1
|
Geiduschek EK, McDowell CM. The Fibro-Inflammatory Response in the Glaucomatous Optic Nerve Head. Int J Mol Sci 2023; 24:13240. [PMID: 37686046 PMCID: PMC10487997 DOI: 10.3390/ijms241713240] [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: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Glaucoma is a progressive disease and the leading cause of irreversible blindness. The limited therapeutics available are only able to manage the common risk factor of glaucoma, elevated intraocular pressure (IOP), indicating a great need for understanding the cellular mechanisms behind optic nerve head (ONH) damage during disease progression. Here we review the known inflammatory and fibrotic changes occurring in the ONH. In addition, we describe a novel mechanism of toll-like receptor 4 (TLR4) and transforming growth factor beta-2 (TGFβ2) signaling crosstalk in the cells of the ONH that contribute to glaucomatous damage. Understanding molecular signaling within and between the cells of the ONH can help identify new drug targets and therapeutics.
Collapse
Affiliation(s)
| | - Colleen M. McDowell
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
2
|
Tahmasebi F, Barati S. The Role of Microglial Depletion Approaches in Pathological Condition of CNS. Cell Mol Neurobiol 2023; 43:2459-2471. [PMID: 36738403 PMCID: PMC11410134 DOI: 10.1007/s10571-023-01326-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Microglia are the primary immune cells of the central nervous system (CNS) that comprise about 5-12% of all cells in the brain. These cells are the first line of defense that protects the CNS from damage and attacking pathogens. Microglia originate from yolk sac macrophages and migrate to the brain before the blood-brain barrier formation. Microglia show key roles in healthy CNS including promoting neurogenesis, synaptic sculpting, and maintaining homeostasis but in pathological conditions of CNS, microglial activation may exacerbate diseases. Thus, microglial depletion of the CNS is a novel approach that could be a useful tool to understand the microglial functions in neurodegenerative and neuroinflammatory diseases. There are methods for microglial ablation and reduction such as genetic tools and pharmacological inhibitors. In this study, we review recent studies that used different microglial ablation models for microglial reduction and repopulation after depletion in pathological states of CNS. Recently, studies showed that microglial depletion as a potential therapeutic application has benefits (such as inflammatory factors reduction, increase synaptogenesis, astrogliosis preventation) in CNS. For these reasons, the inhibition of microglia with these models was considered a therapeutic approach for neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
3
|
Primary Cortical Cell Tri-Culture-Based Screening of Neuroinflammatory Response in Toll-like Receptor Activation. Biomedicines 2022; 10:biomedicines10092122. [PMID: 36140221 PMCID: PMC9495748 DOI: 10.3390/biomedicines10092122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
The activation of toll-like receptors (TLRs) in the central nervous system (CNS) can lead to neuroinflammation and contribute to many neurological disorders, including autoimmune diseases. Cell culture models are powerful tools for studying specific molecular and cellular mechanisms that contribute to these disease states and identifying potential therapeutics. However, most cell culture models have limitations in capturing biologically relevant phenomena, due in part to the non-inclusion of necessary cell types. Neurons, astrocytes, and microglia (critical cell types that play a role in neuroinflammation) all express at least a subset of TLRs. However, the response of each of these cell types to various TLR activation, along with their relative contribution to neuroinflammatory processes, is far from clear. In this study, we demonstrate the screening capabilities of a primary cortical cell tri-culture of neuron, astrocyte, and microglia from neonatal rats. Specifically, we compare the neuroinflammatory response of tri-cultures to that of primary neuron-astrocyte co-cultures to a suite of known TLR agonists. We demonstrate that microglia are required for observation of neurotoxic neuroinflammatory responses, such as increased cell death and apoptosis, in response to TLR2, 3, 4, and 7/8 activation. Additionally, we show that following TLR3 agonist treatment, microglia and astrocytes play opposing roles in the neuroinflammatory response, and that the observed response is dictated by the degree of TLR3 activation. Overall, we demonstrate that microglia play a significant role in the neuroinflammatory response to TLR activation in vitro and, hence, the tri-culture has the potential to serve as a screening platform that better replicates the in vivo responses.
Collapse
|
4
|
Geiduschek EK, Milne PD, Mzyk P, Mavlyutov TA, McDowell CM. TLR4 signaling modulates extracellular matrix production in the lamina cribrosa. FRONTIERS IN OPHTHALMOLOGY 2022; 2:968381. [PMID: 36911656 PMCID: PMC9997209 DOI: 10.3389/fopht.2022.968381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The optic nerve head (ONH) is a place of vulnerability during glaucoma progression due to increased intraocular pressure damaging the retinal ganglion cell axons. The molecular signaling pathways involved in generating glaucomatous ONH damage has not been fully elucidated. There is a great deal of evidence that pro-fibrotic TGFβ2 signaling is involved in modulating the ECM environment within the lamina cribrosa (LC) region of the ONH. Here we investigated the role of signaling crosstalk between the TGFβ2 pathway and the toll-like receptor 4 (TLR4) pathway within the LC. ECM deposition was examined between healthy and glaucomatous human ONH sections, finding increases in fibronectin and fibronectin extra domain A (FN-EDA) an isoform of fibronectin known to be a damage associated molecular pattern (DAMP) that can activate TLR4 signaling. In human LC cell cultures derived from healthy donor eyes, inhibition of TLR4 signaling blocked TGFβ2 induced FN and FN-EDA expression. Activation of TLR4 by cellular FN (cFN) containing the EDA isoform increased both total FN production and Collagen-1 production and this effect was dependent on TLR4 signaling. These studies identify TGFβ2-TLR4 signaling crosstalk in LC cells of the ONH as a novel pathway regulating ECM and DAMP production.
Collapse
Affiliation(s)
- Emma K Geiduschek
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Paige D Milne
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Philip Mzyk
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Timur A Mavlyutov
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Colleen M McDowell
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
5
|
Ghosh P, Singh R, Ganeshpurkar A, Pokle AV, Singh RB, Singh SK, Kumar A. Cellular and molecular influencers of neuroinflammation in Alzheimer's disease: Recent concepts & roles. Neurochem Int 2021; 151:105212. [PMID: 34656693 DOI: 10.1016/j.neuint.2021.105212] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/22/2021] [Accepted: 10/10/2021] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD), an extremely common neurodegenerative disorder of the older generation, is one of the leading causes of death globally. Besides the conventional hallmarks i.e. Amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs), neuroinflammation also serves as a major contributing factor in the pathogenesis of AD. There are mounting evidences to support the fundamental role of cellular (microglia, astrocytes, mast cells, and T-cells) and molecular (cytokines, chemokines, caspases, and complement proteins) influencers of neuroinflammation in producing/promoting neurodegeneration and dementia in AD. Genome-wide association studies (GWAS) have revealed the involvement of various single nucleotide polymorphisms (SNPs) of genes related to neuroinflammation with the risk of developing AD. Modulating the release of the neuroinflammatory molecules and targeting their relevant mechanisms may have beneficial effects on the onset, progress and severity of the disease. Here, we review the distinct role of various mediators and modulators of neuroinflammation that impact the pathogenesis and progression of AD as well as incite further research efforts for the treatment of AD through a neuroinflammatory approach.
Collapse
Affiliation(s)
- Powsali Ghosh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Vyankatrao Pokle
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Bhushan Singh
- Institute of Pharmacy Harischandra PG College, Bawanbigha, Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
6
|
Chaudhary R, Morris RJ, Steinson E. The multifactorial roles of microglia and macrophages in the maintenance and progression of glioblastoma. J Neuroimmunol 2021; 357:577633. [PMID: 34153803 DOI: 10.1016/j.jneuroim.2021.577633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/26/2021] [Accepted: 06/11/2021] [Indexed: 01/18/2023]
Abstract
The functional characteristics of glial cells, in particular microglia, have attained considerable importance in several diseases, including glioblastoma, the most hostile and malignant type of intracranial tumor. Microglia performs a highly significant role in the brain's inflammatory response mechanism. They exhibit anti-tumor properties via phagocytosis and the activation of a number of different cytotoxic substances. Some tumor-derived factors, however, transform these microglial cells into immunosuppressive and tumor-supportive, facilitating survival and progression of tumorigenic cells. Glioma-associated microglia and/or macrophages (GAMs) accounts for a large proportion of glioma infiltrating cells. Once within the tumor, GAMs exhibit a distinct phenotype of initiation that subsequently supports the growth and development of tumorigenic cells, angiogenesis and stimulates the infiltration of healthy brain regions. Interventions that suppress or prohibit the induction of GAMs at the tumor site or attenuate their immunological activities accommodating anti-tumor actions are likely to exert positive impact on glioblastoma treatment. In the present paper, we aim to summarize the most recent knowledge of microglia and its physiology, as well as include a very brief description of different molecular factors involved in microglia and glioblastoma interplay. We further address some of the major signaling pathways that regulate the baseline motility of glioblastoma progression. Finally, we discussed a number of therapeutic approaches regarding glioblastoma treatment.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, India.
| | - Rhianna J Morris
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Emma Steinson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
7
|
Corsi-Zuelli F, Deakin B. Impaired regulatory T cell control of astroglial overdrive and microglial pruning in schizophrenia. Neurosci Biobehav Rev 2021; 125:637-653. [PMID: 33713699 DOI: 10.1016/j.neubiorev.2021.03.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/16/2021] [Accepted: 03/06/2021] [Indexed: 02/07/2023]
Abstract
It is widely held that schizophrenia involves an active process of peripheral inflammation that induces or reflects brain inflammation with activation of microglia, the brain's resident immune cells. However, recent in vivo radioligand binding studies and large-scale transcriptomics in post-mortem brain report reduced markers of microglial inflammation. The findings suggest a contrary hypothesis; that microglia are diverted into their non-inflammatory synaptic remodelling phenotype that interferes with neurodevelopment and perhaps contributes to the relapsing nature of schizophrenia. Recent discoveries on the regulatory interactions between micro- and astroglial cells and immune regulatory T cells (Tregs) cohere with clinical omics data to suggest that: i) disinhibited astrocytes mediate the shift in microglial phenotype via the production of transforming growth factor-beta, which also contributes to the disturbances of dopamine and GABA function in schizophrenia, and ii) systemically impaired functioning of Treg cells contributes to the dysregulation of glial function, the low-grade peripheral inflammation, and the hitherto unexplained predisposition to auto-immunity and reduced life-expectancy in schizophrenia, including greater COVID-19 mortality.
Collapse
Affiliation(s)
- Fabiana Corsi-Zuelli
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Ribeirão Preto, São Paulo, Brazil
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
8
|
López-Pérez Ó, Bernal-Martín M, Hernaiz A, Llorens F, Betancor M, Otero A, Toivonen JM, Zaragoza P, Zerr I, Badiola JJ, Bolea R, Martín-Burriel I. BAMBI and CHGA in Prion Diseases: Neuropathological Assessment and Potential Role as Disease Biomarkers. Biomolecules 2020; 10:biom10050706. [PMID: 32370154 PMCID: PMC7277700 DOI: 10.3390/biom10050706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 12/14/2022] Open
Abstract
Prion diseases affect both animals and humans. Research in the natural animal model of the disease could help in the understanding of neuropathological mechanisms and in the development of biomarkers for human pathologies. For this purpose, we studied the expression of 10 genes involved in prion propagation in vitro in the central nervous system of scrapie-infected sheep. Dysregulated genes (BAMBI and CHGA) were further analysed in a transgenic murine model (Tg338) of scrapie, and their protein distribution was determined using immunohistochemistry and Western blot. Their potential as biomarkers was finally assessed using enzyme-linked immunosorbent assay (ELISA) in cerebrospinal fluid (CSF) of scrapie sheep and Creutzfeldt-Jakob disease (CJD) patients. Protein BAMBI was upregulated in highly affected brain areas and CHGA was overexpressed along the brain in both models. Moreover, BAMBI and CHGA immunostaining scores strongly correlated with spongiosis and microgliosis in mice. Finally, levels of BAMBI were significantly higher in the CSF of clinical sheep and CJD patients. In addition to their potential as biomarkers, our work confirms the role of BAMBI and CHGA in prion neuropathology in vivo, but besides prion replication, they seem to be involved in the characteristic neuroinflammatory response associated to prion infection.
Collapse
Affiliation(s)
- Óscar López-Pérez
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (Ó.L.-P.); (M.B.-M.); (A.H.); (J.M.T.); (P.Z.)
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (M.B.); (A.O.); (J.J.B.); (R.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Marcos Bernal-Martín
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (Ó.L.-P.); (M.B.-M.); (A.H.); (J.M.T.); (P.Z.)
| | - Adelaida Hernaiz
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (Ó.L.-P.); (M.B.-M.); (A.H.); (J.M.T.); (P.Z.)
| | - Franc Llorens
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, 37075 Göttingen, Germany;
| | - Marina Betancor
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (M.B.); (A.O.); (J.J.B.); (R.B.)
| | - Alicia Otero
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (M.B.); (A.O.); (J.J.B.); (R.B.)
- Department of Biological Sciences, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Janne Markus Toivonen
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (Ó.L.-P.); (M.B.-M.); (A.H.); (J.M.T.); (P.Z.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, 28029 Madrid, Spain;
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (Ó.L.-P.); (M.B.-M.); (A.H.); (J.M.T.); (P.Z.)
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, 37075 Göttingen, Germany;
| | - Juan José Badiola
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (M.B.); (A.O.); (J.J.B.); (R.B.)
| | - Rosa Bolea
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (M.B.); (A.O.); (J.J.B.); (R.B.)
| | - Inmaculada Martín-Burriel
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (Ó.L.-P.); (M.B.-M.); (A.H.); (J.M.T.); (P.Z.)
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Universidad de Zaragoza, Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, 50013 Zaragoza, Spain; (M.B.); (A.O.); (J.J.B.); (R.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, 28029 Madrid, Spain;
- Correspondence: ; Tel.: +34-653-638-749
| |
Collapse
|
9
|
Han J, Zhu K, Zhang X, Harris RA. Enforced microglial depletion and repopulation as a promising strategy for the treatment of neurological disorders. Glia 2019; 67:217-231. [PMID: 30378163 PMCID: PMC6635749 DOI: 10.1002/glia.23529] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
Abstract
Microglia are prominent immune cells in the central nervous system (CNS) and are critical players in both neurological development and homeostasis, and in neurological diseases when dysfunctional. Our previous understanding of the phenotypes and functions of microglia has been greatly extended by a dearth of recent investigations. Distinct genetically defined subsets of microglia are now recognized to perform their own independent functions in specific conditions. The molecular profiling of single microglial cells indicates extensively heterogeneous reactions in different neurological disorders, resulting in multiple potentials for crosstalk with other kinds of CNS cells such as astrocytes and neurons. In settings of neurological diseases it could thus be prudent to establish effective cell-based therapies by targeting entire microglial networks. Notably, activated microglial depletion through genetic targeting or pharmacological therapies within a suitable time window can stimulate replenishment of the CNS niche with new microglia. Additionally, enforced repopulation through provision of replacement cells also represents a potential means of exchanging dysfunctional with functional microglia. In each setting the newly repopulated microglia might have the potential to resolve ongoing neuroinflammation. In this review, we aim to summarize the most recent knowledge of microglia and to highlight microglial depletion and subsequent repopulation as a promising cell replacement therapy. Although glial cell replacement therapy is still in its infancy and future translational studies are still required, the approach is scientifically sound and provides new optimism for managing the neurotoxicity and neuroinflammation induced by activated microglia.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Xing‐Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| |
Collapse
|
10
|
Caruso G, Fresta CG, Musso N, Giambirtone M, Grasso M, Spampinato SF, Merlo S, Drago F, Lazzarino G, Sortino MA, Lunte SM, Caraci F. Carnosine Prevents Aβ-Induced Oxidative Stress and Inflammation in Microglial Cells: A Key Role of TGF-β1. Cells 2019; 8:E64. [PMID: 30658430 PMCID: PMC6356400 DOI: 10.3390/cells8010064] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 12/16/2022] Open
Abstract
Carnosine (β-alanyl-L-histidine), a dipeptide, is an endogenous antioxidant widely distributed in excitable tissues like muscles and the brain. Carnosine is involved in cellular defense mechanisms against oxidative stress, including the inhibition of amyloid-beta (Aβ) aggregation and the scavenging of reactive species. Microglia play a central role in the pathogenesis of Alzheimer's disease, promoting neuroinflammation through the secretion of inflammatory mediators and free radicals. However, the effects of carnosine on microglial cells and neuroinflammation are not well understood. In the present work, carnosine was tested for its ability to protect BV-2 microglial cells against oligomeric Aβ1-42-induced oxidative stress and inflammation. Carnosine prevented cell death in BV-2 cells challenged with Aβ oligomers through multiple mechanisms. Specifically, carnosine lowered the oxidative stress by decreasing NO and O₂-• intracellular levels as well as the expression of iNOS and Nox enzymes. Carnosine also decreased the secretion of pro-inflammatory cytokines such as IL-1β, simultaneously rescuing IL-10 levels and increasing the expression and the release of TGF-β1. Carnosine also prevented Aβ-induced neurodegeneration in mixed neuronal cultures challenged with Aβ oligomers, and these neuroprotective effects were completely abolished by SB431542, a selective inhibitor of the type-1 TGF-β receptor. Our data suggest a multimodal mechanism of action of carnosine underlying its protective effects on microglial cells against Aβ toxicity with a key role of TGF-β1 in mediating these protective effects.
Collapse
Affiliation(s)
| | - Claudia G Fresta
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA.
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA.
| | - Nicolò Musso
- Bio-nanotech Research and Innovation Tower (BRIT), University of Catania, 95125 Catania, Italy.
| | | | - Margherita Grasso
- Oasi Research Institute-IRCCS, 94018 Troina, Italy.
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy.
| | - Simona F Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95125 Catania, Italy.
| | - Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95125 Catania, Italy.
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95125 Catania, Italy.
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, 95125 Catania, Italy.
| | - Maria A Sortino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95125 Catania, Italy.
| | - Susan M Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA.
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA.
- Department of Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA.
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, 94018 Troina, Italy.
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy.
| |
Collapse
|