1
|
Amirshahrokhi K, Imani M. Therapeutic Effect of Levetiracetam Against Thioacetamide-Induced Hepatic Encephalopathy Through Inhibition of Oxidative Stress and Downregulation of NF-κB, NLRP3, iNOS/NO, Pro-Inflammatory Cytokines and Apoptosis. Inflammation 2024; 47:1762-1775. [PMID: 38530519 DOI: 10.1007/s10753-024-02007-4] [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: 01/13/2024] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024]
Abstract
Hepatic encephalopathy (HE) is a serious brain disorder which associated with neurological and psychiatric manifestations. Oxidative stress and neuroinflammation and apoptosis play main roles in the development of brain damage in HE. Levetiracetam is an antiseizure drug with established antioxidant and anti-inflammatory activities. In the present study we investigated the therapeutic effects of levetiracetam against brain injury in HE and its underlying mechanisms of action. Male C57BL/6 mice were subjected to the induction of HE by the injection of thioacetamide (200 mg/kg) for 2 days. Mice were treated with levetiracetam at two doses (50 or 100 mg/kg/day) for 3 days in the treatment groups. Animals were subjected to a behavioral test and the brain tissues were dissected for histopathological, biochemical, gene expression and immunofluorescence analysis. The results showed that levetiracetam alleviated body weight loss and improved locomotor activity of mice with HE. Levetiracetam treatment decreased the histopathological changes, lipid peroxidation and protein carbonylation while restored the antioxidants (GSH, SOD and CAT) in the brain. Levetiracetam decreased the expression and activity of NF-κB, NOD-like receptor pyrin domain-containing protein 3 (NLRP3) and pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and IFN-γ) in the brain tissue. Administration of levetiracetam inhibited iNOS/NO pathway and myeloperoxidase (MPO) activity in the brain. Moreover, caspase-3 was decreased and the ratio of Bcl2/Bax was increased in the brain of mice treated with levetiracetam. These findings suggest that levetiracetam may be a promising therapeutic agent for brain injury in HE through inhibiting the oxidative, inflammatory and apoptotic pathways.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, P. O. Box 5618953141, Ardabil, Iran.
| | - Mahsa Imani
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
2
|
Arfaei R, Mikaeili N, Daj F, Boroumand A, Kheyri A, Yaraghi P, Shirzad Z, Keshavarz M, Hassanshahi G, Jafarzadeh A, Shahrokhi VM, Khorramdelazad H. Decoding the role of the CCL2/CCR2 axis in Alzheimer's disease and innovating therapeutic approaches: Keeping All options open. Int Immunopharmacol 2024; 135:112328. [PMID: 38796962 DOI: 10.1016/j.intimp.2024.112328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Alzheimer's disease (AD), as a neurodegenerative disorder, distresses the elderly in large numbers and is characterized by β-amyloid (Aβ) accumulation, elevated tau protein levels, and chronic inflammation. The brain's immune system is aided by microglia and astrocytes, which produce chemokines and cytokines. Nevertheless, dysregulated expression can cause hyperinflammation and lead to neurodegeneration. CCL2/CCR2 chemokines are implicated in neurodegenerative diseases exacerbating. Inflicting damage on nerves and central nervous system (CNS) cells is the function of this axis, which recruits and migrates immune cells, including monocytes and macrophages. It has been shown that targeting the CCL2/CCR2 axis may be a therapeutic option for inflammatory diseases. Using the current knowledge about the involvement of the CCL2/CCR2 axis in the immunopathogenesis of AD, this comprehensive review synthesizes existing information. It also explores potential therapeutic options, including modulation of the CCL2/CCR2 axis as a possible strategy in AD.
Collapse
Affiliation(s)
- Reyhaneh Arfaei
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Narges Mikaeili
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Daj
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Armin Boroumand
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abbas Kheyri
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Pegah Yaraghi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Shirzad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Keshavarz
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Vahid Mohammadi Shahrokhi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
3
|
Bäckström T, Doverskog M, Blackburn TP, Scharschmidt BF, Felipo V. Allopregnanolone and its antagonist modulate neuroinflammation and neurological impairment. Neurosci Biobehav Rev 2024; 161:105668. [PMID: 38608826 DOI: 10.1016/j.neubiorev.2024.105668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
Neuroinflammation accompanies several brain disorders, either as a secondary consequence or as a primary cause and may contribute importantly to disease pathogenesis. Neurosteroids which act as Positive Steroid Allosteric GABA-A receptor Modulators (Steroid-PAM) appear to modulate neuroinflammation and their levels in the brain may vary because of increased or decreased local production or import from the systemic circulation. The increased synthesis of steroid-PAMs is possibly due to increased expression of the mitochondrial cholesterol transporting protein (TSPO) in neuroinflammatory tissue, and reduced production may be due to changes in the enzymatic activity. Microglia and astrocytes play an important role in neuroinflammation, and their production of inflammatory mediators can be both activated and inhibited by steroid-PAMs and GABA. What is surprising is the finding that both allopregnanolone, a steroid-PAM, and golexanolone, a novel GABA-A receptor modulating steroid antagonist (GAMSA), can inhibit microglia and astrocyte activation and normalize their function. This review focuses on the role of steroid-PAMs in neuroinflammation and their importance in new therapeutic approaches to CNS and liver disease.
Collapse
Affiliation(s)
| | | | | | | | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
4
|
Li N, Huang Y, Wu Y, Wang Q, Ji P. Extracellular vesicles derived from monomeric α-synuclein-treated microglia ameliorate neuroinflammation by delivery of miRNAs targeting PRAK. Neurosci Lett 2024; 818:137562. [PMID: 37984486 DOI: 10.1016/j.neulet.2023.137562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Parkinson's disease (PD) is characterized by the formation of Lewy body, which mainly contains misfolded α-synuclein. Microglial activation plays a role in neurodegeneration. The pathologically oligomeric α-synuclein promotes inflammatory microglia, while physiologically monomeric α-synuclein induces anti-inflammatory microglia, the relationship between these two forms in activating microglia and the molecular mechanism is essentially unknown. In this study, using in vivo and in vitro models, we challenged primary or BV2 microglia with exogenous stimuli including α-synuclein. We examined microglial activation and the underlying mechanism by Western blot, RT-PCR, ELISA, IF, FCM, miRNA sequencing and bioinformatic analysis. Oligomeric α-synuclein activatedmicroglia via theinvolvement of the PRAK/MK5 pathway. The specific PRAK inhibitor GLPG0259 could mitigate microglial activation insulted by oligomeric α-synuclein. Monomeric α-synuclein regulated theanti-inflammatory microglia by delivering microglia-derived extracellular vesicles (EVs) in vitro and in vivo. Furthersequencingand bioinformatic analysis of microglial EVs-associated miRNAs indicatedthatmost of these miRNAs targeted PRAK. These results suggest that PRAK serves as an intersection in microglial activation when challenged with conformationally different α-synuclein. EVs derived from microglia treated with monomeric α-synuclein promote anti-inflammatory microglia by delivering miRNAs that target PRAK into recipient microglia.
Collapse
Affiliation(s)
- Na Li
- Department of Immunology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China; Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Yang Huang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yufeng Wu
- Clinical Laboratory Department of Peking University Third Hospital, Beijing 100191, China
| | - Qilong Wang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Pengyu Ji
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, The First School of Clinical Medicine, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
5
|
Sancho-Alonso M, Arenas YM, Izquierdo-Altarejos P, Martinez-Garcia M, Llansola M, Felipo V. Enhanced Activation of the S1PR2-IL-1β-Src-BDNF-TrkB Pathway Mediates Neuroinflammation in the Hippocampus and Cognitive Impairment in Hyperammonemic Rats. Int J Mol Sci 2023; 24:17251. [PMID: 38139078 PMCID: PMC10744193 DOI: 10.3390/ijms242417251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Hyperammonemia contributes to hepatic encephalopathy. In hyperammonemic rats, cognitive function is impaired by altered glutamatergic neurotransmission induced by neuroinflammation. The underlying mechanisms remain unclear. Enhanced sphingosine-1-phosphate receptor 2 (S1PR2) activation in the cerebellum of hyperammonemic rats contributes to neuroinflammation. in In hyperammonemic rats, we assessed if blocking S1PR2 reduced hippocampal neuroinflammation and reversed cognitive impairment and if the signaling pathways were involved. S1PR2 was blocked with intracerebral JTE-013, and cognitive function was evaluated. The signaling pathways inducing neuroinflammation and altered glutamate receptors were analyzed in hippocampal slices. JTE-013 improved cognitive function in the hyperammonemic rats, and hyperammonemia increased S1P. This increased IL-1β, which enhanced Src activity, increased CCL2, activated microglia and increased the membrane expression of the NMDA receptor subunit GLUN2B. This increased p38-MAPK activity, which altered the membrane expression of AMPA receptor subunits and increased BDNF, which activated the TrkB → PI3K → Akt → CREB pathway, inducing sustained neuroinflammation. This report unveils key pathways involved in the induction and maintenance of neuroinflammation in the hippocampus of hyperammonemic rats and supports S1PR2 as a therapeutic target for cognitive impairment.
Collapse
Affiliation(s)
- María Sancho-Alonso
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Yaiza M. Arenas
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Paula Izquierdo-Altarejos
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Mar Martinez-Garcia
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (M.S.-A.); (Y.M.A.); (P.I.-A.); (M.M.-G.); (V.F.)
| |
Collapse
|
6
|
Leclercq B, Weiner A, Zola M, Mejlacowicz D, Lassiaz P, Jonet L, Gélizé E, Perrot J, Viengchareun S, Zhao M, Behar-Cohen F. The choroidal nervous system: a link between mineralocorticoid receptor and pachychoroid. Acta Neuropathol 2023; 146:747-766. [PMID: 37682293 PMCID: PMC10564818 DOI: 10.1007/s00401-023-02628-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
Central serous chorioretinopathy (CSCR) belongs to the pachychoroid spectrum, a pathological phenotype of the choroidal vasculature, in which blood flow is under the choroidal nervous system (ChNS) regulation. The pathogenesis of CSCR is multifactorial, with the most recognised risk factor being intake of glucocorticoids, which activate both the gluco- and the mineralocorticoid (MR) receptors. As MR over-activation is pathogenic in the retina and choroid, it could mediate the pathogenic effects of glucocorticoids in CSCR. But the role of MR signalling in pachychoroid is unknown and whether it affects the ChNS has not been explored. Using anatomo-neurochemical characterisation of the ChNS in rodents and humans, we discovered that beside innervation of arteries, choroidal veins and choriocapillaris are also innervated, suggesting that the entire choroidal vasculature is under neural control. The numerous synapses together with calcitonin gene-related peptide (CGRP) vesicles juxtaposed to choroidal macrophages indicate a neuro-immune crosstalk. Using ultrastructural approaches, we show that transgenic mice overexpressing human MR, display a pachychoroid-like phenotype, with signs of choroidal neuropathy including myelin abnormalities, accumulation and enlargement of mitochondria and nerves vacuolization. Transcriptomic analysis of the RPE/choroid complex in the transgenic mice reveals regulation of corticoids target genes, known to intervene in nerve pathophysiology, such as Lcn2, rdas1/dexras1, S100a8 and S100a9, rabphilin 3a (Rph3a), secretogranin (Scg2) and Kinesin Family Member 5A (Kif5a). Genes belonging to pathways related to vasculature development, hypoxia, epithelial cell apoptosis, epithelial mesenchymal transition, and inflammation, support the pachychoroid phenotype and highlight downstream molecular targets. Hypotheses on the imaging phenotype of pachychoroid in humans are put forward in the light of these new data. Our results provide evidence that MR overactivation causes a choroidal neuropathy that could explain the pachychoroid phenotype found in transgenic mice overexpressing human MR. In patients with pachychoroid and CSCR in which systemic dysautonomia has been demonstrated, MR-induced choroidal neuropathy could be the missing link between corticoids and pachychoroid.
Collapse
Affiliation(s)
- Bastien Leclercq
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Allon Weiner
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Marta Zola
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
- Ophthalmopole Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Hopital Foch, Suresnes, France
| | - Dan Mejlacowicz
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Patricia Lassiaz
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Laurent Jonet
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Emmanuelle Gélizé
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Julie Perrot
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France
| | - Say Viengchareun
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, Inserm, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, 15 rue de l'Ecole de Médecine, 75006, Paris, France.
- Ophthalmopole Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
- Hopital Foch, Suresnes, France.
| |
Collapse
|
7
|
Walrath T, Najarro KM, Giesy LE, Khair S, Frank DN, Robertson CE, Orlicky DJ, Quillinan N, Idrovo JP, McMahan RH, Kovacs EJ. REMOTE BURN INJURY IN AGED MICE INDUCES COLONIC LYMPHOID AGGREGATE EXPANSION AND DYSBIOSIS OF THE FECAL MICROBIOME WHICH CORRELATES WITH NEUROINFLAMMATION. Shock 2023; 60:585-593. [PMID: 37548929 PMCID: PMC10581426 DOI: 10.1097/shk.0000000000002202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/02/2023] [Accepted: 07/28/2023] [Indexed: 08/08/2023]
Abstract
ABSTRACT The Earth's population is aging, and by 2050, one of six people will be 65 years or older. Therefore, proper treatment of injuries that disproportionately impact people of advanced age will be more important. Clinical studies reveal people 65 years or older account for 16.5% of all burn injuries and experience higher morbidity, including neurocognitive decline, and mortality that we and others believe are mediated, in part, by heightened intestinal permeability. Herein, we used our clinically relevant model of scald burn injury in young and aged mice to determine whether age and burn injury cooperate to induce heightened colonic damage, alterations to the fecal microbiome, and whether resultant changes in the microbiome correlate with neuroinflammation. We found that aged, burn-injured mice have an increase in colonic lymphoid aggregates, inflammation, and proinflammatory chemokine expression when compared with young groups and sham-injured aged mice. We then performed fecal microbiota sequencing and found a striking reduction in gut protective bacterial taxa, including Akkermansia , in the aged burn group compared with all other groups. This reduction correlated with an increase in serum fluorescein isothiocyanate-Dextran administered by gavage, indicating heightened intestinal permeability. Furthermore, loss of Akkermansia was highly correlated with increased messenger RNA expression of neuroinflammatory markers in the brain, including chemokine ligand 2, TNF-α, CXC motif ligand 1, and S100 calcium-binding protein A8. Finally, we discovered that postburn alterations in the microbiome correlated with measures of strength in all treatment groups, and those that performed better on the rotarod and hanging wire tests had higher abundance of Akkermansia than those that performed worse. Taken together, these findings indicate that loss of protective bacteria after burn injury in aged mice contributes to alterations in the colon, gut leakiness, neuroinflammation, and strength. Therefore, supplementation of protective bacteria, such as Akkermansia , after burn injury in aged patients may have therapeutic benefit.
Collapse
Affiliation(s)
- Travis Walrath
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Kevin M. Najarro
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Lauren E. Giesy
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Shanawaj Khair
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
- Molecular Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
- Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Daniel N. Frank
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Charles E. Robertson
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - David J. Orlicky
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Nidia Quillinan
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Juan-Pablo Idrovo
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Rachel H. McMahan
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Elizabeth J. Kovacs
- Department of Surgery, Burn Research and Alcohol Research Programs, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
8
|
Bäckström T, Turkmen S, Das R, Doverskog M, Blackburn TP. The GABA system, a new target for medications against cognitive impairment-Associated with neuroactive steroids. J Intern Med 2023; 294:281-294. [PMID: 37518841 DOI: 10.1111/joim.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
The prevalence of cognitive dysfunction, dementia, and neurodegenerative disorders such as Alzheimer's disease (AD) is increasing in parallel with an aging population. Distinct types of chronic stress are thought to be instrumental in the development of cognitive impairment in central nervous system (CNS) disorders where cognitive impairment is a major unmet medical need. Increased GABAergic tone is a mediator of stress effects but is also a result of other factors in CNS disorders. Positive GABA-A receptor modulating stress and sex steroids (steroid-PAMs) such as allopregnanolone (ALLO) and medroxyprogesterone acetate can provoke impaired cognition. As such, ALLO impairs memory and learning in both animals and humans. In transgenic AD animal studies, continuous exposure to ALLO at physiological levels impairs cognition and increases degenerative AD pathology, whereas intermittent ALLO injections enhance cognition, indicating pleiotropic functions of ALLO. We have shown that GABA-A receptor modulating steroid antagonists (GAMSAs) can block the acute negative cognitive impairment of ALLO on memory in animal studies and in patients with cognitive impairment due to hepatic encephalopathy. Here we describe disorders affected by steroid-PAMs and opportunities to treat these adverse effects of steroid-PAMs with novel GAMSAs.
Collapse
Affiliation(s)
| | - Sahruh Turkmen
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
| | - Roshni Das
- Department of Clinical Sciences, University of Umeå, Umeå, Sweden
- Umecrine Cognition AB, Solna, Sweden
| | | | | |
Collapse
|
9
|
Chen Y, Zheng D, Wang H, Zhang S, Zhou Y, Ke X, Chen G. Lipocalin 2 in the Paraventricular Thalamic Nucleus Contributes to DSS-Induced Depressive-Like Behaviors. Neurosci Bull 2023; 39:1263-1277. [PMID: 36920644 PMCID: PMC10387009 DOI: 10.1007/s12264-023-01047-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/23/2023] [Indexed: 03/16/2023] Open
Abstract
The incidence rate of anxiety and depression is significantly higher in patients with inflammatory bowel diseases (IBD) than in the general population. The mechanisms underlying dextran sulfate sodium (DSS)-induced depressive-like behaviors are still unclear. We clarified that IBD mice induced by repeated administration of DSS presented depressive-like behaviors. The paraventricular thalamic nucleus (PVT) was regarded as the activated brain region by the number of c-fos-labeled neurons. RNA-sequencing analysis showed that lipocalin 2 (Lcn2) was upregulated in the PVT of mice with DSS-induced depressive behaviors. Upregulating Lcn2 from neuronal activity induced dendritic spine loss and the secreted protein induced chemokine expression and subsequently contributed to microglial activation leading to blood-brain barrier permeability. Moreover, Lcn2 silencing in the PVT alleviated the DSS-induced depressive-like behaviors. The present study demonstrated that elevated Lcn2 in the PVT is a critical factor for DSS-induced depressive behaviors.
Collapse
Affiliation(s)
- Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Du Zheng
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Hongwei Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Shuxia Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Xinlong Ke
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
10
|
Santos RPC, Toscano ECDB, Rachid MA. Anti-inflammatory strategies for hepatic encephalopathy: preclinical studies. ARQUIVOS DE NEURO-PSIQUIATRIA 2023. [PMID: 37487550 PMCID: PMC10371400 DOI: 10.1055/s-0043-1767819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Hepatic encephalopathy (HE) is a potentially reversible neuropsychiatric syndrome. Often, HE causes cognitive and motor dysfunctions due to an acute or chronic insufficiency of the liver or a shunting between the hepatic portal vein and systemic vasculature. Liver damage induces peripheral changes, such as in the metabolism and peripheral inflammatory responses that trigger exacerbated neuroinflammation. In experimental models, anti-inflammatory strategies have demonstrated neuroprotective effects, leading to a reduction in HE-related cognitive and motor impairments. In this scenario, a growing body of evidence has shown that peripheral and central nervous system inflammation are promising preclinical targets. In this review, we performed an overview of FDA-approved drugs and natural compounds which are used in the treatment of other neurological and nonneurological diseases that have played a neuroprotective role in experimental HE, at least in part, through anti-inflammatory mechanisms. Despite the exciting results from animal models, the available data should be critically interpreted, highlighting the importance of translating the findings for clinical essays.
Collapse
Affiliation(s)
- Rafaela Pinto Coelho Santos
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Laboratório de Patologia Celular e Molecular, Belo Horizonte MG, Brazil
| | - Eliana Cristina de Brito Toscano
- Universidade Federal de Juiz de Fora, Faculdade de Medicina, Departamento de Patologia, Laboratório Integrado de Pesquisa em Patologia, Juiz de Fora MG, Brazil
- Universidade Federal de Juiz e Fora, Faculdade de Medicina, Programa de Pós-Graduação em Saúde, Juiz de Fora MG, Brazil
| | - Milene Alvarenga Rachid
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Laboratório de Patologia Celular e Molecular, Belo Horizonte MG, Brazil
| |
Collapse
|
11
|
Park J, Lee C, Kim YT. Effects of Natural Product-Derived Compounds on Inflammatory Pain via Regulation of Microglial Activation. Pharmaceuticals (Basel) 2023; 16:941. [PMID: 37513853 PMCID: PMC10386117 DOI: 10.3390/ph16070941] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammatory pain is a type of pain caused by tissue damage associated with inflammation and is characterized by hypersensitivity to pain and neuroinflammation in the spinal cord. Neuroinflammation is significantly increased by various neurotransmitters and cytokines that are expressed in activated primary afferent neurons, and it plays a pivotal role in the development of inflammatory pain. The activation of microglia and elevated levels of pro-inflammatory cytokines are the hallmark features of neuroinflammation. During the development of neuroinflammation, various intracellular signaling pathways are activated or inhibited in microglia, leading to the regulation of inflammatory proteins and cytokines. Numerous attempts have been conducted to alleviate inflammatory pain by inhibiting microglial activation. Natural products and their compounds have gained attention as potential candidates for suppressing inflammatory pain due to verified safety through centuries of use. Many studies have also shown that natural product-derived compounds have the potential to suppress microglial activation and alleviate inflammatory pain. Herein, we review the literature on inflammatory mediators and intracellular signaling involved in microglial activation in inflammatory pain, as well as natural product-derived compounds that have been found to suppress microglial activation. This review suggests that natural product-derived compounds have the potential to alleviate inflammatory pain through the suppression of microglial activation.
Collapse
Affiliation(s)
- Joon Park
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
- Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Changho Lee
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju 55365, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
12
|
Neuroblasts migration under control of reactive astrocyte-derived BDNF: a promising therapy in late neurogenesis after traumatic brain injury. Stem Cell Res Ther 2023; 14:2. [PMID: 36600294 DOI: 10.1186/s13287-022-03232-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a disease with high mortality and morbidity, which leads to severe neurological dysfunction. Neurogenesis has provided therapeutic options for treating TBI. Brain derived neurotrophic factor (BDNF) plays a key role in neuroblasts migration. We aimed to investigate to the key regulating principle of BDNF in endogenous neuroblasts migration in a mouse TBI model. METHODS In this study, controlled cortical impact (CCI) mice (C57BL/6J) model was established to mimic TBI. The sham mice served as control. Immunofluorescence staining and enzyme-linked immunosorbent assay were performed on the CCI groups (day 1, 3, 7, 14 and 21 after CCI) and the sham group. All the data were analyzed with Student's t-test or one-way or two-way analysis of variance followed by Tukey's post hoc test. RESULTS Our results revealed that neuroblasts migration initiated as early as day 1, peaking at day 7, and persisted till day 21. The spatiotemporal profile of BDNF expression was similar to that of neuroblasts migration, and BDNF level following CCI was consistently higher in injured cortex than in subventricular zone (SVZ). Reactive astrocytes account for the major resource of BDNF along the migrating path, localized with neuroblasts in proximity. Moreover, injection of exogenous CC chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1, at random sites promoted neuroblasts migration and astrocytic BDNF expression in both normal and CCI mice (day 28). These provoked neuroblasts can also differentiate into mature neurons. CC chemokine ligand receptor 2 antagonist can restrain the neuroblasts migration after TBI. CONCLUSIONS Neuroblasts migrated along the activated astrocytic tunnel, directed by BDNF gradient between SVZ and injured cortex after TBI. CCL2 might be a key regulator in the above endogenous neuroblasts migration. Moreover, delayed CCL2 administration may provide a promising therapeutic strategy for late neurogenesis post-trauma.
Collapse
|
13
|
Sargeant TJ, Fourrier C. Human monocyte-derived microglia-like cell models: A review of the benefits, limitations and recommendations. Brain Behav Immun 2023; 107:98-109. [PMID: 36202170 DOI: 10.1016/j.bbi.2022.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 09/09/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
In the last few decades, mounting evidence has highlighted that microglia have crucial roles in both health and disease. This has led to a growing interest in studying human microglia in disease-relevant models. However, current models present limitations that can make them unsuitable for moderate throughput studies in human cohorts. Primary human microglia are ethically and technically difficult to obtain and only allow low throughput studies; immortalized cell lines have been shown to differ too greatly from primary human microglia; and induced pluripotent stem cell-derived microglia, although physiologically relevant in most contexts, have limited potential for use in large cohorts of people or for personalised drug screening. In this review, we discuss monocyte-derived microglia-like (MDMi) cells, a model that has been developed and increasingly used in the last decade, using human monocytes isolated from blood samples. We describe the variety of protocols that have been used to develop MDMi cell models and highlight a need for standardization across protocols. We then summarize data that validate MDMi cells as an appropriate model to study human microglia in health and disease. We also present the benefits and limitations of using this approach to study human microglia compared with other microglial models, when used in combination with the relevant downstream applications and with cross-validation of findings in other systems. Finally, we summarize the paradigms in which MDMi models have been used to advance research on microglia in immune-related disease. This review is an important reference for scientists who seek to establish MDMi cells as a microglial model for the advancement of our understanding of microglia in human health and disease.
Collapse
Affiliation(s)
- Timothy J Sargeant
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia.
| | - Célia Fourrier
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
14
|
Walrath T, McMahan RH, Idrovo JP, Quillinan N, Kovacs EJ. Cutaneous burn injury induces neuroinflammation and reactive astrocyte activation in the hippocampus of aged mice. Exp Gerontol 2022; 169:111975. [PMID: 36208823 DOI: 10.1016/j.exger.2022.111975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND By 2050, one in six people globally will be 65 or older. Confusion and delirium are significant complications after burn injury, especially in the elderly population. The etiology is still unknown, however complications may be driven by pro-inflammatory activation of astrocytes within the hippocampus (HPC) after burn injury. Reduced levels of phosphorylated cyclic-AMP response binding element (pCREB), caused by elevated systemic pro-inflammatory cytokines, could lead to cognitive decline and memory impairment. METHODS To examine the effects of remote injury on neuroinflammation in advanced age, young and aged mice were subjected to a 15 % total body surface area scald burn or sham injury, and euthanized after 24 h. Expression of ccl2 and tnfa were measured by qPCR in the whole brain and HPC. Astrocyte activation was measured by immunofluorescence within the HPC. pCREB was measured by immunohistochemistry in the dentate gyrus. RESULTS We saw an 80-fold increase in ccl2 and a 30-fold elevation in tnfa after injury in the whole brain of aged mice compared to young groups and aged sham mice (p < 0.05 and p < 0.05, respectively). Additionally, there was a 30-fold increase in ccl2 within isolated HPC of aged injured mice when compared to sham injured animals (p < 0.05). When investigating specific HPC regions, immunofluorescence staining showed a >20 % rise in glial fibrillary acidic protein (GFAP) positive astrocytes within the cornu ammonis 3 (CA3) of aged injured mice when compared to all other groups (p < 0.05). Lastly, we observed a >20 % decrease in pCREB staining by immunohistochemistry in the dentate gyrus of aged mice compared to young regardless of injury (p < 0.05). CONCLUSIONS These novel data suggest that remote injury in aged, but not young, mice is associated with neuroinflammation and astrocyte activation within the HPC. These factors, paired with an age related reduction in pCREB, could help explain the increased cognitive decline seen in burn patients of advanced age. To our knowledge, we are the first group to examine the impact of advanced age combined with burn injury on inflammation and astrocyte activation within the brain.
Collapse
Affiliation(s)
- Travis Walrath
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Rachel H McMahan
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Juan-Pablo Idrovo
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Nidia Quillinan
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elizabeth J Kovacs
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, United States of America.
| |
Collapse
|
15
|
Mincheva G, Gimenez‐Garzo C, Izquierdo‐Altarejos P, Martinez‐Garcia M, Doverskog M, Blackburn TP, Hällgren A, Bäckström T, Llansola M, Felipo V. Golexanolone, a GABA A receptor modulating steroid antagonist, restores motor coordination and cognitive function in hyperammonemic rats by dual effects on peripheral inflammation and neuroinflammation. CNS Neurosci Ther 2022; 28:1861-1874. [PMID: 35880480 PMCID: PMC9532914 DOI: 10.1111/cns.13926] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/29/2022] Open
Abstract
AIMS Hyperammonemic rats show peripheral inflammation, increased GABAergic neurotransmission and neuroinflammation in cerebellum and hippocampus which induce motor incoordination and cognitive impairment. Neuroinflammation enhances GABAergic neurotransmission in cerebellum by enhancing the TNFR1-glutaminase-GAT3 and TNFR1-CCL2-TrkB-KCC2 pathways. Golexanolone reduces GABAA receptors potentiation by allopregnanolone. This work aimed to assess if treatment of hyperammonemic rats with golexanolone reduces peripheral inflammation and neuroinflammation and restores cognitive and motor function and to analyze underlying mechanisms. METHODS Rats were treated with golexanolone and effects on peripheral inflammation, neuroinflammation, TNFR1-glutaminase-GAT3 and TNFR1-CCL2-TrkB-KCC2 pathways, and cognitive and motor function were analyzed. RESULTS Hyperammonemic rats show increased TNFα and reduced IL-10 in plasma, microglia and astrocytes activation in cerebellum and hippocampus, and impaired motor coordination and spatial and short-term memories. Treating hyperammonemic rats with golexanolone reversed changes in peripheral inflammation, microglia and astrocytes activation and restored motor coordination and spatial and short-term memory. This was associated with reversal of the hyperammonemia-enhanced activation in cerebellum of the TNFR1-glutaminase-GAT3 and TNFR1-CCL2-TrkB-KCC2 pathways. CONCLUSION Reducing GABAA receptors activation with golexanolone reduces peripheral inflammation and neuroinflammation and improves cognitive and motor function in hyperammonemic rats. The effects identified would also occur in patients with hepatic encephalopathy and, likely, in other pathologies associated with neuroinflammation.
Collapse
Affiliation(s)
- Gergana Mincheva
- Laboratory of NeurobiologyCentro de Investigación Príncipe FelipeValenciaSpain
| | - Carla Gimenez‐Garzo
- Laboratory of NeurobiologyCentro de Investigación Príncipe FelipeValenciaSpain
| | | | - Mar Martinez‐Garcia
- Laboratory of NeurobiologyCentro de Investigación Príncipe FelipeValenciaSpain
| | | | | | | | - Torbjörn Bäckström
- Umecrine Cognition ABSolnaSweden
- Umeå Neurosteroid Research CenterClinical Sciences at Umeå UniversityUmeåSweden
| | - Marta Llansola
- Laboratory of NeurobiologyCentro de Investigación Príncipe FelipeValenciaSpain
| | - Vicente Felipo
- Laboratory of NeurobiologyCentro de Investigación Príncipe FelipeValenciaSpain
| |
Collapse
|
16
|
Amirshahrokhi K, Niapour A. Carvedilol attenuates brain damage in mice with hepatic encephalopathy. Int Immunopharmacol 2022; 111:109119. [PMID: 35933745 DOI: 10.1016/j.intimp.2022.109119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/17/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022]
Abstract
Brain injury is the most common and serious consequence of hepatic encephalopathy (HE), and its pathophysiology is poorly understood. Excessive inflammatory, oxidative and apoptotic responses are the major mechanisms involved in the progression of brain injury induced by HE. Carvedilol is an adrenergic receptor antagonist with pronouncedantioxidant and anti-inflammatory activity. The present study aimed to investigatethe effects and underlying mechanisms of carvedilol on HE-induced brain damage in mice. Experimental model of HE was induced by the injection of thioacetamide (200 mg/kg) for two consecutive days and then mice were treated with carvedilol (10 or 20 mg/kg/day, orally) for 3 days in treatment groups. After the behavioral test, animals were sacrificed and the brain tissues were collected for biochemical, real time PCR and immunohistochemical analysis. The results showed that carvedilol improved locomotor impairment and reduced mortality rate in mice with HE. Carvedilol treatment decreased the brain levels of oxidative stress markers and induced Nrf2/HO-1 pathway. Carvedilol inhibited the activity of nuclear factor kappa B (NF-κB) and the expression of pro-inflammatory cytokines TNF-α, IL1β and IL-6 in the brain tissues. Treatment of mice with carvedilol caused a significant reduction in the brain levels of iNOS/NO, myeloperoxidase (MPO), cyclooxygenase (COX)-2 and chemokine MCP-1 as proinflammatory mediators in HE. Moreover, the ratio of Bcl2/Bax was increased and apoptotic cell death was decreased in the brain of mice treated with carvedilol. In conclusion, carvedilol exerted protective effect against HE-induced brain injury through increasing antioxidant defense mechanisms and inhibitionof inflammatory and apoptotic pathways.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran; Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali Niapour
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
17
|
Shen H, Pei H, Zhai L, Guan Q, Wang G. Salvianolic acid C improves cerebral ischemia reperfusion injury through suppressing microglial cell M1 polarization and promoting cerebral angiogenesis. Int Immunopharmacol 2022; 110:109021. [PMID: 35810493 DOI: 10.1016/j.intimp.2022.109021] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022]
Abstract
This study aimed to investigate the mechanism of salvianolic acid C (SAC), the active ingredient in Salvia miltiorrhiza, in improving cerebral ischemia injury. The mouse microglial cells BV2 and mouse endothelial cells bEnd.3 were used as the objects of study. LPS/IFN-γ was applied to simulate the BV2 polarization, and bEnd.3 cells were treated under hypoxic condition. The BV2 cell polarization level was measured through flow cytometry (FCM), the TLR4 and MyD88 expression levels were detected by fluorescence staining, whereas the expression of inflammatory factors TNF-α, IL-6 and IL-1β was analyzed through ELISA. Tubule formation assay was also conducted to observe the tubule formation ability of bEnd.3 cells in vitro, and the level of VEGFR2 was detected by fluorescence staining. Cells were treated with the PKM2 inhibitor IN3, aiming to observe the influence of SAC on glycolysis of BV2 cells. In addition, the mouse model of cerebral ischemia was constructed through the middle cerebral artery occlusion (MCAO) method, and the pathological changes in brain tissues were detected after SAC intervention. Meanwhile, the levels of IBA-1, CD31 and ZO-1 were determined through histochemical staining. Nissl staining to detect nerve cell damage. In BV2 cell experiment, SAC suppressed the M1 polarization of BV2 cells, reduced the inflammatory factor levels, and inhibited the activation of TLR4 signal through suppressing glycolysis. When PKM2 was suppressed, the effects of SAC were antagonized. In the bEnd.3 model, SAC promoted tubule formation in bEnd.3 cells under hypoxic condition, and increased the expression of VEGFR2 and Notch1. In the mouse model, SAC improved the neurological function in MCAO mice, and inhibited the activation of microglial cells and the expression of inflammatory factors. At the same time, SAC up-regulated the expression of ZO-1 and CD31, and maintained the blood-brain barrier (BBB) function. As a major component of Salvia miltiorrhiza, SAC can suppress microglial cell polarization and promote tubule formation in endothelial cells to exert the neurological repair function in cerebral ischemia. SAC is a multi-functional neuroprotective small molecule.
Collapse
Affiliation(s)
- Heping Shen
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China
| | - Hongyan Pei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Liping Zhai
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China
| | - Qiaobing Guan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, China.
| | - Genghuan Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Jiaxing University, China.
| |
Collapse
|
18
|
Leone P, Mincheva G, Balzano T, Malaguarnera M, Felipo V, Llansola M. Rifaximin Improves Spatial Learning and Memory Impairment in Rats with Liver Damage-Associated Neuroinflammation. Biomedicines 2022; 10:1263. [PMID: 35740285 PMCID: PMC9219896 DOI: 10.3390/biomedicines10061263] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/04/2022] Open
Abstract
Patients with non-alcoholic fatty liver disease (NAFLD) may show mild cognitive impairment. Neuroinflammation in the hippocampus mediates cognitive impairment in rat models of minimal hepatic encephalopathy (MHE). Treatment with rifaximin reverses cognitive impairment in a large proportion of cirrhotic patients with MHE. However, the underlying mechanisms remain unclear. The aims of this work were to assess if rats with mild liver damage, as a model of NAFLD, show neuroinflammation in the hippocampus and impaired cognitive function, if treatment with rifaximin reverses it, and to study the underlying mechanisms. Mild liver damage was induced with carbon-tetrachloride. Infiltration of immune cells, glial activation, and cytokine expression, as well as glutamate receptors expression in the hippocampus and cognitive function were assessed. We assessed the effects of daily treatment with rifaximin on the alterations showed by these rats. Rats with mild liver damage showed hippocampal neuroinflammation, reduced membrane expression of glutamate N-methyl-D-aspartate (NMDA) receptor subunits, and impaired spatial memory. Increased C-C Motif Chemokine Ligand 2 (CCL2), infiltration of monocytes, microglia activation, and increased tumor necrosis factor α (TNFα) were reversed by rifaximin, that normalized NMDA receptor expression and improved spatial memory. Thus, rifaximin reduces neuroinflammation and improves cognitive function in rats with mild liver damage, being a promising therapy for patients with NAFLD showing mild cognitive impairment.
Collapse
Affiliation(s)
- Paola Leone
- Mar Lab Department of Neuroscience, NYU Grossman School of Medicine Science Building, New York, NY 10016, USA;
| | - Gergana Mincheva
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (G.M.); (M.L.)
| | - Tiziano Balzano
- Centro Integral de Neurociencias, A.C. HM Hospital Universitario Puerta del Sur CINAC, 28938 Madrid, Spain;
| | - Michele Malaguarnera
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, 46010 Valencia, Spain;
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (G.M.); (M.L.)
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (G.M.); (M.L.)
| |
Collapse
|
19
|
Gutiérrez IL, Novellino F, Caso JR, García-Bueno B, Leza JC, Madrigal JLM. CCL2 Inhibition of Pro-Resolving Mediators Potentiates Neuroinflammation in Astrocytes. Int J Mol Sci 2022; 23:ijms23063307. [PMID: 35328727 PMCID: PMC8949263 DOI: 10.3390/ijms23063307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
The chemokine CCL2 participates in multiple neuroinflammatory processes, mainly through the recruitment of glial cells. However, CCL2 has also been proven to exert different types of actions on these cells, including the modification of their response to inflammatory stimuli. In the present study we analyzed the effect of CCL2 on the resolution of inflammation in astrocytes. We observed that genetic removal of CCL2 increases the expression of the enzymes responsible for the synthesis of specialized pro-resolving mediators arachidonate 15-lipoxygenase and arachidonate 5-lipoxygenase in the brain cortex of 5xFAD mice. The expression of FPR2 receptor, known to mediate the activity of pro-resolving mediators was also increased in mice lacking CCL2.The downregulation of these proteins by CCL2 was also observed in cultured astrocytes. This suggests that CCL2 inhibition of the resolution of inflammation could facilitate the progression of neuroinflammatory processes. The production of the pro-inflammatory cytokine IL-1beta by astrocytes was analyzed, and allowed us to confirm that CCL2 potentiates the activation of astrocytes trough the inhibition of pro-resolving pathways mediated by Resolvin D1. In addition, the analysis of the expression of TNFalpha, MIP1alpha and NOS2 further confirmed CCL2 inhibition of inflammation resolution in astrocytes.
Collapse
Affiliation(s)
- Irene L. Gutiérrez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Fabiana Novellino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council, Viale Europa, 88100 Catanzaro, Italy
| | - Javier R. Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Juan C. Leza
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - José L. M. Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Correspondence: ; Tel.: +34-913941463
| |
Collapse
|
20
|
Bioinformatics-Based Analysis of lncRNA-mRNA Interaction Network of Mild Hepatic Encephalopathy in Cirrhosis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:7777699. [PMID: 34938356 PMCID: PMC8687767 DOI: 10.1155/2021/7777699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Backgrounds Serum long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) interaction network was discovered to exert an important role in liver cirrhosis while little is known in mild hepatic encephalopathy (MHE). Therefore, we aim to systematically evaluate the serum lncRNA-mRNA network and its regulatory mechanism in MHE. Methods The data of serum mRNAs and lncRNAs were derived from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were calculated between 11 cirrhotic patients with and without MHE. Next, the biological functions and underlined pathways of DEGs were determined through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Finally, an interactive network between lncRNAs and mRNAs was built, and hub genes were identified, respectively. Results A total of 64 differentially expressed lncRNAs (dif-lncRNAs) were found between patients with and without MHE, including 30 up- and 34 downregulated genes. 187 differentially expressed mRNAs (dif-mRNAs) were identified, including 84 up- and 103 downregulated genes. Functional enrichment analysis suggested that the regulatory pathways involved in MHE mainly consisted of a series of immune and inflammatory responses. Several hub mRNAs involved in regulatory network were identified, including CCL5, CCR5, CXCR3, CD274, STAT1, CXCR6, and EOMES. In addition, lnc-FAM84B-8 and lnc-SAMD3-1 were found to regulate these above hub genes through building a lncRNA-mRNA network. Conclusion This is the first study to construct the serum lncRNA-mRNA network in MHE, demonstrating the critical role of lncRNAs in regulating inflammatory and immunological profiles in the developing of MHE, suggesting a latent mechanism in this pathophysiological process.
Collapse
|
21
|
El Khiat A, El Hiba O, Tamegart L, Rais H, Fdil N, Sellami S, El Mokhtar MA, Gamrani H. Time dependent alteration of locomotor behavior in rat with acute liver failure induced cerebellar neuroinflammation and neuro-astroglial damage. J Chem Neuroanat 2021; 119:102055. [PMID: 34863855 DOI: 10.1016/j.jchemneu.2021.102055] [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: 09/16/2021] [Revised: 11/27/2021] [Accepted: 11/27/2021] [Indexed: 11/26/2022]
Abstract
Hepatic encephalopathy (HE) is a neurophysiological syndrome secondary to acute or chronic liver failure. Studies showed that HE patients exhibit a deficit in motor coordination, which may result from cerebellar functional impairment. The aim of this study is to assess the time-dependent alteration of locomotor behavior and the glial and neuronal alteration in rat with acute HE induced chemically. The study was carried out in male Sprague-Dawley rats with thioacetamide (TAA) induced acute liver failure at different stages 12 h, 24 h and 36 h. Hepatic and renal functions were assessed via various biochemical and histopathological examinations, while the cerebellum and the midbrain were examined using histology and immunohistochemistry for tyrosine hydroxylase (TH), cyclooxygenase-2 (COX-2) and glial fibrillary acidic protein (GFAP). We used as well, the open field test and the Rotarod test for assessing the locomotor activity and coordination. Our data showed a progressive loss of liver function and a progressive alteration in locomotor behavior and motor coordination in acute HE rats. In the cerebellum, we noted an increase in the degeneration of cerebellar Purkinje neurons parallel to increased COX-2 immunoreactivity together with astrocytic morphology and density changes. Likewise, in substantia nigra pars compacta, TH levels were reduced. We showed through the current study, a progressive deterioration in locomotor behavior in acute HE rats, as a result of Purkinje neurons death and a deficient dopaminergic neurotransmission, together with the morpho-functional astroglial modifications involving the oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Abdelaati El Khiat
- Laboratory of Clinical and Experimental Neurosciences and Environment, faculty of Medicine and Pharmacy, Cadi Ayyad University, 4000 Marrakech, Morocco; Higher Institute of Nursing Professions and Health Techniques, Ouarzazate, Morocco.
| | - Omar El Hiba
- Nutritional Physiopathologies and Toxicology Team, faculty of Sciences, Chouaib Doukkali University, El Jadida, Morocco.
| | - Lahcen Tamegart
- Laboratory of Clinical and Experimental Neurosciences and Environment, faculty of Medicine and Pharmacy, Cadi Ayyad University, 4000 Marrakech, Morocco; Department of Biology, Faculty of Science, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Hanane Rais
- Laboratory of Morphosciences, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Morocco; Mohammed VI University Hospital, Marrakech, Morocco
| | - Naima Fdil
- Metabolics platform, Biochemistry Laboratory, Faculty of Medicine, Cadi Ayyad University, Sidi Abbad, BP 40000 Marrakech, Morocco
| | | | - Mohamed Ait El Mokhtar
- Laboratory of Biochemistry, Environment &Agri-food URAC 36, Department of Biology, Faculty of Sciences and Techniques, Mohmmedia, Hassan II University of Casablanca, Morocco
| | - Halima Gamrani
- Laboratory of Clinical and Experimental Neurosciences and Environment, faculty of Medicine and Pharmacy, Cadi Ayyad University, 4000 Marrakech, Morocco.
| |
Collapse
|
22
|
Claeys W, Van Hoecke L, Lefere S, Geerts A, Verhelst X, Van Vlierberghe H, Degroote H, Devisscher L, Vandenbroucke RE, Van Steenkiste C. The neurogliovascular unit in hepatic encephalopathy. JHEP Rep 2021; 3:100352. [PMID: 34611619 PMCID: PMC8476774 DOI: 10.1016/j.jhepr.2021.100352] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatic encephalopathy (HE) is a neurological complication of hepatic dysfunction and portosystemic shunting. It is highly prevalent in patients with cirrhosis and is associated with poor outcomes. New insights into the role of peripheral origins in HE have led to the development of innovative treatment strategies like faecal microbiota transplantation. However, this broadening of view has not been applied fully to perturbations in the central nervous system. The old paradigm that HE is the clinical manifestation of ammonia-induced astrocyte dysfunction and its secondary neuronal consequences requires updating. In this review, we will use the holistic concept of the neurogliovascular unit to describe central nervous system disturbances in HE, an approach that has proven instrumental in other neurological disorders. We will describe HE as a global dysfunction of the neurogliovascular unit, where blood flow and nutrient supply to the brain, as well as the function of the blood-brain barrier, are impaired. This leads to an accumulation of neurotoxic substances, chief among them ammonia and inflammatory mediators, causing dysfunction of astrocytes and microglia. Finally, glymphatic dysfunction impairs the clearance of these neurotoxins, further aggravating their effect on the brain. Taking a broader view of central nervous system alterations in liver disease could serve as the basis for further research into the specific brain pathophysiology of HE, as well as the development of therapeutic strategies specifically aimed at counteracting the often irreversible central nervous system damage seen in these patients.
Collapse
Key Words
- ABC, ATP-binding cassette
- ACLF, acute-on-chronic liver failure
- AD, acute decompensation
- ALF, acute liver failure
- AOM, azoxymethane
- AQP4, aquaporin 4
- Acute Liver Failure
- Ammonia
- BBB, blood-brain barrier
- BCRP, breast cancer resistance protein
- BDL, bile duct ligation
- Blood-brain barrier
- Brain edema
- CCL, chemokine ligand
- CCR, C-C chemokine receptor
- CE, cerebral oedema
- CLD, chronic liver disease
- CLDN, claudin
- CNS, central nervous system
- CSF, cerebrospinal fluid
- Cirrhosis
- Energy metabolism
- GS, glutamine synthetase
- Glymphatic system
- HE, hepatic encephalopathy
- HO-1, heme oxygenase 1
- IL-, interleukin
- MMP-9, matrix metalloproteinase 9
- MRP, multidrug resistance associated protein
- NGVU
- NGVU, neurogliovascular unit
- NKCC1, Na-K-2Cl cotransporter 1
- Neuroinflammation
- OCLN, occludin
- ONS, oxidative and nitrosative stress
- Oxidative stress
- P-gp, P-glycoprotein
- PCA, portacaval anastomosis
- PSS, portosystemic shunt
- S1PR2, sphingosine-1-phosphate receptor 2
- SUR1, sulfonylurea receptor 1
- Systemic inflammation
- TAA, thioacetamide
- TGFβ, transforming growth factor beta
- TJ, tight junction
- TNF, tumour necrosis factor
- TNFR1, tumour necrosis factor receptor 1
- ZO, zonula occludens
- mPT, mitochondrial pore transition
Collapse
Affiliation(s)
- Wouter Claeys
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
- Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sander Lefere
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences; Liver Research Center Ghent; Ghent University, Ghent, Belgium
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Xavier Verhelst
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Hans Van Vlierberghe
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Helena Degroote
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Liver Research Center Ghent, Ghent University, Ghent, Belgium
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences; Liver Research Center Ghent; Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christophe Van Steenkiste
- Antwerp University, Department of Gastroenterology and Hepatology, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| |
Collapse
|
23
|
Balzano T, Leone P, Ivaylova G, Castro MC, Reyes L, Ramón C, Malaguarnera M, Llansola M, Felipo V. Rifaximin Prevents T-Lymphocytes and Macrophages Infiltration in Cerebellum and Restores Motor Incoordination in Rats with Mild Liver Damage. Biomedicines 2021; 9:1002. [PMID: 34440206 PMCID: PMC8393984 DOI: 10.3390/biomedicines9081002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/30/2022] Open
Abstract
In patients with liver cirrhosis, minimal hepatic encephalopathy (MHE) is triggered by a shift in peripheral inflammation, promoting lymphocyte infiltration into the brain. Rifaximin improves neurological function in MHE by normalizing peripheral inflammation. Patients who died with steatohepatitis showed T-lymphocyte infiltration and neuroinflammation in the cerebellum, suggesting that MHE may already occur in these patients. The aims of this work were to assess, in a rat model of mild liver damage similar to steatohepatitis, whether: (1) the rats show impaired motor coordination in the early phases of liver damage; (2) this is associated with changes in the immune system and infiltration of immune cells into the brain; and (3) rifaximin improves motor incoordination, associated with improved peripheral inflammation, reduced infiltration of immune cells and neuroinflammation in the cerebellum, and restoration of the alterations in neurotransmission. Liver damage was induced by carbon tetrachloride (CCl4) injection over four weeks. Peripheral inflammation, immune cell infiltration, neuroinflammation, and neurotransmission in the cerebellum and motor coordination were assessed. Mild liver damage induces neuroinflammation and altered neurotransmission in the cerebellum and motor incoordination. These alterations are associated with increased TNFa, CCL20, and CX3CL1 in plasma and cerebellum, IL-17 and IL-15 in plasma, and CCL2 in cerebellum. This promotes T-lymphocyte and macrophage infiltration in the cerebellum. Early treatment with rifaximin prevents the shift in peripheral inflammation, immune cell infiltration, neuroinflammation, and motor incoordination. This report provides new clues regarding the mechanisms of the beneficial effects of rifaximin, suggesting that early rifaximin treatment could prevent neurological impairment in patients with steatohepatitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marta Llansola
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, 46012 Valencia, Spain; (T.B.); (P.L.); (G.I.); (M.C.C.); (L.R.); (C.R.); (M.M.); (V.F.)
| | | |
Collapse
|
24
|
Hammond BP, Manek R, Kerr BJ, Macauley MS, Plemel JR. Regulation of microglia population dynamics throughout development, health, and disease. Glia 2021; 69:2771-2797. [PMID: 34115410 DOI: 10.1002/glia.24047] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022]
Abstract
The dynamic expansions and contractions of the microglia population in the central nervous system (CNS) to achieve homeostasis are likely vital for their function. Microglia respond to injury or disease but also help guide neurodevelopment, modulate neural circuitry throughout life, and direct regeneration. Throughout these processes, microglia density changes, as does the volume of area that each microglia surveys. Given that microglia are responsible for sensing subtle alterations to their environment, a change in their density could affect their capacity to mobilize rapidly. In this review, we attempt to synthesize the current literature on the ligands and conditions that promote microglial proliferation across development, adulthood, and neurodegenerative conditions. Microglia display an impressive proliferative capacity during development and in neurodegenerative diseases that is almost completely absent at homeostasis. However, the appropriate function of microglia in each state is critically dependent on density fluctuations that are primarily induced by proliferation. Proliferation is a natural microglial response to insult and often serves neuroprotective functions. In contrast, inappropriate microglial proliferation, whether too much or too little, often precipitates undesirable consequences for nervous system health. Thus, fluctuations in the microglia population are tightly regulated to ensure these immune cells can execute their diverse functions.
Collapse
Affiliation(s)
- Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rupali Manek
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
25
|
Sepehrinezhad A, Shahbazi A, Sahab Negah S, Joghataei MT, Larsen FS. Drug-induced-acute liver failure: A critical appraisal of the thioacetamide model for the study of hepatic encephalopathy. Toxicol Rep 2021; 8:962-970. [PMID: 34026559 PMCID: PMC8122178 DOI: 10.1016/j.toxrep.2021.04.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/17/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic encephalopathy (HE) following acute and chronic liver failure is defined as a complex of neuropsychiatric abnormalities, such as discrete personal changes, sleep disorder, forgetfulness, confusion, and decreasing the level of consciousness to coma. The use and design of suitable animal models that represent clinical features and pathological changes of HE are valuable to map the molecular mechanisms that result in HE. Among different types of animal models, thioacetamide (TAA) has been used extensively for the induction of acute liver injury and HE. This agent is not directly hepatotoxic but its metabolites induce liver injury through the induction of oxidative stress and produce systemic inflammation similar to that seen in acute HE patients. In this short review article, we shortly review the most important pathological findings in animal models of acute HE following the administration of TAA.
Collapse
Key Words
- ALT, alanine aminotransferase
- AQP4, aquaporin 4 water channel
- AST, aspartate aminotransferase
- Acute liver failure
- Animal model
- B7, B7 molecules (CD80+CD86)
- BBB, blood-brain barrier
- CBF, cerebral blood flow
- CCL2, chemokine ligand 2
- CNS, central nervous system
- CTLA4, Cytotoxic T-lymphocyte-associated Protein 4
- CYP2E1, Cytochrome P450 family 2 subfamily E member 1
- GFAP, glial fibrillary acidic protein
- HE, hepatic encephalopathy
- Hepatic encephalopathy
- IL-6, interleukin 6
- IL-β, interleukin 1 β
- Iba1, ionized calcium-binding adaptor molecule 1
- JNK, c-Jun N-terminal kinase
- NAC, N-acetylcysteine
- NF-κB, nuclear factor κB
- OA, L-ornithine-l-aspartate
- ROS, reactive oxygen species
- TAA, thioacetamide
- TASO, thioacetamide sulfoxide
- TASO2, thioacetamide sulfdioxide
- TLR-2, toll-like receptor 2
- TLR-4, toll-like receptor 4
- TNFα, tumor necrosis factor α
- Thioacetamide
- Toxicity pathway
Collapse
Affiliation(s)
- Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fin Stolze Larsen
- Department of Hepatology CA-3163, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
26
|
Singh G, Segura BJ, Georgieff MK, Gisslen T. Fetal inflammation induces acute immune tolerance in the neonatal rat hippocampus. J Neuroinflammation 2021; 18:69. [PMID: 33706765 PMCID: PMC7953777 DOI: 10.1186/s12974-021-02119-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Infants born preterm due to chorioamnionitis are frequently affected by a fetal inflammatory response syndrome (FIRS) and then by subsequent postnatal infections. FIRS and postnatal systemic inflammatory events independently contribute to poor neurocognitive outcomes of preterm infants. Developmental integrity of the hippocampus is crucial for intact neurocognitive outcomes in preterms and hippocampally dependent behaviors are particularly vulnerable to preterm systemic inflammation. How FIRS modulates the hippocampal immune response to acute postnatal inflammatory events is not well understood. METHODS Prenatal LPS exposed (FIRS) and control neonatal rats received i.p. LPS or saline at postnatal day (P) 5. On P7, immune response was evaluated in the hippocampus of four treatment groups by measuring gene expression of inflammatory mediators and cytosolic and nuclear NFκB pathway proteins. Microglial activation was determined by CD11b+ and Iba1+ immunohistochemistry (IHC) and inflammatory gene expression of isolated microglia. Astrocyte reactivity was measured using Gfap+ IHC. RESULTS Postnatal LPS resulted in a robust hippocampal inflammatory response. In contrast, FIRS induced by prenatal LPS attenuated the response to postnatal LPS exposure, evidenced by decreased gene expression of inflammatory mediators, decreased nuclear NFκB p65 protein, and fewer activated CD11b+ and Iba1+ microglia. Isolated microglia demonstrated inflammatory gene upregulation to postnatal LPS without evidence of immune tolerance by prenatal LPS. CONCLUSION Prenatal LPS exposure induced immune tolerance to subsequent postnatal LPS exposure in the hippocampus. Microglia demonstrate a robust inflammatory response to postnatal LPS, but only a partial immune tolerance response.
Collapse
Affiliation(s)
- Garima Singh
- Division of Neonatology, Department of Pediatrics, University of Minnesota, East Building MB630, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA
| | - Bradley J Segura
- Division of Pediatric Surgery, Department of Surgery, University of Minnesota, East Building MB630, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, East Building MB630, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA
| | - Tate Gisslen
- Division of Neonatology, Department of Pediatrics, University of Minnesota, East Building MB630, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA.
| |
Collapse
|
27
|
Cho H, Kambhampati SP, Lai MJ, Zhou L, Lee G, Xie Y, Hui Q, Kannan RM, Duh EJ. Dendrimer-Triamcinolone Acetonide Reduces Neuroinflammation, Pathological Angiogenesis, and Neuroretinal Dysfunction in Ischemic Retinopathy. ADVANCED THERAPEUTICS 2021; 4:2000181. [PMID: 34527806 PMCID: PMC8436818 DOI: 10.1002/adtp.202000181] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Indexed: 12/11/2022]
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness in working-age adults. Severe visual loss in DR is primarily due to proliferative diabetic retinopathy, characterized by pathologic preretinal angiogenesis driven by retinal ischemia. Microglia, the resident immune cells of the retina, have emerged as a potentially important regulator of pathologic retinal angiogenesis. Corticosteroids including triamcinolone acetonide (TA), known for their antiangiogenic effects, are used in treating retinal diseases, but their use is significantly limited by side effects including cataracts and glaucoma. Generation-4 hydroxyl polyamidoamine dendrimer nanoparticles are utilized to deliver TA to activated microglia in the ischemic retina in a mouse model of oxygen-induced retinopathy (OIR). Following intravitreal injection, dendrimer-conjugated TA (D-TA) exhibits selective localization and sustained retention in activated microglia in disease-associated areas of the retina. D-TA, but not free TA, suppresses inflammatory cytokine production, microglial activation, and preretinal neovascularization in OIR. In addition, D-TA, but not free TA, ameliorates OIR-induced neuroretinal and visual dysfunction. These results indicate that activated microglia are a promising therapeutic target for retinal angiogenesis and neuroprotection in ischemic retinal diseases. Furthermore, dendrimer-based targeted therapy and specifically D-TA constitute a promising treatment approach for DR, offering increased and sustained drug efficacy with reduced side effects.
Collapse
Affiliation(s)
- Hongkwan Cho
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Siva P Kambhampati
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA; Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Michael J Lai
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Lingli Zhou
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Grace Lee
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Yangyiran Xie
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Qiaoyan Hui
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA
| | - Rangaramanujam M Kannan
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA; Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University Baltimore, MD 21218, USA
| | - Elia J Duh
- Department of Ophthalmology, School of Medicine Johns Hopkins University, Baltimore, MD 21231, USA; Center for Nanomedicine at the Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
28
|
Lorenzen K, Mathy NW, Whiteford ER, Eischeid A, Chen J, Behrens M, Chen XM, Shibata A. Microglia induce neurogenic protein expression in primary cortical cells by stimulating PI3K/AKT intracellular signaling in vitro. Mol Biol Rep 2021; 48:563-584. [PMID: 33387198 PMCID: PMC7884585 DOI: 10.1007/s11033-020-06092-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
Emerging evidence suggests that microglia can support neurogenesis. Little is known about the mechanisms by which microglia regulate the cortical environment and stimulate cortical neurogenesis. We used an in vitro co-culture model system to investigate the hypothesis that microglia respond to soluble signals from cortical cells, particularly following mechanical injury, to alter the cortical environment and promote cortical cell proliferation, differentiation, and survival. Analyses of cortical cell proliferation, cell death, neurogenic protein expression, and intracellular signaling were performed on uninjured and injured cortical cells in co-culture with microglial cell lines. Microglia soluble cues enhanced cortical cell viability and proliferation cortical cells. Co-culture of injured cortical cells with microglia significantly reduced cell death of cortical cells. Microglial co-culture significantly increased Nestin + and α-internexin + cortical cells. Multiplex ELISA and RT-PCR showed decreased pro-inflammatory cytokine production by microglia co-cultured with injured cortical cells. Inhibition of AKT phosphorylation in cortical cells blocked microglial-enhanced cortical cell viability and expression of neurogenic markers in vitro. This in vitro model system allows for assessment of the effect of microglial-derived soluble signals on cortical cell viability, proliferation, and stages of differentiation during homeostasis or following mechanical injury. These data suggest that microglia cells can downregulate inflammatory cytokine production following activation by mechanical injury to enhance proliferation of new cells capable of neurogenesis via activation of AKT intracellular signaling. Increasing our understanding of the mechanisms that drive microglial-enhanced cortical neurogenesis during homeostasis and following injury in vitro will provide useful information for future primary cell and in vivo studies.
Collapse
Affiliation(s)
- Kristi Lorenzen
- Biology Department, Creighton University, Omaha, NE, USA
- University of Nebraska Medical Center, Omaha, NE, USA
| | - Nicholas W Mathy
- Biology Department, Creighton University, Omaha, NE, USA
- Pediatric Medicine, St. Joseph Heritage Healthcare, Chino Hills, CA, USA
| | - Erin R Whiteford
- Biology Department, Creighton University, Omaha, NE, USA
- Pediatric Medicine, St. Joseph Heritage Healthcare, Chino Hills, CA, USA
| | - Alex Eischeid
- Biology Department, Creighton University, Omaha, NE, USA
- Stanford Hospital and Clinics, 300 Pasteur Dr, Stanford, CA, USA
| | - Jing Chen
- Biology Department, Creighton University, Omaha, NE, USA
- Pediatric Medicine, St. Joseph Heritage Healthcare, Chino Hills, CA, USA
| | - Matthew Behrens
- Biology Department, Creighton University, Omaha, NE, USA
- University of Nebraska College of Medicine, Omaha, NE, USA
| | - Xian-Ming Chen
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Creighton University, Omaha, NE, USA
| | | |
Collapse
|
29
|
Fatoba O, Itokazu T, Yamashita T. Microglia as therapeutic target in central nervous system disorders. J Pharmacol Sci 2020; 144:102-118. [PMID: 32921391 DOI: 10.1016/j.jphs.2020.07.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic microglial activation is associated with the pathogenesis of several CNS disorders. Microglia show phenotypic diversity and functional complexity in diseased CNS. Thus, understanding the pathology-specific heterogeneity of microglial behavior is crucial for the future development of microglia-modulating therapy for variety of CNS disorders. This review summarizes up-to-date knowledge on how microglia contribute to CNS homeostasis during development and throughout adulthood. We discuss the heterogeneity of microglial phenotypes in the context of CNS disorders with an emphasis on neurodegenerative diseases, demyelinating diseases, CNS trauma, and epilepsy. We conclude this review with a discussion about the disease-specific heterogeneity of microglial function and how it could be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Oluwaseun Fatoba
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
30
|
Uemura MT, Maki T, Ihara M, Lee VMY, Trojanowski JQ. Brain Microvascular Pericytes in Vascular Cognitive Impairment and Dementia. Front Aging Neurosci 2020; 12:80. [PMID: 32317958 PMCID: PMC7171590 DOI: 10.3389/fnagi.2020.00080] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Pericytes are unique, multi-functional mural cells localized at the abluminal side of the perivascular space in microvessels. Originally discovered in 19th century, pericytes had drawn less attention until decades ago mainly due to lack of specific markers. Recently, however, a growing body of evidence has revealed that pericytes play various important roles: development and maintenance of blood–brain barrier (BBB), regulation of the neurovascular system (e.g., vascular stability, vessel formation, cerebral blood flow, etc.), trafficking of inflammatory cells, clearance of toxic waste products from the brain, and acquisition of stem cell-like properties. In the neurovascular unit, pericytes perform these functions through coordinated crosstalk with neighboring cells including endothelial, glial, and neuronal cells. Dysfunction of pericytes contribute to a wide variety of diseases that lead to cognitive impairments such as cerebral small vessel disease (SVD), acute stroke, Alzheimer’s disease (AD), and other neurological disorders. For instance, in SVDs, pericyte degeneration leads to microvessel instability and demyelination while in stroke, pericyte constriction after ischemia causes a no-reflow phenomenon in brain capillaries. In AD, which shares some common risk factors with vascular dementia, reduction in pericyte coverage and subsequent microvascular impairments are observed in association with white matter attenuation and contribute to impaired cognition. Pericyte loss causes BBB-breakdown, which stagnates amyloid β clearance and the leakage of neurotoxic molecules into the brain parenchyma. In this review, we first summarize the characteristics of brain microvessel pericytes, and their roles in the central nervous system. Then, we focus on how dysfunctional pericytes contribute to the pathogenesis of vascular cognitive impairment including cerebral ‘small vessel’ and ‘large vessel’ diseases, as well as AD. Finally, we discuss therapeutic implications for these disorders by targeting pericytes.
Collapse
Affiliation(s)
- Maiko T Uemura
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,JSPS Overseas Research Fellowship Program, Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Virginia M Y Lee
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John Q Trojanowski
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
31
|
Balzano T, Arenas YM, Dadsetan S, Forteza J, Gil-Perotin S, Cubas-Nuñez L, Casanova B, Gracià F, Varela-Andrés N, Montoliu C, Llansola M, Felipo V. Sustained hyperammonemia induces TNF-a IN Purkinje neurons by activating the TNFR1-NF-κB pathway. J Neuroinflammation 2020; 17:70. [PMID: 32087723 PMCID: PMC7035786 DOI: 10.1186/s12974-020-01746-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with liver cirrhosis may develop hepatic encephalopathy. Rats with chronic hyperammonemia exhibit neurological alterations mediated by peripheral inflammation and neuroinflammation. Motor incoordination is due to increased TNF-a levels and activation of its receptor TNFR1 in the cerebellum. The aims were to assess (a) whether peripheral inflammation is responsible for TNF-a induction in hyperammonemic rats, (b) the cell type(s) in which TNF-a is increased, (c) whether this increase is associated with increased nuclear NF-κB and TNFR1 activation, (d) the time course of TNF-a induction, and (e) if TNF-a is induced in the Purkinje neurons of patients who die with liver cirrhosis. METHODS We analyzed the level of TNF-a mRNA and NF-κB in microglia, astrocytes, and Purkinje neurons in the cerebellum after 1, 2, and 4 weeks of hyperammonemia. We assessed whether preventing peripheral inflammation by administering an anti-TNF-a antibody prevents TNF-a induction. We tested whether TNF-a induction is reversed by R7050, which inhibits the TNFR1-NF-κB pathway, in ex vivo cerebellar slices. RESULTS Hyperammonemia induced microglial and astrocyte activation at 1 week. This was followed by TNF-a induction in both glial cell types at 2 weeks and in Purkinje neurons at 4 weeks. The level of TNF-a mRNA increased in parallel with the TNF-a protein level, indicating that TNF-a was synthesized in Purkinje cells. This increase was associated with increased NF-κB nuclear translocation. The nuclear translocation of NF-κB and the increase in TNF-a were reversed by R7050, indicating that they were mediated by the activation of TNFR1. Preventing peripheral inflammation with an anti-TNF-a antibody prevents TNF-a induction. CONCLUSION Sustained (4 weeks) but not short-term hyperammonemia induces TNF-a in Purkinje neurons in rats. This is mediated by peripheral inflammation. TNF-a is also increased in the Purkinje neurons of patients who die with liver cirrhosis. The results suggest that hyperammonemia induces TNF-a in glial cells and that TNF-a released by glial cells activates TNFR1 in Purkinje neurons, leading to NF-κB nuclear translocation and the induction of TNF-a expression, which may contribute to the neurological alterations observed in hyperammonemia and hepatic encephalopathy.
Collapse
Affiliation(s)
- Tiziano Balzano
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Yaiza M Arenas
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Sherry Dadsetan
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Jerónimo Forteza
- Instituto Valenciano de Patología, Unidad Mixta de Patología Molecular, Centro Investigación Príncipe Felipe/Universidad Católica de Valencia, Valencia, Spain
| | - Sara Gil-Perotin
- Multiple Sclerosis and Neuroimmunology Research Group, Fundación para la Investigación La Fe, Valencia, Spain
| | - Laura Cubas-Nuñez
- Multiple Sclerosis and Neuroimmunology Research Group, Fundación para la Investigación La Fe, Valencia, Spain
| | - Bonaventura Casanova
- Multiple Sclerosis and Neuroimmunology Research Group, Fundación para la Investigación La Fe, Valencia, Spain
| | - Francisco Gracià
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Natalia Varela-Andrés
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Carmina Montoliu
- Instituto de Investigacion Sanitaria INCLIVA, Hospital Clinico de Valencia, Valencia, Spain
| | - Marta Llansola
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain.
| |
Collapse
|
32
|
Jefferson B, Ali M, Grant S, Frampton G, Ploof M, Andry S, DeMorrow S, McMillin M. Thrombospondin-1 Exacerbates Acute Liver Failure and Hepatic Encephalopathy Pathology in Mice by Activating Transforming Growth Factor β1. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:347-357. [PMID: 31734229 PMCID: PMC7013272 DOI: 10.1016/j.ajpath.2019.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/28/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022]
Abstract
Severe hepatic insults can lead to acute liver failure and hepatic encephalopathy (HE). Transforming growth factor β1 (TGFβ1) has been shown to contribute to HE during acute liver failure; however, TGFβ1 must be activated to bind its receptor and generate downstream effects. One protein that can activate TGFβ1 is thrombospondin-1 (TSP-1). Therefore, the aim of this study was to assess TSP-1 during acute liver failure and HE pathogenesis. C57Bl/6 or TSP-1 knockout (TSP-1-/-) mice were injected with azoxymethane (AOM) to induce acute liver failure and HE. Liver damage, neurologic decline, and molecular analyses of TSP-1 and TGFβ1 signaling were performed. AOM-treated mice had increased TSP-1 and TGFβ1 mRNA and protein expression in the liver. TSP-1-/- mice administered AOM had reduced liver injury as assessed by histology and serum transaminase levels compared with C57Bl/6 AOM-treated mice. TSP-1-/- mice treated with AOM had reduced TGFβ1 signaling that was associated with less hepatic cell death as assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and cleaved caspase 3 expression. TSP-1-/- AOM-treated mice had a reduced rate of neurologic decline, less cerebral edema, and a decrease in microglia activation in comparison with C57Bl/6 mice treated with AOM. Taken together, TSP-1 is an activator of TGFβ1 signaling during AOM-induced acute liver failure and contributes to both liver pathology and HE progression.
Collapse
Affiliation(s)
| | - Malaika Ali
- Central Texas Veterans Health Care System, Austin, Texas
| | - Stephanie Grant
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas; Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Gabriel Frampton
- Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas; Department of Internal Medicine, The University of Texas at Austin Dell Medical School, Austin, Texas
| | - Michaela Ploof
- Central Texas Veterans Health Care System, Austin, Texas
| | - Sarah Andry
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Sharon DeMorrow
- Central Texas Veterans Health Care System, Austin, Texas; Department of Medical Physiology, Texas A&M University Health Science Center, Temple, Texas; Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, The University of Texas at Austin Dell Medical School, Austin, Texas
| | - Matthew McMillin
- Central Texas Veterans Health Care System, Austin, Texas; Department of Internal Medicine, The University of Texas at Austin Dell Medical School, Austin, Texas.
| |
Collapse
|
33
|
Dominant Role of the Gut Microbiota in Chemotherapy Induced Neuropathic Pain. Sci Rep 2019; 9:20324. [PMID: 31889131 PMCID: PMC6937259 DOI: 10.1038/s41598-019-56832-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023] Open
Abstract
Chemotherapy induced peripheral neuropathy (CIPN), a toxic side effect of some cancer treatments, negatively impacts patient outcomes and drastically reduces survivor’s quality of life (QOL). Uncovering the mechanisms driving chemotherapy-induced CIPN is urgently needed to facilitate the development of effective treatments, as currently there are none. Observing that C57BL/6 (B6) and 129SvEv (129) mice are respectively sensitive and resistant to Paclitaxel-induced pain, we investigated the involvement of the gut microbiota in this extreme phenotypic response. Reciprocal gut microbiota transfers between B6 and 129 mice as well as antibiotic depletion causally linked gut microbes to Paclitaxel-induced pain sensitivity and resistance. Microglia proliferated in the spinal cords of Paclitaxel treated mice harboring the pain-sensitive B6 microbiota but not the pain-resistant 129 microbiota, which exhibited a notable absence of infiltrating immune cells. Paclitaxel decreased the abundance of Akkermansia muciniphila, which could compromise barrier integrity resulting in systemic exposure to bacterial metabolites and products – that acting via the gut-immune-brain axis – could result in altered brain function. Other bacterial taxa that consistently associated with both bacteria and pain as well as microglia and pain were identified, lending support to our hypothesis that microglia are causally involved in CIPN, and that gut bacteria are drivers of this phenotype.
Collapse
|
34
|
Jaeger V, DeMorrow S, McMillin M. The Direct Contribution of Astrocytes and Microglia to the Pathogenesis of Hepatic Encephalopathy. J Clin Transl Hepatol 2019; 7:352-361. [PMID: 31915605 PMCID: PMC6943208 DOI: 10.14218/jcth.2019.00025] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/07/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatic encephalopathy is a neurological complication resulting from loss of hepatic function and is associated with poor clinical outcomes. During acute liver failure over 20% of mortality can be associated with the development of hepatic encephalopathy. In patients with liver cirrhosis, 1-year survival for those that develop overt hepatic encephalopathy is under 50%. The pathogenesis of hepatic encephalopathy is complicated due to the multiple disruptions in homeostasis that occur following a reduction in liver function. Of these, elevations of ammonia and neuroinflammation have been shown to play a significant contributing role to the development of hepatic encephalopathy. Disruption of the urea cycle following liver dysfunction leads to elevations of circulating ammonia, which enter the brain and disrupt the functioning of astrocytes. This results in dysregulation of metabolic pathways in astrocytes, oxidative stress and cerebral edema. Besides ammonia, circulating chemokines and cytokines are increased following liver injury, leading to activation of microglia and a subsequent neuroinflammatory response. The combination of astrocyte dysfunction and microglia activation are significant contributing factors to the pathogenesis of hepatic encephalopathy.
Collapse
Affiliation(s)
- Victoria Jaeger
- Baylor Scott & White Health, Department of Internal Medicine, Temple, TX, USA
| | - Sharon DeMorrow
- Texas A&M University Health Science Center, Department of Medical Physiology, Temple, TX, USA
- Central Texas Veterans Health Care System, Temple, TX, USA
- University of Texas at Austin, Dell Medical School, Department of Internal Medicine, Austin, TX, USA
- University of Texas at Austin, College of Pharmacy, Austin, TX, USA
| | - Matthew McMillin
- Texas A&M University Health Science Center, Department of Medical Physiology, Temple, TX, USA
- Central Texas Veterans Health Care System, Temple, TX, USA
- University of Texas at Austin, Dell Medical School, Department of Internal Medicine, Austin, TX, USA
- Correspondence to: Matthew McMillin, University of Texas at Austin Dell Medical School, 1601 Trinity Street, Building B, Austin, TX 78701, USA. Tel: +1-512-495-5037, Fax: +1-512-495-5839, E-mail:
| |
Collapse
|
35
|
Pang H, Ren Y, Li H, Chen C, Zheng X. LncRNAs linc00311 and AK141205 are identified as new regulators in STAT3-mediated neuropathic pain in bCCI rats. Eur J Pharmacol 2019; 868:172880. [PMID: 31863767 DOI: 10.1016/j.ejphar.2019.172880] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/11/2019] [Accepted: 12/17/2019] [Indexed: 12/30/2022]
Abstract
Neuropathic pain is a severe disease caused by lesions or diseases in the somatosensory system. Long non-coding RNAs (lncRNAs) are important in the development and maintenance of neuropathic pain. However, the precise role of lncRNAs in regulating neuropathic pain remains largely unknown. In this study, a rat model of bilateral chronic constriction injury (bCCI) was established, and microarray was applied to analyze differentially expressed lncRNAs among sham group, bCCI group and the experimental group (bCCI rats administrated with a specific STAT3 inhibitor WP1066). Linc00311 and lncRNA-AK141205 were uncharacterized lncRNAs both upregulated by > 2 folds in bCCI model compared with Sham group, and they were downregulated by > 2 folds following WP1066 administration compared with bCCI group. Downregulation of linc00311 and lncRNA-AK141205 by specific siRNAs significantly attenuated mechanical allodynia, thermal and cold hyperalgesia in bCCI rats. In addition, inhibition of linc00311 and lncRNA-AK141205 inactivated the signal transducer and activator of transcription 3 (STAT3) signaling in spinal microglia in vivo and in vitro. Inhibition of linc00311 and lncRNA-AK141205 could reduce activation of STAT3 and production of proinflammatory cytokines. Moreover, activating STAT3 with SD19 could antagonize the effect of the suppressive effect of siRNAs on production of proinflammatory cytokines. Hence, it is likely that silencing linc00311 and lncRNA-AK141205 may be a promising and novel treatment for neuropathic pain.
Collapse
Affiliation(s)
- Hongli Pang
- Department of Anesthesiology, The First Affiliated Hospital, Henan University, Kaifeng, 475001, China
| | - Yifeng Ren
- Department of Anesthesiology, The First Affiliated Hospital, Henan University, Kaifeng, 475001, China
| | - Huifang Li
- Department of Anesthesiology, The First Affiliated Hospital, Henan University, Kaifeng, 475001, China
| | - Chengzhe Chen
- Department of Anesthesiology, The First Affiliated Hospital, Henan University, Kaifeng, 475001, China
| | - Xiaozhen Zheng
- Department of Anesthesiology, The First Affiliated Hospital, Henan University, Kaifeng, 475001, China.
| |
Collapse
|
36
|
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: 18] [Impact Index Per Article: 3.0] [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
|
37
|
Cabrera-Pastor A, Llansola M, Montoliu C, Malaguarnera M, Balzano T, Taoro-Gonzalez L, García-García R, Mangas-Losada A, Izquierdo-Altarejos P, Arenas YM, Leone P, Felipo V. Peripheral inflammation induces neuroinflammation that alters neurotransmission and cognitive and motor function in hepatic encephalopathy: Underlying mechanisms and therapeutic implications. Acta Physiol (Oxf) 2019; 226:e13270. [PMID: 30830722 DOI: 10.1111/apha.13270] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022]
Abstract
Several million patients with liver cirrhosis suffer minimal hepatic encephalopathy (MHE), with mild cognitive and coordination impairments that reduce their quality of life and life span. Hyperammonaemia and peripheral inflammation act synergistically to induce these neurological alterations. We propose that MHE appearance is because of the changes in peripheral immune system, which are transmitted to brain, leading to neuroinflammation that alters neurotransmission leading to cognitive and motor alterations. We summarize studies showing that MHE in cirrhotic patients is associated with alterations in the immune system and that patients died with HE show neuroinflammation in cerebellum, with microglial and astrocytic activation and Purkinje cell loss. We also summarize studies in animal models of MHE on the role of peripheral inflammation in neuroinflammation induction, how neuroinflammation alters neurotransmission and how this leads to cognitive and motor alterations. These studies identify therapeutic targets and treatments that improve cognitive and motor function. Rats with MHE show neuroinflammation in hippocampus and altered NMDA and AMPA receptor membrane expression, which impairs spatial learning and memory. Neuroinflammation in cerebellum is associated with altered GABA transporters and extracellular GABA, which impair motor coordination and learning in a Y maze. These alterations are reversed by treatments that reduce peripheral inflammation (anti-TNFα, ibuprofen), neuroinflammation (sulphoraphane, p38 inhibitors), GABAergic tone (bicuculline, pregnenolone sulphate) or increase extracellular cGMP (sildenafil or cGMP). The mechanisms identified would also occur in other chronic diseases associated with inflammation, aging and some mental and neurodegenerative diseases. Treatments that improve MHE may also be beneficial to treat these pathologies.
Collapse
Affiliation(s)
- Andrea Cabrera-Pastor
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
- Fundacion Investigacion Hospital Clinico Valencia, INCLIVA, Valencia, Spain
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Carmina Montoliu
- Fundacion Investigacion Hospital Clinico Valencia, INCLIVA, Valencia, Spain
| | - Michele Malaguarnera
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Tiziano Balzano
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Lucas Taoro-Gonzalez
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Raquel García-García
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Alba Mangas-Losada
- Fundacion Investigacion Hospital Clinico Valencia, INCLIVA, Valencia, Spain
| | | | - Yaiza M Arenas
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Paola Leone
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| |
Collapse
|
38
|
McMillin M, Grant S, Frampton G, Petrescu AD, Williams E, Jefferson B, Thomas A, Brahmaroutu A, DeMorrow S. Elevated circulating TGFβ1 during acute liver failure activates TGFβR2 on cortical neurons and exacerbates neuroinflammation and hepatic encephalopathy in mice. J Neuroinflammation 2019; 16:69. [PMID: 30940161 PMCID: PMC6446280 DOI: 10.1186/s12974-019-1455-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Acute liver failure resulting from drug-induced liver injury can lead to the development of neurological complications called hepatic encephalopathy (HE). Hepatic transforming growth factor beta 1 (TGFβ1) is upregulated due to liver failure in mice and inhibiting circulating TGFβ reduced HE progression. However, the specific contributions of TGFβ1 on brain cell populations and neuroinflammation during HE are not known. Therefore, the aim of this study was to characterize hepatic and brain TGFβ1 signaling during acute liver failure and its contribution to HE progression using a combination of pharmacological and genetic approaches. METHODS C57Bl/6 or neuron-specific transforming growth factor beta receptor 2 (TGFβR2) null mice (TGFβR2ΔNeu) were treated with azoxymethane (AOM) to induce acute liver failure and HE. The activity of circulating TGFβ1 was inhibited in C57Bl/6 mice via injection of a neutralizing antibody against TGFβ1 (anti-TGFβ1) prior to AOM injection. In all mouse treatment groups, liver damage, neuroinflammation, and neurological deficits were assessed. Inflammatory signaling between neurons and microglia were investigated in in vitro studies through the use of pharmacological inhibitors of TGFβ1 signaling in HT-22 and EOC-20 cells. RESULTS TGFβ1 was expressed and upregulated in the liver following AOM injection. Pharmacological inhibition of TGFβ1 after AOM injection attenuated neurological decline, microglia activation, and neuroinflammation with no significant changes in liver damage. TGFβR2ΔNeu mice administered AOM showed no effect on liver pathology but significantly reduced neurological decline compared to control mice. Microglia activation and neuroinflammation were attenuated in mice with pharmacological inhibition of TGFβ1 or in TGFβR2ΔNeu mice. TGFβ1 increased chemokine ligand 2 (CCL2) and decreased C-X3-C motif ligand 1 (CX3CL1) expression in HT-22 cells and reduced interleukin-1 beta (IL-1ß) expression, tumor necrosis factor alpha (TNFα) expression, and phagocytosis activity in EOC-20 cells. CONCLUSION Increased circulating TGFβ1 following acute liver failure results in activation of neuronal TGFβR2 signaling, driving neuroinflammation and neurological decline during AOM-induced HE.
Collapse
Affiliation(s)
- Matthew McMillin
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Stephanie Grant
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Gabriel Frampton
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Anca D Petrescu
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Elaina Williams
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA.,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Brandi Jefferson
- Central Texas Veterans Health Care System, Temple, TX, USA.,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA
| | - Alison Thomas
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA
| | - Ankita Brahmaroutu
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA
| | - Sharon DeMorrow
- Central Texas Veterans Health Care System, Temple, TX, USA. .,Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, USA. .,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA. .,Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
39
|
González-Regueiro J, la Tijera MHD, Moreno-Alcántar R, Torre A. Pathophysiology of hepatic encephalopathy and future treatment options. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO (ENGLISH EDITION) 2019. [DOI: 10.1016/j.rgmxen.2019.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
40
|
González-Regueiro JA, Higuera-de la Tijera MF, Moreno-Alcántar R, Torre A. Pathophysiology of hepatic encephalopathy and future treatment options. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2019; 84:195-203. [PMID: 31014748 DOI: 10.1016/j.rgmx.2019.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/28/2019] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
Understanding of the pathophysiology of hepatic encephalopathy has conditioned new treatment options. Ammonia detoxification in hepatic encephalopathy is regulated by two enzymes: glutaminase or glutamine synthetase. The first produces ammonia and the second detoxifies the ammonia, which is why treatments are aimed at glutaminase inhibition or glutamine synthetase activation. At present, we know that both enzymes are found not only in the liver, but also in the muscle, intestine, kidney, and brain. Therefore, current treatments can be directed at each enzyme at different sites. Awareness of those potential treatment sites makes different options of approach possible in the patient with hepatic encephalopathy, and each approach should be personalized.
Collapse
Affiliation(s)
- J A González-Regueiro
- Departamento de Gastroenterología, Instituto Nacional de Ciencias Médicas y Nutrición «Salvador Zubirán», Ciudad de México, México
| | | | - R Moreno-Alcántar
- Departamento de Gastroenterología, Centro Médico Nacional Siglo XXI, Ciudad de México, México
| | - A Torre
- Departamento de Gastroenterología, Instituto Nacional de Ciencias Médicas y Nutrición «Salvador Zubirán», Ciudad de México, México; Unidad de Hepatología y Trasplante Hepático, Departamento de Gastroenterología, Instituto Nacional de Ciencias Médicas y Nutrición «Salvador Zubirán», Ciudad de México, México.
| |
Collapse
|