mTOR in microglia and its importance in Alzheimer's disease (AD) have remained largely uncharacterized. We report that selective loss of Tsc1, a negative regulator of mTOR, in microglia in mice of both sexes, caused mTOR activation and upregulation of Trem2 with enhanced β-Amyloid (Aβ) clearance, reduced spine loss, and improved cognitive function in the 5XFAD AD mouse model. Combined loss of Tsc1 and Trem2 in microglia led to reduced Aβ clearance and increased Aβ plaque burden revealing that Trem2 functions downstream of mTOR. Tsc1 mutant microglia showed increased phagocytosis with upregulation of CD68 and Lamp1 lysosomal proteins. In vitro studies using Tsc1-deficient microglia revealed enhanced endocytosis of the lysosomal tracker indicator Green DND-26 suggesting increased lysosomal activity. Incubation of Tsc1-deficient microglia with fluorescent-labeled Aβ revealed enhanced Aβ uptake and clearance, which was blunted by rapamycin, an mTOR inhibitor. In vivo treatment of mice of relevant genotypes in the 5XFAD background with rapamycin, affected microglial activity, decreased Trem2 expression and reduced Aβ clearance causing an increase in Aβ plaque burden. Prolonged treatment with rapamycin caused even further reduction of mTOR activity, reduction in Trem2 expression, and increase in Aβ levels. Together, our findings reveal that mTOR signaling in microglia is critically linked to Trem2 regulation and lysosomal biogenesis, and that the upregulation of Trem2 in microglia through mTOR activation could be exploited toward better therapeutic avenues to Aβ-related AD pathologies.SIGNIFICANCE STATEMENT Mechanistic target of rapamycin (mTOR) signaling pathway is a key regulator for major cellular metabolic processes. However, the link between mTOR signaling and Alzheimer's disease (AD) is not well understood. In this study, we provide compelling in vivo evidence that mTOR activation in microglia would benefit β-Amyloid (Aβ)-related AD pathologies, as it upregulates Trem2, a key receptor for Aβ plaque uptake. Inhibition of mTOR pathway with rapamycin, a well-established immunosuppressant, downregulated Trem2 in microglia and reduced Aβ plaque clearance indicating that mTOR inactivation may be detrimental in Aβ-associated AD patients. This finding will have a significant public health impact and benefit, regarding the usage of rapamycin in AD patients, which we believe will aggravate the Aβ-related AD pathologies.
Collapse
Affiliation(s)
- Qian Shi
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Cheng Chang
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Afaf Saliba
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| |
Collapse
|
50
|
Seo Y, Bang S, Son J, Kim D, Jeong Y, Kim P, Yang J, Eom JH, Choi N, Kim HN. Brain physiome: A concept bridging in vitro 3D brain models and in silico models for predicting drug toxicity in the brain. Bioact Mater 2022; 13:135-148. [PMID: 35224297 PMCID: PMC8843968 DOI: 10.1016/j.bioactmat.2021.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, adverse reactions to pharmaceuticals have been evaluated using 2D in vitro models and animal models. However, with increasing computational power, and as the key drivers of cellular behavior have been identified, in silico models have emerged. These models are time-efficient and cost-effective, but the prediction of adverse reactions to unknown drugs using these models requires relevant experimental input. Accordingly, the physiome concept has emerged to bridge experimental datasets with in silico models. The brain physiome describes the systemic interactions of its components, which are organized into a multilevel hierarchy. Because of the limitations in obtaining experimental data corresponding to each physiome component from 2D in vitro models and animal models, 3D in vitro brain models, including brain organoids and brain-on-a-chip, have been developed. In this review, we present the concept of the brain physiome and its hierarchical organization, including cell- and tissue-level organizations. We also summarize recently developed 3D in vitro brain models and link them with the elements of the brain physiome as a guideline for dataset collection. The connection between in vitro 3D brain models and in silico modeling will lead to the establishment of cost-effective and time-efficient in silico models for the prediction of the safety of unknown drugs.
Collapse
Affiliation(s)
- Yoojin Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yong Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihun Yang
- Next&Bio Inc., Seoul, 02841, Republic of Korea
| | - Joon-Ho Eom
- Medical Device Research Division, National Institute of Food and Drug Safety Evaluation, Cheongju, 28159, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|