1
|
Serradas ML, Ding Y, Martorell PV, Kulińska I, Castro-Gomez S. Therapeutic Targets in Innate Immunity to Tackle Alzheimer's Disease. Cells 2024; 13:1426. [PMID: 39272998 PMCID: PMC11394242 DOI: 10.3390/cells13171426] [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: 07/30/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
There is an urgent need for effective disease-modifying therapeutic interventions for Alzheimer's disease (AD)-the most prevalent cause of dementia with a profound socioeconomic burden. Most clinical trials targeting the classical hallmarks of this disease-β-amyloid plaques and neurofibrillary tangles-failed, showed discrete clinical effects, or were accompanied by concerning side effects. There has been an ongoing search for novel therapeutic targets. Neuroinflammation, now widely recognized as a hallmark of all neurodegenerative diseases, has been proven to be a major contributor to AD pathology. Here, we summarize the role of neuroinflammation in the pathogenesis and progression of AD and discuss potential targets such as microglia, TREM2, the complement system, inflammasomes, and cytosolic DNA sensors. We also present an overview of ongoing studies targeting specific innate immune system components, highlighting the progress in this field of drug research while bringing attention to the delicate nature of innate immune modulations in AD.
Collapse
Affiliation(s)
- Maria L Serradas
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Yingying Ding
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Paula V Martorell
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Ida Kulińska
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Sergio Castro-Gomez
- Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
- Center for Neurology, Department of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
2
|
Fink A, Ben Hur D, Wani NA, Cohen H, Segev-Zarko LA, Arnusch CJ, Shai Y. Development of Nontoxic Peptides for Lipopolysaccharide Neutralization and Sepsis Treatment. ACS Pharmacol Transl Sci 2024; 7:1795-1806. [PMID: 38898940 PMCID: PMC11184611 DOI: 10.1021/acsptsci.4c00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 06/21/2024]
Abstract
Host defense peptides (HDPs), also named antimicrobial peptides (AMPs), are increasingly being recognized for serving multiple functions in protecting the host from infection and disease. Previous studies have shown that various HDPs can also neutralize lipopolysaccharide (LPS, endotoxin), as well as lipoteichoic acid (LTA), inducing macrophage activation. However, antimicrobial activity is usually accompanied by systemic toxicity which makes it difficult to use HDPs as antiendotoxin agents. Here we report that key parameters can uncouple these two functions yielding nontoxic peptides with potent LPS and LTA neutralization activities in vitro and in animal models. The data reveal that peptide length, the number, and the placement of positive charges are important parameters involved in LPS neutralization. Crucially, the peptide exhibited a separation between its membrane-disrupting and antimicrobial properties, effectively decoupling them from its ability to neutralize LPS. This essential distinction prevented systemic toxicity and led to the peptide's complete rescue of mice suffering from severe septic shock in two distinct models. Strong binding to LPS, changes in structure, and oligomerization state upon LPS binding were important factors that determined the activity of the peptides. In the face of the increasing threat of septic shock worldwide, it is crucial to grasp how we can neutralize harmful substances like LPS. This knowledge is vital for creating nontoxic treatments for sepsis.
Collapse
Affiliation(s)
- Avner Fink
- Department
of Biomolecular Sciences, Weizmann Institute
of Science, Rehovot 76100, Israel
- MilliporeSigma
Life Science, Kiryat
Hamada 13, 9777613 Jerusalem, Israel
| | - Daniel Ben Hur
- Department
of Biomolecular Sciences, Weizmann Institute
of Science, Rehovot 76100, Israel
| | - Naiem Ahmad Wani
- Department
of Biomolecular Sciences, Weizmann Institute
of Science, Rehovot 76100, Israel
| | - Hadar Cohen
- Department
of Biomolecular Sciences, Weizmann Institute
of Science, Rehovot 76100, Israel
| | - Li-Av Segev-Zarko
- Department
of Biomolecular Sciences, Weizmann Institute
of Science, Rehovot 76100, Israel
| | - Christopher J. Arnusch
- Department
of Desalination and Water Treatment, Zuckerberg Institute for Water
Research, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede-Boqer Campus 8499000, Israel
| | - Yechiel Shai
- Department
of Biomolecular Sciences, Weizmann Institute
of Science, Rehovot 76100, Israel
| |
Collapse
|
3
|
Castro-Gomez S, Heneka MT. Innate immune activation in neurodegenerative diseases. Immunity 2024; 57:790-814. [PMID: 38599171 DOI: 10.1016/j.immuni.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Activation of the innate immune system following pattern recognition receptor binding has emerged as one of the major pathogenic mechanisms in neurodegenerative disease. Experimental, epidemiological, pathological, and genetic evidence underscores the meaning of innate immune activation during the prodromal as well as clinical phases of several neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia. Importantly, innate immune activation and the subsequent release of inflammatory mediators contribute mechanistically to other hallmarks of neurodegenerative diseases such as aberrant proteostatis, pathological protein aggregation, cytoskeleton abnormalities, altered energy homeostasis, RNA and DNA defects, and synaptic and network disbalance and ultimately to the induction of neuronal cell death. In this review, we discuss common mechanisms of innate immune activation in neurodegeneration, with particular emphasis on the pattern recognition receptors (PRRs) and other receptors involved in the detection of damage-associated molecular patterns (DAMPs).
Collapse
Affiliation(s)
- Sergio Castro-Gomez
- Center for Neurology, Department of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, 53127 Bonn, Germany; Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Paramasivam S, Perumal SS, Ekambaram SP. Computational Deciphering of the Role of S100A8 and S100A9 Proteins and Their Changes in the Structure Assembly Influences Their Interaction with TLR4, RAGE, and CD36. Protein J 2024; 43:243-258. [PMID: 38431537 DOI: 10.1007/s10930-024-10186-0] [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] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
S100A8 and S100A9 belong to the calcium-binding, damage associated molecular pattern (DAMP) proteins shown to aggravate the pathogenesis of rheumatoid arthritis (RA) through their interaction with the TLR4, RAGE and CD36 receptors. S100A8 and S100A9 proteins tend to exist in monomeric, homo and heterodimeric forms, which have been implicated in the pathogenesis of RA, via interacting with Pattern Recognition receptors (PRRs). The study aims to assess the influence of changes in the structure and biological assembly of S100A8 and S100A9 proteins as well as their interaction with significant receptors in RA through computational methods and surface plasmon resonance (SPR) analysis. Molecular docking analysis revealed that the S100A9 homodimer and S100A8/A9 heterodimer showed higher binding affinity towards the target receptors. Most S100 proteins showed good binding affinity towards TLR4 compared to other receptors. Based on the 50 ns MD simulations, TLR4, RAGE, and CD36 formed stable complexes with the monomeric and dimeric forms of S100A8 and S100A9 proteins. However, SPR analysis showed that the S100A8/A9 heterodimers formed stable complexes and exhibited high binding affinity towards the receptors. SPR data also indicated that TLR4 and its interactions with S100A8/A9 proteins may play a primary role in the pathogenesis of RA, with additional contributions from CD36 and RAGE interactions. Subsequent in vitro and in vivo investigations are warranted to corroborate the involvement of S100A8/A9 and the expression of TLR4, RAGE, and CD36 in the pathophysiology of RA.
Collapse
Affiliation(s)
- Sivasakthi Paramasivam
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Senthamil Selvan Perumal
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Sanmuga Priya Ekambaram
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
5
|
Peggion C, Calì T, Brini M. Mitochondria Dysfunction and Neuroinflammation in Neurodegeneration: Who Comes First? Antioxidants (Basel) 2024; 13:240. [PMID: 38397838 PMCID: PMC10885966 DOI: 10.3390/antiox13020240] [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: 02/01/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) encompass an assorted array of disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, each characterised by distinct clinical manifestations and underlying pathological mechanisms. While some cases have a genetic basis, many NDs occur sporadically. Despite their differences, these diseases commonly feature chronic neuroinflammation as a hallmark. Consensus has recently been reached on the possibility that mitochondria dysfunction and protein aggregation can mutually contribute to the activation of neuroinflammatory response and thus to the onset and progression of these disorders. In the present review, we discuss the contribution of mitochondria dysfunction and neuroinflammation to the aetiology and progression of NDs, highlighting the possibility that new potential therapeutic targets can be identified to tackle neurodegenerative processes and alleviate the progression of these pathologies.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biology, University of Padova, 35131 Padova, Italy;
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
| | - Marisa Brini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
6
|
Hamzeh O, Rabiei F, Shakeri M, Parsian H, Saadat P, Rostami-Mansoor S. Mitochondrial dysfunction and inflammasome activation in neurodegenerative diseases: Mechanisms and therapeutic implications. Mitochondrion 2023; 73:S1567-7249(23)00087-9. [PMID: 39492438 DOI: 10.1016/j.mito.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/02/2023] [Accepted: 10/28/2023] [Indexed: 11/05/2024]
Abstract
Impaired mitochondrial function is crucial to the pathogenesis of several neurodegenerative diseases. It causes the release of mitochondrial DNA (mtDNA), mitochondrial reactive oxygen species (mtROS), ATP, and cardiolipin, which activate the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. NLRP3 inflammasome is an important innate immune system element contributing to neuroinflammation and neurodegeneration. Therefore, targeting the NLRP3 inflammasome has become an interesting therapeutic approach for treating neurodegenerative diseases. This review describes the role of mitochondrial abnormalities and over-activated inflammasomes in the progression of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple sclerosis (MS), Amyotrophic lateral sclerosis (ALS), and Friedrich ataxia (FRDA). We also discuss the therapeutic strategies focusing on signaling pathways associated with inflammasome activation, which potentially alleviate neurodegenerative symptoms and impede disease progression.
Collapse
Affiliation(s)
- Olia Hamzeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Rabiei
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mahdi Shakeri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Payam Saadat
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sahar Rostami-Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
7
|
Hyun GH, Cho IH, Yang YY, Jeong DH, Kang YP, Kim YS, Lee SJ, Kwon SW. Mechanisms of interactions in pattern-recognition of common glycostructures across pectin-derived heteropolysaccharides by Toll-like receptor 4. Carbohydr Polym 2023; 314:120921. [PMID: 37173020 DOI: 10.1016/j.carbpol.2023.120921] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
Complex pectin, originating from terrestrial plant cell walls has been attracting research attention as a promising source of a new innate immune modulator. Numerous bioactive polysaccharides associated with pectin are newly reported every year, but the general mechanism of their immunological action remains unclear owing to the complexity and heterogeneity of pectin. Herein, we systematically investigated the interactions in pattern-recognition for common glycostructures of pectic heteropolysaccharides (HPSs) by Toll-like receptors (TLRs). The compositional similarity of glycosyl residues derived from pectic HPS was confirmed by conducting systematic reviews, leading to molecular modeling of representative pectic segments. Via structural investigation, the inner concavity of leucine-rich repeats of TLR4 was predicted to act as a binding motif for carbohydrate recognition, and subsequent simulations predicted the binding modes and conformations. We experimentally demonstrated that pectic HPS exhibits the non-canonical and multivalent binding aspects for TLR4 resulting in receptor activation. Furthermore, we showed that pectic HPSs were selectively clustered with TLR4 during endocytosis, inducing downstream signals to cause phenotypic activation of macrophages. Overall, we have presented a better explanation for the pattern recognition of pectic HPS and further proposed an approach to understand the interaction between complex carbohydrates and proteins.
Collapse
Affiliation(s)
- Gyu Hwan Hyun
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - In Ho Cho
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon Young Yang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Da-Hye Jeong
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Yun Pyo Kang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Seul Ji Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Tang Y, Hu YW, Wang SH, Zhou M, Ding YJ, Cai SQ, Tang B, Wang SG. RNAi-mediated CrebA silencing inhibits reproduction and immunity in Locusta migratoria manilensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104711. [PMID: 37062456 DOI: 10.1016/j.dci.2023.104711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/27/2023]
Abstract
Locusta migratoria manilensis is a major agricultural pest that causes severe direct and indirect damage to several crops. Thus, to provide a theoretical foundation for pest control, the role of CrebA in the reproduction and immune regulation of L. migratoria was investigated. CrebA is a bZIP transcription factor that critically regulates intracellular protein secretion. In this study, CrebA was widely expressed in the brain, fat body, integument, midgut, and reproductive tissues of different maturity stages of adult locusts, especially in the female fat body. RNA interfering (RNAi)-mediated silencing of CrebA inhibited locusts ovarian development, and key reproduction gene expressions, Vgs, VgRs, Chico, and JHAMT were downregulated. After the locusts were injected with Micrococcus luteus or Escherichia coli, M. luteus activated lysozyme expression, while the E. coli activated both phenol oxidase cascade and lysozyme expression. Furthermore, both bacteria stimulated the upregulation of the antimicrobial peptide genes DEF3 and DEF4. However, CrebA silencing is fatal to locusts infection with E. coli, with a mortality rate of up to 96.3%, and resulted in a significant decrease in the expression of DEF3 and DEF4 and changes in the activities of phenol oxidase and lysozyme of locusts infected by bacteria. Collectively, CrebA may be involved in diverse biological processes, including reproduction and immunity. CrebA inhibited locusts reproduction by regulating JH signaling pathway and inhibits the expression of immune genes TLR6, IMD, and AMPs. These results demonstrate CrebA seems to play a crucial role in reproduction and innate immunity.
Collapse
Affiliation(s)
- Ya Tang
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Yao-Wen Hu
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Shao-Hua Wang
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Min Zhou
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Yan-Juan Ding
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Si-Qi Cai
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Bin Tang
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China
| | - Shi-Gui Wang
- Hangzhou Key Laboratory of Animal Adaptation and Evolution, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, PR China.
| |
Collapse
|
9
|
Gu L, Ju Y, Hu M, Zheng M, Li Q, Zhang X. Research progress of PPARγ regulation of cholesterol and inflammation in Alzheimer's disease. Metab Brain Dis 2023; 38:839-854. [PMID: 36723831 DOI: 10.1007/s11011-022-01139-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/29/2022] [Indexed: 02/02/2023]
Abstract
Peroxidase proliferator receptors (PPARs) are defined as key sensors and regulators of cell metabolism, transcription factors activated by ligands, involved in lipid, glucose and amino acid metabolism, participating in the processes of cell differentiation, apoptosis, inflammation regulation, and acute and chronic nerve damage. Among them, PPARγ is expressed in different brain regions and can regulate lipid metabolism, mitochondrial disorders, oxidative stress, and cell apoptosis. It has anti-inflammatory activity and shows neuroprotection. The regulation of Aβ levels in Alzheimer's disease involves cholesterol metabolism and inflammation, so this article first analyzes the biological functions of PPARγ, then mainly focuses on the relationship between cholesterol and inflammation and Aβ, and elaborates on the regulation of PPARγ on key proteins and the corresponding molecules, which provides new ideas for the treatment of AD.
Collapse
Affiliation(s)
- Lili Gu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Yue Ju
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Min Hu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Miao Zheng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Qin Li
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Xinyue Zhang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China.
| |
Collapse
|
10
|
Hou J, Chen Y, Grajales-Reyes G, Colonna M. TREM2 dependent and independent functions of microglia in Alzheimer's disease. Mol Neurodegener 2022; 17:84. [PMID: 36564824 PMCID: PMC9783481 DOI: 10.1186/s13024-022-00588-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/02/2022] [Indexed: 12/25/2022] Open
Abstract
Microglia are central players in brain innate immunity and have been the subject of extensive research in Alzheimer's disease (AD). In this review, we aim to summarize the genetic and functional discoveries that have advanced our understanding of microglia reactivity to AD pathology. Given the heightened AD risk posed by rare variants of the microglial triggering receptor expressed on myeloid cells 2 (TREM2), we will focus on the studies addressing the impact of this receptor on microglia responses to amyloid plaques, tauopathy and demyelination pathologies in mouse and human. Finally, we will discuss the implications of recent discoveries on microglia and TREM2 biology on potential therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jinchao Hou
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yun Chen
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Gary Grajales-Reyes
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Marco Colonna
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
11
|
Al-Ghraiybah NF, Wang J, Alkhalifa AE, Roberts AB, Raj R, Yang E, Kaddoumi A. Glial Cell-Mediated Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2022; 23:10572. [PMID: 36142483 PMCID: PMC9502483 DOI: 10.3390/ijms231810572] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells' microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation.
Collapse
Affiliation(s)
- Nour F. Al-Ghraiybah
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amer E. Alkhalifa
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Andrew B. Roberts
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Euitaek Yang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
| |
Collapse
|
12
|
Navia-Pelaez JM, Silva Dias MT, Ariza Orellano LA, Campos GP, Alvarez-Leite J, Campos PP, Aggum Capettini LS. Dual effect of amitriptyline in the control of vascular tone: Direct blockade of calcium channel in smooth muscle cells and reduction of TLR4-dependent NO production in endothelial cells. Eur J Pharmacol 2022; 934:175255. [PMID: 36088982 DOI: 10.1016/j.ejphar.2022.175255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/01/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND PURPOSE Amitriptyline (AM) is a classical and typical tricyclic antidepressant drug. Despite its well-known effects on the nervous system, it has been described to work as a TLR4 antagonist and several clinical works suggested some unexpected cardiovascular effects. The role of amitriptyline on vascular tone is not clear, thus we hypothesized that amitriptyline has a double effect on vascular tone by both endothelial TLR4-dependent nitric oxide down-regulation and calcium channel blockade in smooth muscle cells. EXPERIMENTAL APPROACH Changes in isometric tension were recorded on a wire myograph. NO production was evaluated by fluorescence microscopy and flow cytometry in the mouse aorta and EAhy926 cells using DAF fluorescence intensity. Calcium influx was evaluated in A7r5 cells by flow cytometry. Western blot was used to analyze eNOS and nNOS phosphorylation. KEY RESULTS AM reduced PE-induced contraction by calcium influx diminution in smooth muscle cells (F/F0 = 225.6 ± 15.9 and 118.6 ± 17.6 to CT and AM, respectively). AM impaired Ach-dependent vasodilation (Emax = 95.8 ± 1.4; 78.1 ± 1.8; 60.4 ± 2.9 and -7.4 ± 1.0 for CT, 0.01, 0,1 and 1 μmol/L AM, respectively) through reduction of calcium influx and NO availability and TLR4 antagonism in a concentration-dependent manner. AM or TLR4 gene deletion significantly reduced NO production (Fluorescence = 9503 ± 871.7, 2561 ± 282, 4771 ± 728 and 1029 ± 103 to CT, AM, TLR4-/- and AM + TLR4-/-, respectively) by an increase in nNOSser852 and reduction in eNOSser1177 phosphorylation in endothelial cells. CONCLUSIONS AND IMPLICATIONS Our data show that amitriptyline impaired vascular function through two different mechanisms: blockade of TLR4 in endothelial cells and consequent decrease in NO production and calcium influx reduction in smooth muscle and endothelial cells. We also suggest, for the first time, nNOS activity reduction by AM in non-neuronal cells.
Collapse
Affiliation(s)
- Juliana Maria Navia-Pelaez
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627 - Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte, MG, Brazil; Department of Medicine. University of California San Diego, Biomedical Sciences Building, Room 1081 9500 Gilman Drive, La Jolla, CA, 92093-0682, USA.
| | - Melissa Tainan Silva Dias
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627 - Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte, MG, Brazil.
| | - Laura Alejandra Ariza Orellano
- Department of General Pathology, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Pampulha, 31270-901, Belo Horizonte, MG, Brazil.
| | - Gianne Paul Campos
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627 - Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte, MG, Brazil.
| | - Jacqueline Alvarez-Leite
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627 - Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte, MG, Brazil.
| | - Paula Peixoto Campos
- Department of General Pathology, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil. Av. Antônio Carlos, 6627, Pampulha, 31270-901, Belo Horizonte, MG, Brazil.
| | - Luciano Santos Aggum Capettini
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos 6627 - Campus Pampulha, Cx Post 468, CEP 31270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
13
|
Wu J, Li T, Mao G, Cha X, Fei S, Miao B. The involvement of Pellino-1 downregulation in the modulation of visceral hypersensitivity via the TLR4/NF-κB pathway in the rat fastigial nucleus. Neurosci Lett 2022; 787:136815. [PMID: 35901910 DOI: 10.1016/j.neulet.2022.136815] [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: 02/18/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022]
Abstract
Irritable bowel syndrome (IBS) is a common functional bowel disorder whose key characteristics include chronic visceral hypersensitivity (CVH) and abnormal brain-gut interactions. Pellino-1 is an E3 ubiquitin ligase, mediating the degradation or modification of targeted proteins. Some brain regions, such as the fastigial nucleus (FN), may play important roles in CVH; however, the molecular mechanism underlying this phenomenon is not clear. In this study, we assessed the roles of Pellino-1 within the FN in modulating VH by generating a colorectal distention (CRD) model in male Sprague-Dawley rats. Our results showed that the downregulation of Pellino-1 in the fastigial nucleus (FN) was involved in the modulation of visceral hypersensitivity. The expression of Pellino-1 was downregulated in the FN of adult CRD rats compared with control rats, whereas TLR4 and NF-κB were upregulated in the CRD model. To overexpress Pellino-1, a lentivirus specifically expressing Pellino-1 and green fluorescent protein was administered into the FN. The overexpression of Pellino-1 increased the visceral sensitivity of CRD rats, and the expression of TLR4 and NF-κB increased further. After administration of TAK-242 (a specific TLR4 inhibitor), the visceral response to overexpression of Pellino-1 was reversed. Overall, the findings indicated the involvement of the FN in the development of CVH; the downregulation of Pellino-1 in the FN acted through the TLR4/NF-κB pathway to protect against CVH in a CRD rat model.
Collapse
Affiliation(s)
- Jiaojiao Wu
- Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Tao Li
- Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Guangtong Mao
- Department of Pathology, Xinyi People's Hospital, 16 Renmin Road, Xinyi 221400, Jiangsu Province, China
| | - Xiuli Cha
- Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Sujuan Fei
- Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China; Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China.
| | - Bei Miao
- Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China; Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China.
| |
Collapse
|
14
|
García-Martínez M, Cortez LM, Otero A, Betancor M, Serrano-Pérez B, Bolea R, Badiola JJ, Garza MC. Distinctive Toll-like Receptors Gene Expression and Glial Response in Different Brain Regions of Natural Scrapie. Int J Mol Sci 2022; 23:ijms23073579. [PMID: 35408945 PMCID: PMC8998348 DOI: 10.3390/ijms23073579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022] Open
Abstract
Prion diseases are chronic and fatal neurodegenerative diseases characterized by the accumulation of disease-specific prion protein (PrPSc), spongiform changes, neuronal loss, and gliosis. Growing evidence shows that the neuroinflammatory response is a key component of prion diseases and contributes to neurodegeneration. Toll-like receptors (TLRs) have been proposed as important mediators of innate immune responses triggered in the central nervous system in other human neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. However, little is known about the role of TLRs in prion diseases, and their involvement in the neuropathology of natural scrapie has not been studied. We assessed the gene expression of ovine TLRs in four anatomically distinct brain regions in natural scrapie-infected sheep and evaluated the possible correlations between gene expression and the pathological hallmarks of prion disease. We observed significant changes in TLR expression in scrapie-infected sheep that correlate with the degree of spongiosis, PrPSc deposition, and gliosis in each of the regions studied. Remarkably, TLR4 was the only gene upregulated in all regions, regardless of the severity of neuropathology. In the hippocampus, we observed milder neuropathology associated with a distinct TLR gene expression profile and the presence of a peculiar microglial morphology, called rod microglia, described here for the first time in the brain of scrapie-infected sheep. The concurrence of these features suggests partial neuroprotection of the hippocampus. Finally, a comparison of the findings in naturallyinfected sheep versus an ovinized mouse model (tg338 mice) revealed distinct patterns of TLRgene expression.
Collapse
Affiliation(s)
- Mirta García-Martínez
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, IA2, IIS Aragón, Universidad de Zaragoza, 50013 Zaragoza, Spain; (M.G.-M.); (M.B.); (R.B.); (J.J.B.)
| | - Leonardo M. Cortez
- Department of Medicine and Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Correspondence: (L.M.C.); (A.O.)
| | - Alicia Otero
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, IA2, IIS Aragón, Universidad de Zaragoza, 50013 Zaragoza, Spain; (M.G.-M.); (M.B.); (R.B.); (J.J.B.)
- Correspondence: (L.M.C.); (A.O.)
| | - Marina Betancor
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, IA2, IIS Aragón, Universidad de Zaragoza, 50013 Zaragoza, Spain; (M.G.-M.); (M.B.); (R.B.); (J.J.B.)
| | - Beatriz Serrano-Pérez
- Agrotecnio-CERCA Center, Department of Animal Science, University of Lleida, 25198 Lleida, Spain;
| | - Rosa Bolea
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, IA2, IIS Aragón, Universidad de Zaragoza, 50013 Zaragoza, Spain; (M.G.-M.); (M.B.); (R.B.); (J.J.B.)
| | - Juan J. Badiola
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, IA2, IIS Aragón, Universidad de Zaragoza, 50013 Zaragoza, Spain; (M.G.-M.); (M.B.); (R.B.); (J.J.B.)
| | - María Carmen Garza
- Departamento de Anatomía e Histología Humanas, IIS Aragón, Universidad de Zaragoza, 50009 Zaragoza, Spain;
| |
Collapse
|
15
|
Tian Y, Huang B, Li J, Tian X, Zeng X. Identification of the Association Between Toll-Like Receptors and T-Cell Activation in Takayasu’s Arteritis. Front Immunol 2022; 12:792901. [PMID: 35126357 PMCID: PMC8812403 DOI: 10.3389/fimmu.2021.792901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/22/2021] [Indexed: 12/26/2022] Open
Abstract
To explore the relationships between Toll-like receptors (TLRs) and the activation and differentiation of T-cells in Takayasu’s arteritis (TAK), using real-time fluorescence quantitative polymerase chain reaction, mRNA abundance of 29 target genes in peripheral blood mononuclear cells (PBMCs) were detected from 27 TAK patients and 10 healthy controls. Compared with the healthy control group, the untreated TAK group and the treated TAK group had an increased mRNA level of TLR2 and TLR4. A sample-to-sample matrix revealed that 80% of healthy controls could be separated from the TAK patients. Correlation analysis showed that the inactive-treated TAK group exhibited a unique pattern of inverse correlations between the TLRs gene clusters (including TLR1/2/4/6/8, BCL6, TIGIT, NR4A1, etc) and the gene cluster associated with T-cell activation and differentiation (including TCR, CD28, T-bet, GATA3, FOXP3, CCL5, etc). The dynamic gene co-expression network indicated the TAK groups had more active communication between TLRs and T-cell activation than healthy controls. BCL6, CCL5, FOXP3, GATA3, CD28, T-bet, TIGIT, IκBα, and NR4A1 were likely to have a close functional relation with TLRs at the inactive stage. The co-expression of TLR4 and TLR6 could serve as a biomarker of disease activity in treated TAK (the area under curve/sensitivity/specificity, 0.919/100%/90.9%). The largest gene co-expression cluster of the inactive-treated TAK group was associated with TLR signaling pathways, while the largest gene co-expression cluster of the active-treated TAK group was associated with the activation and differentiation of T-cells. The miRNA sequencing of the plasma exosomes combining miRDB, DIANA-TarBase, and miRTarBase databases suggested that the miR-548 family miR-584, miR-3613, and miR-335 might play an important role in the cross-talk between TLRs and T-cells at the inactive stage. This study found a novel relation between TLRs and T-cell in the pathogenesis of autoimmune diseases, proposed a new concept of TLR-co-expression signature which might distinguish different disease activity of TAK, and highlighted the miRNA of exosomes in TLR signaling pathway in TAK.
Collapse
Affiliation(s)
- Yixiao Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Biqing Huang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Jing Li, ; Xiaofeng Zeng,
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Peking Union Medical College Hospital (PUMCH), Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Jing Li, ; Xiaofeng Zeng,
| |
Collapse
|
16
|
Uniyal A, Tiwari V, Rani M, Tiwari V. Immune-microbiome interplay and its implications in neurodegenerative disorders. Metab Brain Dis 2022; 37:17-37. [PMID: 34357554 DOI: 10.1007/s11011-021-00807-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/22/2021] [Indexed: 12/28/2022]
Abstract
The neurodegeneration and its related CNS pathologies need an urgent toolbox to minimize the global mental health burden. The neuroimmune system critically regulates the brain maturation and survival of neurons across the nervous system. The chronic manipulated immunological drive can accelerate the neuronal pathology hence promoting the burden of neurodegenerative disorders. The gut is home for trillions of microorganisms having a mutual relationship with the host system. The gut-brain axis is a unique biochemical pathway through which the gut residing microbes connects with the brain cells and regulates various physiological and pathological cascades. The gut microbiota and CNS communicate using a common language that synchronizes the tuning of immune cells. The intestinal gut microbial community has a profound role in the maturation of the immune system as well as the development of the nervous system. We have critically summarised the clinical and preclinical reports from the past a decade emphasising that the significant changes in gut microbiota can enhance the host susceptibility towards neurodegenerative disorders. In this review, we have discussed how the gut microbiota-mediated immune response inclines the host physiology towards neurodegeneration and indicated the gut microbiota as a potential future candidate for the management of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ankit Uniyal
- Department of Pharmaceutical Engineering and Technology, Neuroscience and Pain Research Laboratory, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Vineeta Tiwari
- Department of Pharmaceutical Engineering and Technology, Neuroscience and Pain Research Laboratory, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Mousmi Rani
- Department of Pharmaceutical Engineering and Technology, Neuroscience and Pain Research Laboratory, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering and Technology, Neuroscience and Pain Research Laboratory, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
17
|
Toll-Like Receptors: General Molecular and Structural Biology. J Immunol Res 2021; 2021:9914854. [PMID: 34195298 PMCID: PMC8181103 DOI: 10.1155/2021/9914854] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background/Aim Toll-like receptors (TLRs) are pivotal biomolecules in the immune system. Today, we are all aware of the importance of TLRs in bridging innate and adaptive immune system to each other. The TLRs are activated through binding to damage/danger-associated molecular patterns (DAMPs), microbial/microbe-associated molecular patterns (MAMPs), pathogen-associated molecular patterns (PAMPs), and xenobiotic-associated molecular patterns (XAMPs). The immunogenetic molecules of TLRs have their own functions, structures, coreceptors, and ligands which make them unique. These properties of TLRs give us an opportunity to find out how we can employ this knowledge for ligand-drug discovery strategies to control TLRs functions and contribution, signaling pathways, and indirect activities. Hence, the authors of this paper have a deep observation on the molecular and structural biology of human TLRs (hTLRs). Methods and Materials To prepare this paper and fulfill our goals, different search engines (e.g., GOOGLE SCHOLAR), Databases (e.g., MEDLINE), and websites (e.g., SCOPUS) were recruited to search and find effective papers and investigations. To reach this purpose, we tried with papers published in the English language with no limitation in time. The iCite bibliometrics was exploited to check the quality of the collected publications. Results Each TLR molecule has its own molecular and structural biology, coreceptor(s), and abilities which make them unique or a complementary portion of the others. These immunogenetic molecules have remarkable roles and are much more important in different sections of immune and nonimmune systems rather than that we understand to date. Conclusion TLRs are suitable targets for ligand-drug discovery strategies to establish new therapeutics in the fields of infectious and autoimmune diseases, cancers, and other inflammatory diseases and disorders.
Collapse
|
18
|
Ioghen O, Chițoiu L, Gherghiceanu M, Ceafalan LC, Hinescu ME. CD36 - A novel molecular target in the neurovascular unit. Eur J Neurosci 2021; 53:2500-2510. [PMID: 33560561 PMCID: PMC8247892 DOI: 10.1111/ejn.15147] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
CD36 is an integral membrane protein primarily known for its function as a fatty acid transporter, yet also playing other biological roles from lipid metabolism to inflammation modulation. These pleiotropic effects are explained by the existence of multiple different ligands and the extensive distribution in numerous cell types. Moreover, the receptor is related to various pathologies and it may prove to be a good target for prospective therapeutic strategies. In the neurovascular unit (NVU), CD36 is expressed in cells like microglia, microvascular endothelial cells, astrocytes and neurons. In the normal brain, CD36 was proven to be involved in phagocytosis of apoptotic cells, oro‐sensory detection of dietary lipids, and fatty acid transport across the blood brain barrier (BBB). CD36 was also acknowledged as a potentially important player in central nervous system (CNS) disorders, such as Alzheimer Disease‐associated vascular dysfunction and oxidative stress and the neuroinflammatory response in stroke. Despite continuous efforts, the therapeutic arsenal for such diseases is still scarce and there is an increasing interest in discovering new molecular targets for more specific therapeutic approaches. In this review, we summarize the role of CD36 in the normal function of the NVU and in several CNS disorders, focusing on the dysregulation of the NVU and the potential therapeutic modulation.
Collapse
Affiliation(s)
- Octavian Ioghen
- Ultrastructural Pathology and Bioimaging Laboratory, Victor Babes Institute of Pathology, Bucharest, Romania
| | - Leona Chițoiu
- Ultrastructural Pathology and Bioimaging Laboratory, Victor Babes Institute of Pathology, Bucharest, Romania
| | - Mihaela Gherghiceanu
- Ultrastructural Pathology and Bioimaging Laboratory, Victor Babes Institute of Pathology, Bucharest, Romania.,Department of Cellular and Molecular Biology and Histology, School of Medicine, Carol Davila Faculty of Medicine, Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, School of Medicine, Carol Davila Faculty of Medicine, Bucharest, Romania.,Cell Biology, Neurosciences and Experimental Myology Laboratory, Victor Babes Institute of Pathology, Bucharest, Romania
| | - Mihail Eugen Hinescu
- Department of Cellular and Molecular Biology and Histology, School of Medicine, Carol Davila Faculty of Medicine, Bucharest, Romania.,Cell Biology, Neurosciences and Experimental Myology Laboratory, Victor Babes Institute of Pathology, Bucharest, Romania
| |
Collapse
|
19
|
Simons ES, Smith MA, Dengler-Crish CM, Crish SD. Retinal ganglion cell loss and gliosis in the retinofugal projection following intravitreal exposure to amyloid-beta. Neurobiol Dis 2021; 147:105146. [PMID: 33122075 DOI: 10.1016/j.nbd.2020.105146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
Pathological accumulations of amyloid-beta (Aβ) peptide are found in retina early in Alzheimer's disease, yet its effects on retinal neuronal structure remain unknown. To investigate this, we injected fibrillized Aβ1-42 protein into the eye of adult C57BL/6 J mice and analyzed the retina, optic nerve (ON), and the superior colliculus (SC), the primary retinal target in mice. We found that retinal Aβ exposure stimulated microglial activation and retinal ganglion cell (RGC) loss as early as 1-week post-injection. Pathology was not limited to the retina, but propagated into other areas of the central nervous system. Microgliosis spread throughout the retinal projection (retina, ON, and SC), with multiplex protein quantitation demonstrating an increase in endogenously produced Aβ in the ON and SC corresponding to the injected retinas. Surprisingly, this pathology spread to the opposite side, with unilateral Aβ eye injections driving increased Aβ levels, neuroinflammation, and RGC death in the opposite, un-injected retinal projection. As Aβ-mediated microglial activation has been shown to propagate Aβ pathology, we also investigated the role of the Aβ-binding microglial scavenger receptor CD36 in this pathology. Transgenic mice lacking the CD36 receptor were resistant to Aβ-induced inflammation and RGC death up to 2 weeks following exposure. These results indicate that Aβ pathology drives regional neuropathology in the retina and does not remain isolated to the affected eye, but spreads throughout the nervous system. Further, CD36 may serve as a promising target to prevent Aβ-mediated inflammatory damage.
Collapse
Affiliation(s)
- E S Simons
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - M A Smith
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States; Akron Children's Hospital, Rebecca D. Considine Research Institute, Akron, OH 44302, United States
| | - C M Dengler-Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - S D Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States.
| |
Collapse
|
20
|
Dobri AM, Dudău M, Enciu AM, Hinescu ME. CD36 in Alzheimer's Disease: An Overview of Molecular Mechanisms and Therapeutic Targeting. Neuroscience 2020; 453:301-311. [PMID: 33212223 DOI: 10.1016/j.neuroscience.2020.11.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/17/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
CD36 is a membrane protein with wide distribution in the human body, is enriched in the monocyte-macrophage system and endothelial cells, and is involved in the cellular uptake of long chain fatty acids (LCFA) and oxidized low-density lipoproteins. It is also a scavenger receptor, binding hydrophobic amyloid fibrils found in the Alzheimer's disease (AD) brain. In neurobiology research, it has been mostly studied in relationship with chronic ischemia and stroke, but it was also related to amyloid clearance by microglial phagocytosis. In AD animal models, amyloid binding to CD36 has been consistently correlated with a pro-inflammatory response. Therapeutic approaches have two main focuses: CD36 blockade with monoclonal antibodies or small molecules, which is beneficial in terms of the inflammatory milieu, and upregulation of CD36 for increased amyloid clearance. The balance of the two approaches, centered on microglia, is poorly understood. Furthermore, CD36 evaluation in AD clinical studies is still at a very early stage and there is a gap in the knowledge regarding the impact of LCFA on AD progression and CD36 expression and genetic phenotype. This review summarizes the role played by CD36 in the pathogenic amyloid cascade and explore the translatability of preclinical data towards clinical research.
Collapse
Affiliation(s)
- Ana-Maria Dobri
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania.
| | - Maria Dudău
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania.
| | - Ana-Maria Enciu
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania.
| | - Mihail Eugen Hinescu
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; "Carol Davila" University of Medicine and Pharmacy, 5 Eroilor Sanitari Blvd, 050047 Bucharest, Romania
| |
Collapse
|
21
|
Toll-like receptors in Alzheimer's disease. J Neuroimmunol 2020; 348:577362. [DOI: 10.1016/j.jneuroim.2020.577362] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/04/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023]
|
22
|
Narayanankutty A. Toll-like Receptors as a Novel Therapeutic Target for Natural Products Against Chronic Diseases. Curr Drug Targets 2020; 20:1068-1080. [PMID: 30806312 DOI: 10.2174/1389450120666190222181506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/25/2019] [Accepted: 02/14/2019] [Indexed: 02/08/2023]
Abstract
Toll-like receptors (TLR) are one among the initial responders of the immune system which participate in the activation inflammatory processes. Several different types of TLR such as TLR2, TLR4, TLR7 and TLR9 have been identified in various cell types, each having distinct ligands like lipids, lipoproteins, nucleic acids and proteins. Though its prime concern is xenobiotic defences, TLR signalling has also recognized as an activator of inflammation and associated development of chronic degenerative disorders (CDDs) including obesity, type 2 diabetes mellitus (T2DM), fatty liver disease, cardiovascular and neurodegenerative disorders as well as various types of cancers. Numerous drugs are in use to prevent these disorders, which specifically inhibit different pathways associated with the development of CDDs. Compared to these drug targets, inhibition of TLR, which specifically responsible for the inflammatory insults has proven to be a better drug target. Several natural products have emerged as inhibitors of CDDs, which specifically targets TLR signalling, among these, many are in the clinical trials. This review is intended to summarize the recent progress on TLR association with CDDs and to list possible use of natural products, their combinations and their synthetic derivative in the prevention of TLR-driven CDD development.
Collapse
Affiliation(s)
- Arunaksharan Narayanankutty
- Post Graduate & Research Department of Zoology, St. Joseph's College (Autonomous), Devagiri, Calicut, Kerala, 680 555, India
| |
Collapse
|
23
|
Wang Y, Zhang S, Li H, Wang H, Zhang T, Hutchinson MR, Yin H, Wang X. Small-Molecule Modulators of Toll-like Receptors. Acc Chem Res 2020; 53:1046-1055. [PMID: 32233400 DOI: 10.1021/acs.accounts.9b00631] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are the "gatekeepers" of the immune system in humans and other animals to protect the host from invading bacteria, viruses, and other microorganisms. Since TLR4 was discovered as the receptor for endotoxin in the late 1990s, significant progress has been made in exploiting an understanding of the function of TLRs. The TLR-signaling pathway is crucial for the induction and progression of various diseases. Dysregulation of TLR signaling contributes to numerous pathological conditions, including chronic inflammation, sepsis, cancers, asthma, neuropathic pain, drug addiction, and autoimmune diseases. Therefore, manipulation of TLR signaling is promising to halt their activity in inflammatory diseases, to enhance their signaling to fight cancers, to modulate their role in autoimmune diseases, and to suppress them to treat drug addiction. TLR agonists have demonstrated great potential as antimicrobial agents and vaccine adjuvants, whereas TLR antagonists are being developed as reagents and drugs to dampen immune responses. Because of their pivotal potential therapeutic applications, fruitful small-molecule compounds and peptide fragments have been discovered, and many of them have advanced to various stages of clinical trials (though only two have been approved by the Food and Drug Administration (FDA): MPLA as a TLR4 agonist and imiquimod as a TLR7 agonist).In this Account, we focus on the progress in developing TLR signaling pathway modulators (mainly focused on the Yin and Wang laboratories) over the past decade and highlight the accomplishments and currently existing challenges in the development of TLR modulators. First, we briefly describe the members of the human TLR family along with their natural modulators. Second, we illustrate our endeavors to discover TLR-targeted agents using comprehensive approaches. Specifically, a discussion of identification and characterization of new chemical entities, determination of modes of action, and further applications is presented. For instance, the TLR3 antagonist was first discovered through in silico screening, and the inhibitory activity was confirmed in murine cells. Considering the glycosylation on TLR3, a new direction for TLR3 modulator design was pointed out to target asparagine glycosylation. We have particularly focused on the discovery of TLR4 antagonists and have assessed their great potential in the clinical treatment of drug addiction and alcohol use disorders. In addition, we discuss multiple other popular and robust techniques for modulator discovery. Not only small organic modulators but also stapled peptides and peptidomimetics will attract more and more attention in the future. Finally, current challenges, opportunities, and future perspectives for TLR-targeted agents are also discussed.
Collapse
Affiliation(s)
- Yibo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Shuting Zhang
- School of Pharmaceutical Sciences, Tsinghua University−Peking University Joint Center for Life Sciences, Tsinghua University, Beijing 100082, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Tianshu Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Mark R. Hutchinson
- Discipline of Physiology, Adelaide Medical School, and ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University−Peking University Joint Center for Life Sciences, Tsinghua University, Beijing 100082, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
24
|
Miller YI, Navia-Pelaez JM, Corr M, Yaksh TL. Lipid rafts in glial cells: role in neuroinflammation and pain processing. J Lipid Res 2020; 61:655-666. [PMID: 31862695 PMCID: PMC7193960 DOI: 10.1194/jlr.tr119000468] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/06/2019] [Indexed: 12/27/2022] Open
Abstract
Activation of microglia and astrocytes secondary to inflammatory processes contributes to the development and perpetuation of pain with a neuropathic phenotype. This pain state presents as a chronic debilitating condition and affects a large population of patients with conditions like rheumatoid arthritis and diabetes, or after surgery, trauma, or chemotherapy. Here, we review the regulation of lipid rafts in glial cells and the role they play as a key component of neuroinflammatory sensitization of central pain signaling pathways. In this context, we introduce the concept of an inflammaraft (i-raft), enlarged lipid rafts harboring activated receptors and adaptor molecules and serving as an organizing platform to initiate inflammatory signaling and the cellular response. Characteristics of the inflammaraft include increased relative abundance of lipid rafts in inflammatory cells, increased content of cholesterol per raft, and increased levels of inflammatory receptors, such as toll-like receptor (TLR)4, adaptor molecules, ion channels, and enzymes in lipid rafts. This inflammaraft motif serves an important role in the membrane assembly of protein complexes, for example, TLR4 dimerization. Operating within this framework, we demonstrate the involvement of inflammatory receptors, redox molecules, and ion channels in the inflammaraft formation and the regulation of cholesterol and sphingolipid metabolism in the inflammaraft maintenance and disruption. Strategies for targeting inflammarafts, without affecting the integrity of lipid rafts in noninflammatory cells, may lead to developing novel therapies for neuropathic pain states and other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Yury I Miller
- Departments of MedicineUniversity of California San Diego, La Jolla, CA. mailto:
| | | | - Maripat Corr
- Departments of MedicineUniversity of California San Diego, La Jolla, CA
| | - Tony L Yaksh
- Anesthesiology,University of California San Diego, La Jolla, CA
| |
Collapse
|
25
|
Gross-Vered M, Shmuel-Galia L, Zarmi B, Humphries F, Thaiss C, Salame TM, David E, Chappell-Maor L, Fitzgerald KA, Shai Y, Jung S. TLR2 Dimerization Blockade Allows Generation of Homeostatic Intestinal Macrophages under Acute Colitis Challenge. THE JOURNAL OF IMMUNOLOGY 2020; 204:707-717. [PMID: 31882517 DOI: 10.4049/jimmunol.1900470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 11/16/2019] [Indexed: 11/19/2022]
Abstract
Recruited blood monocytes contribute to the establishment, perpetuation, and resolution of tissue inflammation. Specifically, in the inflamed intestine, monocyte ablation was shown to ameliorate colitis scores in preclinical animal models. However, the majority of intestinal macrophages that seed the healthy gut are also monocyte derived. Monocyte ablation aimed to curb inflammation would therefore likely interfere with intestinal homeostasis. In this study, we used a TLR2 trans-membrane peptide that blocks TLR2 dimerization that is critical for TLR2/1 and TLR2/6 heterodimer signaling to blunt inflammation in a murine colitis model. We show that although the TLR2 peptide treatment ameliorated colitis, it allowed recruited monocytes to give rise to macrophages that lack the detrimental proinflammatory gene signature and reduced potentially damaging neutrophil infiltrates. Finally, we demonstrate TLR blocking activity of the peptide on in vitro cultured human monocyte-derived macrophages. Collectively, we provide a significantly improved anti-inflammatory TLR2 peptide and critical insights in its mechanism of action toward future potential use in the clinic.
Collapse
Affiliation(s)
- Mor Gross-Vered
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liraz Shmuel-Galia
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Batya Zarmi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Fiachra Humphries
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Christoph Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tomer-Meir Salame
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Katherine A Fitzgerald
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Yechiel Shai
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
26
|
Review on Cross Talk between Neurotransmitters and Neuroinflammation in Striatum and Cerebellum in the Mediation of Motor Behaviour. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1767203. [PMID: 31815123 PMCID: PMC6877979 DOI: 10.1155/2019/1767203] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/06/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023]
Abstract
Neurological diseases particularly Alzheimer's disease (AD), Parkinson's disease (PD), stroke, and epilepsy are on the rise all around the world causing morbidity and mortality globally with a common symptom of gradual loss or impairment of motor behaviour. Striatum, which is a component of the basal ganglia, is involved in facilitating voluntary movement while the cerebellum is involved in the maintenance of balance and coordination of voluntary movements. Dopamine, serotonin, gamma-aminobutyric acid (GABA), and glutamate, to name a few, interact in regulating the excitation and inhibition of motor neurons. In another hand, interestingly, the motor loss associated with neurological diseases is possibly resulted from neuroinflammation induced by the neuroimmune system. Toll-like receptors (TLRs) are present in the central nervous system (CNS), specifically and primarily expressed in microglia and are also found on neurons and astrocytes, functioning mainly in the regulation of proinflammatory cytokine production. TLRs are always found to be associated or involved in the induction of neuroinflammation in neurodegenerative diseases. Activation of toll-like receptor 4 (TLR4) through TLR4 agonist, lipopolysaccharide (LPS), stimulation initiate a signaling cascade whereby the TLR4-LPS interaction has been found to result in physiological and behavioural changes including retardation of motor activity in the mouse model. TLR4 inhibitor TAK-242 was reflected in the reduction of the spinal cord pathology along with the motor improvement in ALS mouse. There is cross talk with neuroinflammation and neurochemicals. For example, TLR4 activation by LPS is noted to release proinflammatory cytokines, IL-1β, from microglia that subsequently suppresses GABA receptor activities at the postsynaptic site and reduces GABA synthesis at the presynaptic site. Glial glutamate transporter activities are also found to be suppressed, showing the association between TLR4 activation and the related neurotransmitters and corresponding receptors and transporters in the event of neuroinflammation. This review is helpful to understand the connection between neurotransmitter and neuroinflammation in striatum- and cerebellum-mediated motor behaviour.
Collapse
|
27
|
Aurelian L, Balan I. GABA AR α2-activated neuroimmune signal controls binge drinking and impulsivity through regulation of the CCL2/CX3CL1 balance. Psychopharmacology (Berl) 2019; 236:3023-3043. [PMID: 31030249 DOI: 10.1007/s00213-019-05220-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Toll-like receptors (TLRs) are a family of innate immune system receptors that respond to pathogen-derived and tissue damage-related ligands and are increasingly recognized for their impact on homeostasis and its dysregulation in the nervous system. TLR signaling participates in brain injury and addiction, but its role in the alcohol-seeking behavior, which initiates alcohol drinking, is still poorly understood. In this review, we discuss our findings designed to elucidate the potential contribution of the activated TLR4 signal located in neurons, on impulsivity and the predisposition to initiate alcohol drinking (binge drinking). RESULTS Our findings indicate that the TLR4 signal is innately activated in neurons from alcohol-preferring subjects, identifying a genetic contribution to the regulation of impulsivity and the alcohol-seeking propensity. Signal activation is through the non-canonical, previously unknown, binding of TLR4 to the α2 subunit of the γ-aminobutyric 2 acid A receptor (GABAAR α2). Activation is sustained by the stress hormone corticotrophin-releasing factor (CRF) and additional still poorly recognized ligand/scaffold proteins. Focus is on the effect of TLR4 signal activation on the balance between pro- and anti-inflammatory chemokines [chemokine (C-C motif) ligand 2 (CCL2)/chemokine (C-X3-C motif) ligand 1 (CX3CL1)] and its effect on binge drinking. CONCLUSION The results are discussed within the context of current findings on the distinct activation and functions of TLR signals located in neurons, as opposed to immune cells. They indicate that the balance between pro- and anti-inflammatory TLR4 signaling plays a major role in binge drinking. These findings have major impact on future basic and translational research, including the development of potential therapeutic and preventative strategies.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Stanford University School of Medicine OFDD, Stanford, CA, 94305, USA.
| | - Irina Balan
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Department of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
28
|
Cell Type Specific Expression of Toll-Like Receptors in Human Brains and Implications in Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7420189. [PMID: 31396533 PMCID: PMC6668540 DOI: 10.1155/2019/7420189] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/07/2019] [Indexed: 12/20/2022]
Abstract
Toll-like receptors mediate important cellular immune responses upon activation via various pathogenic stimuli such as bacterial or viral components. The activation and subsequent secretion of cytokines and proinflammatory factors occurs in the whole body including the brain. The subsequent inflammatory response is crucial for the immune system to clear the pathogen(s) from the body via the innate and adaptive immune response. Within the brain, astrocytes, neurons, microglia, and oligodendrocytes all bear unique compositions of Toll-like receptors. Besides pathogens, cellular damage and abnormally folded protein aggregates, such as tau and Amyloid beta peptides, have been shown to activate Toll-like receptors in neurodegenerative diseases such as Alzheimer's disease. This review provides an overview of the different cell type-specific Toll-like receptors of the human brain, their activation mode, and subsequent cellular response, as well as their activation in Alzheimer's disease. Finally, we critically evaluate the therapeutic potential of targeting Toll-like receptors for treatment of Alzheimer's disease as well as discussing the limitation of mouse models in understanding Toll-like receptor function in general and in Alzheimer's disease.
Collapse
|
29
|
Alzheimer's disease: Key developments support promising perspectives for therapy. Pharmacol Res 2019; 146:104316. [PMID: 31260730 DOI: 10.1016/j.phrs.2019.104316] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/15/2019] [Accepted: 06/15/2019] [Indexed: 12/13/2022]
Abstract
Alzheimer's is the neurodegenerative disease affecting the largest number of patients in the world. In spite of the intense research of the last decades, progress about its knowledge and therapy was limited. In particular, various cytotoxic processes remained debated, while the few drugs approved for therapy were of only marginal relevance. Recent studies have identified key aspects of the disease, such as the mechanisms governing the development of pathology. In order to operate the Aβ peptide, known as the key factor, requires a complex assembled by its high affinity binding to PrPc, a cell surface prion protein, and mGluR5, a metabotropic glutamate receptor. Aβ and its associates bind also phosphorylated tau transferred to the extracellular space, with final activation of intracellular cytotoxic signals. Pathology is further affected by factors (including genes, receptors and their agonists) and by glial cells governing (via vesicles, cytokines and enzymes) cell immunology, inflammation and oxidative stress. Concomitant to pathology studies, strong attempts have been made for the development of new, effective therapies. Critical for this are biomarkers, by which Alzheimer's patients are recognized even before appearance of their symptoms. The question was whether patients take advantage from drugs not yet approved. The latter, first identified in mice, were found effective also in men, however only before appearance or at early stage of the disease. In other words, the drugs not yet approved induce effective protection of patients still healthy or in a preliminary stage of the disease. In contrast, developed Alzheimer's disease is practically irreversible.
Collapse
|
30
|
Colabufo NA, Contino M, Cantore M, Berardi F, Perrone R, Tonazzi A, Console L, Panaro MA, Savolainen H, Luurtsema G. An innovative small molecule for promoting neuroreparative strategies. RSC Adv 2018; 8:5451-5458. [PMID: 35542423 PMCID: PMC9078125 DOI: 10.1039/c7ra11812k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/22/2018] [Indexed: 11/21/2022] Open
Abstract
In this study, a new regenerative strategy to treat several neurodegenerative diseases is suggested by the use of a multitarget approach induced by our small molecule, MC111. Considering the importance of P-gp and BCRP expression on stem cell differentiation and the involvement of TLR4 on neurodegeneration processes, we investigated the effect of MC111, belonging to our library of P-gp active compounds on: (i) TLR4 signaling; (ii) P-gp and BCRP activity and expression; (iii) neurite sprouting. The observed findings exerted by MC111, open a new scenario for a multitarget and regenerative approach in neurodegenerative diseases encouraging the in vivo evaluation of MC111 as new tool in neuroreparative medicine. In this study, a new regenerative strategy to treat several neurodegenerative diseases is suggested by the use of a multitarget approach induced by our small molecule, MC111.![]()
Collapse
Affiliation(s)
- Nicola Antonio Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco
- University of Bari “Aldo Moro”
- Bari
- Italy
- Biofordrug srl
| | | | - Mariangela Cantore
- Biofordrug srl
- Dipartimento di Farmacia-Scienze del Farmaco
- University of Bari “Aldo Moro”
- Bari
- Italy
| | - Francesco Berardi
- Dipartimento di Farmacia-Scienze del Farmaco
- University of Bari “Aldo Moro”
- Bari
- Italy
| | - Roberto Perrone
- Dipartimento di Farmacia-Scienze del Farmaco
- University of Bari “Aldo Moro”
- Bari
- Italy
| | - Annamaria Tonazzi
- CNR-IBIOM (Institute of Biomembrane, Bioenergetics and Molecular Biotechnologies)
- 70126 Bari
- Italy
- Dipartimento di Bioscienze
- Biotecnologie e Biofarmaceutica
| | - Lara Console
- Department BEST (Biologia, Ecologia, Scienze della Terra)
- Unit of Biochemistry and Molecular Biotechnology
- University of Calabria
- Italy
| | - Maria Antonietta Panaro
- Dipartimento di Bioscienze
- Biotecnologie e Biofarmaceutica
- University of Bari “Aldo Moro”
- Bari
- Italy
| | - Heli Savolainen
- University of Groningen
- University Medical Center Groningen
- Department of Nuclear Medicine and Molecular Imaging
- Groningen
- Netherlands
| | - Gert Luurtsema
- University of Groningen
- University Medical Center Groningen
- Department of Nuclear Medicine and Molecular Imaging
- Groningen
- Netherlands
| |
Collapse
|