1
|
Lin YE, Lin HP, Lu KH, Huang YJ, Panyod S, Liu WT, Lu YS, Chen MH, Sheen LY. Cordyceps militaris and Armillaria mellea formula alleviates depressive behaviors via microglia regulation in an unpredictable chronic mild stress animal model. J Tradit Complement Med 2025; 15:24-35. [PMID: 39807265 PMCID: PMC11725130 DOI: 10.1016/j.jtcme.2024.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 01/16/2025] Open
Abstract
Background and aim Cordyceps militaris (CM) and Armillaria mellea (AM) are medicinal mushrooms with potential applications in the treatment of mood disorders, including depression and anxiety. While research suggests that both CM and AM possess anti-inflammatory properties and hold potential for treating depression when administered separately, there is limited knowledge about their efficacy when combined in a formula, as well as the underlying mechanism involving the modulation of microglia. Experimental procedure Rats received oral administrations of the low-dose formulation, medium-dose formulation, and high-dose formulation over 28 consecutive days as part of the UCMS protocols. The concentrations of serotonin, dopamine, and the corresponding metabolites in the rat prefrontal cortex and hippocampus were assessed. Blood samples were collected to examine corticosterone levels, and the brains were dissected for evaluating activated microglia morphologies and associated pro- and anti-inflammatory signaling pathways. Results and conclusion The CM-AM formula effectively averted abnormal behaviors triggered by UCMS, such as anhedonia and hypoactivity, and decreased the turnover rate of monoamines in both the prefrontal cortex and hippocampus. The formula mitigated the increase in serum corticosterone levels induced by chronic stress. Furthermore, the formula alleviated stress-induced microglia activation in the hippocampus, achieving this by down-regulating hyperactivated pro-inflammatory proteins and up-regulating hypoactivated anti-inflammatory proteins in the hippocampus. The antidepressant-like effects potentially stemming from the regulation of neurotransmitters and immunomodulation, likely by restoring the balance of M1 and M2 microglia fractions in the hippocampus. Consequently, the CM-AM formula could be explored as a prospective complementary and alternative therapy for depression.
Collapse
Affiliation(s)
- Yu-En Lin
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Hui-Ping Lin
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Kuan-Hung Lu
- Institute of Food Safety and Health, National Taiwan University, Taipei, Taiwan
- Institute of Environmental and Occupational Health Sciences, National Taiwan University, Taipei, Taiwan
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Suraphan Panyod
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Wei-Ting Liu
- Taiwan Agricultural Research Institute, Council of Agricultural, Taichung, Taiwan
| | - Yun-Sheng Lu
- Taiwan Agricultural Research Institute, Council of Agricultural, Taichung, Taiwan
| | - Mei-Hsing Chen
- Taiwan Agricultural Research Institute, Council of Agricultural, Taichung, Taiwan
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan
- National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
2
|
Maya-Arteaga JP, Martínez-Orozco H, Diaz-Cintra S. MorphoGlia, an interactive method to identify and map microglia morphologies, demonstrates differences in hippocampal subregions of an Alzheimer's disease mouse model. Front Cell Neurosci 2024; 18:1505048. [PMID: 39698052 PMCID: PMC11653188 DOI: 10.3389/fncel.2024.1505048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Microglia are dynamic central nervous system cells crucial for maintaining homeostasis and responding to neuroinflammation, as evidenced by their varied morphologies. Existing morphology analysis often fails to detect subtle variations within the full spectrum of microglial morphologies due to their reliance on predefined categories. Here, we present MorphoGlia, an interactive, user-friendly pipeline that objectively characterizes microglial morphologies. MorphoGlia employs a machine learning ensemble to select relevant morphological features of microglia cells, perform dimensionality reduction, cluster these features, and subsequently map the clustered cells back onto the tissue, providing a spatial context for the identified microglial morphologies. We applied this pipeline to compare the responses between saline solution (SS) and scopolamine (SCOP) groups in a SCOP-induced mouse model of Alzheimer's disease, with a specific focus on the hippocampal subregions CA1 and Hilus. Next, we assessed microglial morphologies across four groups: SS-CA1, SCOP-CA1, SS-Hilus, and SCOP-Hilus. The results demonstrated that MorphoGlia effectively differentiated between SS and SCOP-treated groups, identifying distinct clusters of microglial morphologies commonly associated with pro-inflammatory states in the SCOP groups. Additionally, MorphoGlia enabled spatial mapping of these clusters, identifying the most affected hippocampal layers. This study highlights MorphoGlia's capability to provide unbiased analysis and clustering of microglial morphological states, making it a valuable tool for exploring microglial heterogeneity and its implications for central nervous system pathologies.
Collapse
Affiliation(s)
| | | | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Santiago de Querétaro, Mexico
| |
Collapse
|
3
|
Niemeyer CS, Merle L, Bubak AN, Baxter BD, Gentile Polese A, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and trigeminal routes of HSV-1 CNS infection with regional microglial heterogeneity. J Virol 2024; 98:e0096824. [PMID: 39475273 PMCID: PMC11575344 DOI: 10.1128/jvi.00968-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/29/2024] [Indexed: 11/06/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal ganglion (TG) and other peripheral ganglia. HSV-1 can also infect and become latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem and other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases.IMPORTANCEThis study shows how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveal the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigens. Previous reports have identified specific brain regions found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the brain regions consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Christy S Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Andrew N Bubak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - B Dnate' Baxter
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Arianna Gentile Polese
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katherine Colon-Reyes
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sandy Vang
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James E Hassell
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kimberley D Bruce
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria A Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
4
|
Niemeyer CS, Merle L, Bubak AN, Dnate' Baxter B, Polese AG, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and Trigeminal Routes of HSV-1 CNS Infection with Regional Microglial Heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614340. [PMID: 39386674 PMCID: PMC11463476 DOI: 10.1101/2024.09.22.614340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal and other peripheral ganglia (TG). HSV-1 can also infect and go latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE Infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem, as well as other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus, while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases. Importance This study sheds light on how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveals the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigen. Previous reports have identified specific regions of the brain found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the regions of the brain consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
|
5
|
Hoyer-Kimura C, Hay M, Konhilas JP, Morrison HW, Methajit M, Strom J, Polt R, Salcedo V, Fricks JP, Kalya A, Pires PW. PNA5, A Novel Mas Receptor Agonist, Improves Neurovascular and Blood-Brain-Barrier Function in a Mouse Model of Vascular Cognitive Impairment and Dementia. Aging Dis 2024; 15:1927-1951. [PMID: 37815905 PMCID: PMC11272189 DOI: 10.14336/ad.2023.0928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
It is well established that decreased brain blood flow, increased reactive oxygen species production (ROS), and pro-inflammatory mechanisms accelerate neurodegenerative disease progressions, including vascular cognitive impairment and dementia (VCID). Previous studies in our laboratory have shown that our novel glycosylated Angiotensin-(1-7) Mas receptor agonist PNA5 reverses cognitive deficits, decreases ROS production, and inhibits inflammatory cytokine production in our preclinical mouse model of VCID that is induced by chronic heart failure (VCID-HF). In the present study, the effects of VCID-HF and treatment with PNA5 on microglia activation, blood-brain-barrier (BBB) integrity, and neurovascular coupling were assessed in our mouse model of VCID-HF. Three-month-old male C57BL/6J mice were subjected to myocardial infarction (MI) to induce heart failure for four weeks and then treated with subcutaneous injections of extended-release PNA5. Microglia activation, BBB permeability, cerebral perfusion, and neurovascular coupling were assessed. Results show that in our VCID-HF model, there was an increase in microglial activation and recruitment within the CA1 and CA3 regions of the hippocampus, a disruption in BBB integrity, and a decrease in neurovascular coupling. Treatment with PNA5 reversed these neuropathological effects of VCID-HF, suggesting that PNA5 may be an effective disease-modifying therapy to treat and prevent VCID. This study identifies potential mechanisms by which heart failure may induce VCID and highlights the possible mechanisms by which treatment with our novel glycosylated Angiotensin-(1-7) Mas receptor agonist, PNA5, may protect cognitive function in our model of VCID.
Collapse
Affiliation(s)
| | - Meredith Hay
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
- Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ 85724, USA.
- ProNeurogen, Inc, Tucson, AZ, USA
| | - John P Konhilas
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | - Helena W Morrison
- College of Nursing, The University of Arizona, Tucson, AZ 85724, USA.
| | - Methawasin Methajit
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85724, USA.
| | - Joshua Strom
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85724, USA.
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85724, USA.
| | - Victoria Salcedo
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | | | - Anjna Kalya
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| | - Paulo W Pires
- Department of Physiology, The University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
6
|
Sciarretta F, Zaccaria F, Ninni A, Ceci V, Turchi R, Apolloni S, Milani M, Della Valle I, Tiberi M, Chiurchiù V, D'Ambrosi N, Pedretti S, Mitro N, Volontè C, Amadio S, Aquilano K, Lettieri-Barbato D. Frataxin deficiency shifts metabolism to promote reactive microglia via glucose catabolism. Life Sci Alliance 2024; 7:e202402609. [PMID: 38631900 PMCID: PMC11024345 DOI: 10.26508/lsa.202402609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Immunometabolism investigates the intricate relationship between the immune system and cellular metabolism. This study delves into the consequences of mitochondrial frataxin (FXN) depletion, the primary cause of Friedreich's ataxia (FRDA), a debilitating neurodegenerative condition characterized by impaired coordination and muscle control. By using single-cell RNA sequencing, we have identified distinct cellular clusters within the cerebellum of an FRDA mouse model, emphasizing a significant loss in the homeostatic response of microglial cells lacking FXN. Remarkably, these microglia deficient in FXN display heightened reactive responses to inflammatory stimuli. Furthermore, our metabolomic analyses reveal a shift towards glycolysis and itaconate production in these cells. Remarkably, treatment with butyrate counteracts these immunometabolic changes, triggering an antioxidant response via the itaconate-Nrf2-GSH pathways and suppressing the expression of inflammatory genes. Furthermore, we identify Hcar2 (GPR109A) as a mediator involved in restoring the homeostasis of microglia without FXN. Motor function tests conducted on FRDA mice underscore the neuroprotective attributes of butyrate supplementation, enhancing neuromotor performance. In conclusion, our findings elucidate the role of disrupted homeostatic function in cerebellar microglia in the pathogenesis of FRDA. Moreover, they underscore the potential of butyrate to mitigate inflammatory gene expression, correct metabolic imbalances, and improve neuromotor capabilities in FRDA.
Collapse
Affiliation(s)
- Francesca Sciarretta
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| | - Fabio Zaccaria
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Evolutionary Biology and Ecology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| | - Andrea Ninni
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Evolutionary Biology and Ecology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| | - Veronica Ceci
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Evolutionary Biology and Ecology, University of Rome Tor Vergata, Rome, Italy
| | - Riccardo Turchi
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Savina Apolloni
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Martina Milani
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Cellular and Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | - Ilaria Della Valle
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- PhD Program in Cellular and Molecular Biology, University of Rome Tor Vergata, Rome, Italy
| | - Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
- Institute of Translational Pharmacology, IFT-CNR, Rome, Italy
| | - Nadia D'Ambrosi
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Pedretti
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy
| | - Nico Mitro
- DiSFeB, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Cinzia Volontè
- National Research Council, Institute for Systems Analysis and Computer Science "A. Ruberti", Rome, Italy
- Santa Lucia Foundation IRCCS, Experimental Neuroscience and Neurological Disease Models, Rome, Italy
| | - Susanna Amadio
- Santa Lucia Foundation IRCCS, Experimental Neuroscience and Neurological Disease Models, Rome, Italy
| | - Katia Aquilano
- Department Biology, University of Rome Tor Vergata, Rome, Italy
| | - Daniele Lettieri-Barbato
- Department Biology, University of Rome Tor Vergata, Rome, Italy
- IRCCS Fondazione Bietti, Rome, Italy
| |
Collapse
|
7
|
Dabrowska S, Turano E, Scambi I, Virla F, Nodari A, Pezzini F, Galiè M, Bonetti B, Mariotti R. A Cellular Model of Amyotrophic Lateral Sclerosis to Study the Therapeutic Effects of Extracellular Vesicles from Adipose Mesenchymal Stem Cells on Microglial Activation. Int J Mol Sci 2024; 25:5707. [PMID: 38891895 PMCID: PMC11171908 DOI: 10.3390/ijms25115707] [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: 03/19/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of upper and lower motor neurons (MNs) in the brain and spinal cord, leading to progressive paralysis and death. Increasing evidence indicates that neuroinflammation plays an important role in ALS's pathogenesis and disease progression. Neuroinflammatory responses, primarily driven by activated microglia and astrocytes, and followed by infiltrating peripheral immune cells, contribute to exacerbate/accelerate MN death. In particular, the role of the microglia in ALS remains unclear, partly due to the lack of experimental models that can fully recapitulate the complexity of ALS's pathology. In this study, we developed and characterized a microglial cell line, SIM-A9-expressing human mutant protein Cu+/Zn+ superoxide dismutase_1 (SIM-A9hSOD1(G93A)), as a suitable model in vitro mimicking the microglia activity in ALS. The expression of hSOD1(G93A) in SIM-A9 cells induced a change in their metabolic activity, causing polarization into a pro-inflammatory phenotype and enhancing reactive oxygen species production, which is known to activate cell death processes and apoptosis. Afterward, we used our microglial model as an experimental set-up to investigate the therapeutic action of extracellular vesicles isolated from adipose mesenchymal stem cells (ASC-EVs). ASC-EVs represent a promising therapeutic treatment for ALS due to their neuroprotective and immunomodulatory properties. Here, we demonstrated that treatment with ASC-EVs is able to modulate activated ALS microglia, reducing their metabolic activity and polarizing their phenotype toward an anti-inflammatory one through a mechanism of reduction of reactive oxygen species.
Collapse
Affiliation(s)
- Sylwia Dabrowska
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego Street 5, 02-106 Warsaw, Poland
| | - Ermanna Turano
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Ilaria Scambi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Federica Virla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Alice Nodari
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Francesco Pezzini
- Department of Surgery, Dentistry, Paediatrics and Gynaecology (Child Neurology and Psychiatry), University of Verona, 37134 Verona, Italy;
| | - Mirco Galiè
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| | - Bruno Bonetti
- Neurology Unit, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy;
| | - Raffaella Mariotti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (S.D.); (E.T.); (I.S.); (F.V.); (A.N.); (M.G.)
| |
Collapse
|
8
|
Jithoo A, Penny TR, Pham Y, Sutherland AE, Smith MJ, Petraki M, Fahey MC, Jenkin G, Malhotra A, Miller SL, McDonald CA. The Temporal Relationship between Blood-Brain Barrier Integrity and Microglial Response following Neonatal Hypoxia Ischemia. Cells 2024; 13:660. [PMID: 38667275 PMCID: PMC11049639 DOI: 10.3390/cells13080660] [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: 02/29/2024] [Revised: 04/05/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
Blood-brain barrier (BBB) dysfunction and neuroinflammation are key mechanisms of brain injury. We performed a time-course study following neonatal hypoxia-ischemia (HI) to characterize these events. HI brain injury was induced in postnatal day 10 rats by single carotid artery ligation followed by hypoxia (8% oxygen, 90 min). At 6, 12, 24, and 72 h (h) post-HI, brains were collected to assess neuropathology and BBB dysfunction. A significant breakdown of the BBB was observed in the HI injury group compared to the sham group from 6 h in the cortex and hippocampus (p < 0.001), including a significant increase in albumin extravasation (p < 0.0033) and decrease in basal lamina integrity and tight-junction proteins. There was a decrease in resting microglia (p < 0.0001) transitioning to an intermediate state from as early as 6 h post-HI, with the intermediate microglia peaking at 12 h (p < 0.0001), which significantly correlated to the peak of microbleeds. Neonatal HI insult leads to significant brain injury over the first 72 h that is mediated by BBB disruption within 6 h and a transitioning state of the resident microglia. Key BBB events coincide with the appearance of the intermediate microglial state and this relationship warrants further research and may be a key target for therapeutic intervention.
Collapse
Affiliation(s)
- Arya Jithoo
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Tayla R. Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Amy E. Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Madeleine J. Smith
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Maria Petraki
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
| | - Michael C. Fahey
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia;
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Atul Malhotra
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia;
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Courtney A. McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (A.J.); (T.R.P.); (Y.P.); (A.E.S.); (M.J.S.); (G.J.); (A.M.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
9
|
Virla F, Turano E, Scambi I, Schiaffino L, Boido M, Mariotti R. Administration of adipose-derived stem cells extracellular vesicles in a murine model of spinal muscular atrophy: effects of a new potential therapeutic strategy. Stem Cell Res Ther 2024; 15:94. [PMID: 38561840 PMCID: PMC10986013 DOI: 10.1186/s13287-024-03693-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Spinal Muscular Atrophy (SMA) is an autosomal-recessive neuromuscular disease affecting children. It is caused by the mutation or deletion of the survival motor neuron 1 (SMN1) gene resulting in lower motor neuron (MN) degeneration followed by motor impairment, progressive skeletal muscle paralysis and respiratory failure. In addition to the already existing therapies, a possible combinatorial strategy could be represented by the use of adipose-derived mesenchymal stem cells (ASCs) that can be obtained easily and in large amounts from adipose tissue. Their efficacy seems to be correlated to their paracrine activity and the production of soluble factors released through extracellular vesicles (EVs). EVs are important mediators of intercellular communication with a diameter between 30 and 100 nm. Their use in other neurodegenerative disorders showed a neuroprotective effect thanks to the release of their content, especially proteins, miRNAs and mRNAs. METHODS In this study, we evaluated the effect of EVs isolated from ASCs (ASC-EVs) in the SMNΔ7 mice, a severe SMA model. With this purpose, we performed two administrations of ASC-EVs (0.5 µg) in SMA pups via intracerebroventricular injections at post-natal day 3 (P3) and P6. We then assessed the treatment efficacy by behavioural test from P2 to P10 and histological analyses at P10. RESULTS The results showed positive effects of ASC-EVs on the disease progression, with improved motor performance and a significant delay in spinal MN degeneration of treated animals. ASC-EVs could also reduce the apoptotic activation (cleaved Caspase-3) and modulate the neuroinflammation with an observed decreased glial activation in lumbar spinal cord, while at peripheral level ASC-EVs could only partially limit the muscular atrophy and fiber denervation. CONCLUSIONS Our results could encourage the use of ASC-EVs as a therapeutic combinatorial treatment for SMA, bypassing the controversial use of stem cells.
Collapse
Affiliation(s)
- Federica Virla
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Ermanna Turano
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Ilaria Scambi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Lorenzo Schiaffino
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Boido
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Raffaella Mariotti
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
10
|
Pandya CD, Vekaria HJ, Zamorano M, Trout AL, Ritzel RM, Guzman GU, Bolden C, Sullivan PG, Gensel JC, Miller BA. Azithromycin reduces hemoglobin-induced innate neuroimmune activation. Exp Neurol 2024; 372:114574. [PMID: 37852468 PMCID: PMC11706628 DOI: 10.1016/j.expneurol.2023.114574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/11/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Neonatal intraventricular hemorrhage (IVH) releases blood products into the lateral ventricles and brain parenchyma. There are currently no medical treatments for IVH and surgery is used to treat a delayed effect of IVH, post-hemorrhagic hydrocephalus. However, surgery is not a cure for intrinsic brain injury from IVH, and is performed in a subacute time frame. Like many neurological diseases and injuries, innate immune activation is implicated in the pathogenesis of IVH. Innate immune activation is a pharmaceutically targetable mechanism to reduce brain injury and post-hemorrhagic hydrocephalus after IVH. Here, we tested the macrolide antibiotic azithromycin, which has immunomodulatory properties, to reduce innate immune activation in an in vitro model of microglial activation using the blood product hemoglobin (Hgb). We then utilized azithromycin in our in vivo model of IVH, using intraventricular blood injection into the lateral ventricle of post-natal day 5 rat pups. In both models, azithromycin modulated innate immune activation by several outcome measures including mitochondrial bioenergetic analysis, cytokine expression and flow cytometric analysis. This suggests that azithromycin, which is safe for neonates, could hold promise for modulating innate immune activation after IVH.
Collapse
Affiliation(s)
- Chirayu D Pandya
- Center for Advanced Translational Stroke Science (CATSS), Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - Hemendra J Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - Miriam Zamorano
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 77030, United States of America
| | - Amanda L Trout
- Center for Advanced Translational Stroke Science (CATSS), Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - Rodney M Ritzel
- Lexington Veterans' Affairs Healthcare System, Lexington, KY 40502, United States of America
| | - Gary U Guzman
- Lexington Veterans' Affairs Healthcare System, Lexington, KY 40502, United States of America
| | - Christopher Bolden
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 77030, United States of America
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center (SCoBIRC), Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America; Lexington Veterans' Affairs Healthcare System, Lexington, KY 40502, United States of America
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center (SCoBIRC), Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - Brandon A Miller
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 77030, United States of America.
| |
Collapse
|
11
|
Lankhuijzen LM, Ridler T. Opioids, microglia, and temporal lobe epilepsy. Front Neurol 2024; 14:1298489. [PMID: 38249734 PMCID: PMC10796828 DOI: 10.3389/fneur.2023.1298489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
A lack of treatment options for temporal lobe epilepsy (TLE) demands an urgent quest for new therapies to recover neuronal damage and reduce seizures, potentially interrupting the neurotoxic cascades that fuel hyper-excitability. Endogenous opioids, along with their respective receptors, particularly dynorphin and kappa-opioid-receptor, present as attractive candidates for controlling neuronal excitability and therapeutics in epilepsy. We perform a critical review of the literature to evaluate the role of opioids in modulating microglial function and morphology in epilepsy. We find that, in accordance with anticonvulsant effects, acute opioid receptor activation has unique abilities to modulate microglial activation through toll-like 4 receptors, regulating downstream secretion of cytokines. Abnormal activation of microglia is a dominant feature of neuroinflammation, and inflammatory cytokines are found to aggravate TLE, inspiring the challenge to alter microglial activation by opioids to suppress seizures. We further evaluate how opioids can modulate microglial activation in epilepsy to enhance neuroprotection and reduce seizures. With controlled application, opioids may interrupt inflammatory cycles in epilepsy, to protect neuronal function and reduce seizures. Research on opioid-microglia interactions has important implications for epilepsy and healthcare approaches. However, preclinical research on opioid modulation of microglia supports a new therapeutic pathway for TLE.
Collapse
Affiliation(s)
| | - Thomas Ridler
- Hatherly Laboratories, Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
12
|
Reddaway J, Richardson PE, Bevan RJ, Stoneman J, Palombo M. Microglial morphometric analysis: so many options, so little consistency. Front Neuroinform 2023; 17:1211188. [PMID: 37637472 PMCID: PMC10448193 DOI: 10.3389/fninf.2023.1211188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/05/2023] [Indexed: 08/29/2023] Open
Abstract
Quantification of microglial activation through morphometric analysis has long been a staple of the neuroimmunologist's toolkit. Microglial morphological phenomics can be conducted through either manual classification or constructing a digital skeleton and extracting morphometric data from it. Multiple open-access and paid software packages are available to generate these skeletons via semi-automated and/or fully automated methods with varying degrees of accuracy. Despite advancements in methods to generate morphometrics (quantitative measures of cellular morphology), there has been limited development of tools to analyze the datasets they generate, in particular those containing parameters from tens of thousands of cells analyzed by fully automated pipelines. In this review, we compare and critique the approaches using cluster analysis and machine learning driven predictive algorithms that have been developed to tackle these large datasets, and propose improvements for these methods. In particular, we highlight the need for a commitment to open science from groups developing these classifiers. Furthermore, we call attention to a need for communication between those with a strong software engineering/computer science background and neuroimmunologists to produce effective analytical tools with simplified operability if we are to see their wide-spread adoption by the glia biology community.
Collapse
Affiliation(s)
- Jack Reddaway
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, Neuroscience and Mental Health Innovation Institute (NMHII), Cardiff University, Cardiff, United Kingdom
| | | | - Ryan J. Bevan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Jessica Stoneman
- Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Marco Palombo
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
- School of Computer Science and Informatics, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
13
|
Toutonji A, Krieg C, Borucki DM, Mandava M, Guglietta S, Tomlinson S. Mass cytometric analysis of the immune cell landscape after traumatic brain injury elucidates the role of complement and complement receptors in neurologic outcomes. Acta Neuropathol Commun 2023; 11:92. [PMID: 37308987 DOI: 10.1186/s40478-023-01583-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/18/2023] [Indexed: 06/14/2023] Open
Abstract
Following traumatic brain injury (TBI), a neuroinflammatory response can persist for years and contribute to the development of chronic neurological manifestations. Complement plays a central role in post-TBI neuroinflammation, and C3 opsonins and the anaphylatoxins (C3a and C5a) have been implicated in promoting secondary injury. We used single cell mass cytometry to characterize the immune cell landscape of the brain at different time points after TBI. To specifically investigate how complement shapes the post-TBI immune cell landscape, we analyzed TBI brains in the context of CR2-Crry treatment, an inhibitor of C3 activation. We analyzed 13 immune cell types, including peripheral and brain resident cells, and assessed expression of various receptors. TBI modulated the expression of phagocytic and complement receptors on both brain resident and infiltrating peripheral immune cells, and distinct functional clusters were identified within same cell populations that emerge at different phases after TBI. In particular, a CD11c+ (CR4) microglia subpopulation continued to expand over 28 days after injury, and was the only receptor to show continuous increase over time. Complement inhibition affected the abundance of brain resident immune cells in the injured hemisphere and impacted the expression of functional receptors on infiltrating cells. A role for C5a has also been indicated in models of brain injury, and we found significant upregulation of C5aR1 on many immune cell types after TBI. However, we demonstrated experimentally that while C5aR1 is involved in the infiltration of peripheral immune cells into the brain after injury, it does not alone affect histological or behavioral outcomes. However, CR2-Crry improved post-TBI outcomes and reduced resident immune cell populations, as well as complement and phagocytic receptor expression, indicating that its neuroprotective effects are mediated upstream of C5a generation, likely via modulating C3 opsonization and complement receptor expression.
Collapse
Affiliation(s)
- Amer Toutonji
- College of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Carsten Krieg
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
- Hollings Cancer Center, Charleston, SC, 29425, USA
| | - Davis M Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Mamatha Mandava
- Immune Deficiency Cellular Therapy Program (IDCTP), National Institutes of Health, Bethesda, USA
| | - Silvia Guglietta
- Hollings Cancer Center, Charleston, SC, 29425, USA.
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Ralph Johnson VA Medical Center, Charleston, SC, 29401, USA.
| |
Collapse
|
14
|
Xiong J, Lv Y, Ma X, Peng G, Wu C, Hou J, Zhang Y, Wu C, Chen-Yi Liu T, Yang L. Neuroprotective Effect of Sub-lethal Hyperthermia Preconditioning in a Rat Model of Repeated Closed Head Injury. Neuroscience 2023; 522:57-68. [PMID: 37164305 DOI: 10.1016/j.neuroscience.2023.04.030] [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: 12/17/2022] [Revised: 03/28/2023] [Accepted: 04/29/2023] [Indexed: 05/12/2023]
Abstract
Repeated mild traumatic brain injury (rTBI), one of the most common forms of traumatic brain injury, is a worldwide severe public health concern. rTBI induces cumulative neuronal injury, neurological dysfunction, and cognitive deficits. Although there are clinical treatment methods, there is still an urgent need to develop preventive approaches for susceptible populations. Using a repeated closed head injury (rCHI) rat model, we interrogate the effect of sub-lethal hyperthermia preconditioning (SHP) on rCHI-induced neuronal injury and behavioral changes. Our study applied the repeated weight-drop model to induce the rCHI. According to the changes of heat shock protein 70 (HSP 70) in the cortex and hippocampus following a single SHP treatment in normal rats, the SHP was delivered to the rats 18 hours before rCHI. We found that HSP significantly alleviated rCHI-induced anxiety-like behaviors and impairments in motor abilities and spatial memory. SHP exerts significant neuroprotection against rCHI-induced neuronal damage, apoptosis, and neuroinflammation. Our findings support the potential use of SHP as a preventative approach for alleviating rCHI-induced brain damage.
Collapse
Affiliation(s)
- Jing Xiong
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China; Guangzhou Cadre Health Management Center, Guangzhou, China 510006, China
| | - Ying Lv
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Xu Ma
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Guangcong Peng
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Chunyi Wu
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Jun Hou
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Yulan Zhang
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Chongyun Wu
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China.
| | - Timon Chen-Yi Liu
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China.
| | - Luodan Yang
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China.
| |
Collapse
|
15
|
Hani Hojjati S, Butler TA, Chiang GC, Habeck C, RoyChoudhury A, Feiz F, Shteingart J, Nayak S, Ozoria S, Fernández A, Stern Y, Luchsinger JA, Devanand DP, Razlighi QR. Distinct and joint effects of low and high levels of Aβ and tau deposition on cortical thickness. Neuroimage Clin 2023; 38:103409. [PMID: 37104927 PMCID: PMC10165160 DOI: 10.1016/j.nicl.2023.103409] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
Alzheimer's disease (AD) is defined by the presence of Amyloid-β (Aβ),tau, and neurodegeneration (ATN framework) in the human cerebral cortex. Yet, prior studies have suggested that Aβ deposition can be associated with both cortical thinning and thickening. These contradictory results are attributed to small sample sizes, the presence versus absence of tau, and limited detectability in the earliest phase of protein deposition, which may begin in young adulthood and cannot be captured in studies enrolling only older subjects. In this study, we aimed to find the distinct and joint effects of Aβ andtau on neurodegeneration during the progression from normal to abnormal stages of pathologies that remain elusive. We used18F-MK6240 and 18F-Florbetaben/18F-Florbetapir positron emission tomography (PET) and magnetic resonance imaging (MRI) to quantify tau, Aβ, and cortical thickness in 590 participants ranging in age from 20 to 90. We performed multiple regression analyses to assess the distinct and joint effects of Aβ and tau on cortical thickness using 590 healthy control (HC) and mild cognitive impairment (MCI) participants (141 young, 394 HC elderlies, 52 MCI). We showed thatin participants with normal levels of global Aβdeposition, Aβ uptakewassignificantly associated with increasedcortical thickness regardless of tau (e.g., left entorhinal cortex with t > 3.241, p < 0.0013). The relationship between tau deposition and neurodegeneration was more complex: in participants with abnormal levels of global tau, tau uptake was associated with cortical thinning in several regions of the brain (e.g., left entorhinal with t < -2.80, p < 0.0096 and left insula with t-value < -4.284, p < 0.0001), as reported on prior neuroimaging and neuropathological studies. Surprisingly, in participants with normal levels of global tau, tau was found to be associated with cortical thickening. Moreover, in participants with abnormal levels of global Aβandtau, theresonancebetween them, defined as their correlation throughout the cortex, wasassociated strongly with cortical thinning even when controlling for a direct linear effect. We confirm prior findings of an association between Aβ deposition and cortical thickening and suggest this may also be the case in the earliest stages of deposition in normal aging. We also illustrate that resonance between high levels of Aβ and tau uptake is strongly associated with cortical thinning, emphasizing the effects of Aβ/tau synergy inAD pathogenesis.
Collapse
Affiliation(s)
- Seyed Hani Hojjati
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States.
| | - Tracy A Butler
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Gloria C Chiang
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Christian Habeck
- Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, United States
| | - Arindam RoyChoudhury
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Farnia Feiz
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Jacob Shteingart
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Siddharth Nayak
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Sindy Ozoria
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Antonio Fernández
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| | - Yaakov Stern
- Departments of Neurology, Psychiatry, GH Sergievsky Center, the Taub Institute for the Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, United States
| | - José A Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Davangere P Devanand
- Division of Geriatric Psychiatry, New York State Psychiatric Institute, Columbia University Irving Medical Center, New York, NY, United States; Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, United States; Department of Psychiatry, New York State Psychiatric Institute, Columbia University Irving Medical Center, New York, NY, United States
| | - Qolamreza R Razlighi
- Quantitative Neuroimaging Laboratory, Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
16
|
Doust YV, Bindoff A, Holloway OG, Wilson R, King AE, Ziebell JM. Temporal changes in the microglial proteome of male and female mice after a diffuse brain injury using label-free quantitative proteomics. Glia 2023; 71:880-903. [PMID: 36468604 PMCID: PMC10952308 DOI: 10.1002/glia.24313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) triggers neuroinflammatory cascades mediated by microglia, which promotes tissue repair in the short-term. These cascades may exacerbate TBI-induced tissue damage and symptoms in the months to years post-injury. However, the progression of the microglial function across time post-injury and whether this differs between biological sexes is not well understood. In this study, we examined the microglial proteome at 3-, 7-, or 28-days after a midline fluid percussion injury (mFPI) in male and female mice using label-free quantitative proteomics. Data are available via ProteomeXchange with identifier PXD033628. We identified a reduction in microglial proteins involved with clearance of neuronal debris via phagocytosis at 3- and 7-days post-injury. At 28 days post-injury, pro-inflammatory proteins were decreased and anti-inflammatory proteins were increased in microglia. These results indicate a reduction in microglial clearance of neuronal debris in the days post-injury with a shift to anti-inflammatory function by 28 days following TBI. The changes in the microglial proteome that occurred across time post-injury did not differ between biological sexes. However, we did identify an increase in microglial proteins related to pro-inflammation and phagocytosis as well as insulin and estrogen signaling in males compared with female mice that occurred with or without a brain injury. Although the microglial response was similar between males and females up to 28 days following TBI, biological sex differences in the microglial proteome, regardless of TBI, has implications for the efficacy of treatment strategies targeting the microglial response post-injury.
Collapse
Affiliation(s)
- Yasmine V. Doust
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Olivia G. Holloway
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Richard Wilson
- Central Science Laboratory (CSL)University of TasmaniaHobartTasmaniaAustralia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Jenna M. Ziebell
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
17
|
Demuth L, Ohm M, Michaelsen-Preusse K, Schulze K, Riese P, Guzmán CA, Korte M, Hosseini S. Influenza vaccine is able to prevent neuroinflammation triggered by H7N7 IAV infection. Front Pharmacol 2023; 14:1142639. [PMID: 37063291 PMCID: PMC10090407 DOI: 10.3389/fphar.2023.1142639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Influenza A virus (IAV) subtypes are a major cause of illness and mortality worldwide and pose a threat to human health. Although IAV infection is considered a self-limiting respiratory syndrome, an expanded spectrum of cerebral manifestations has been reported following IAV infection. Neurotropic IAVs, such as the H7N7 subtype, are capable of invading the central nervous system (CNS) and replicating in brain cells, resulting in microglia-induced neuroinflammation. Microglial cells, the brain’s resident immune cells, are instrumental in the inflammatory response to viral infection. While activation of microglia is important to initially contain the virus, excessive activation of these cells leads to neuronal damage. Previous studies have shown that acute and even long-term IAV-induced neuroinflammation leads to CNS damage. Therefore, the search for possible preventive or therapeutic strategies is of great importance. In this study, we investigated the potential effect of vaccination against acute neuroinflammation induced by H7N7 infection and subsequent neuronal damage in the hippocampus, a particularly vulnerable brain region, comparing young and aged mice. Immunosenescence is one of the striking pathophysiological changes during mammalian aging that leads to “inflammaging” and critically limits the protection by vaccines in the elderly. The results suggest that formalin-inactivated H7N7 vaccine has a preventive effect against the inflammatory responses in the periphery and also in the CNS after H7N7 infection. Cytokine and chemokine levels, increased microglial density, and cell volume after H7N7 infection were all attenuated by vaccination. Further structural analysis of microglial cells also revealed a change in branching complexity after H7N7 infection, most likely reflecting the neuroprotective effect of the vaccination. In addition, synapse loss was prevented in vaccinated mice. Remarkably, engulfment of post-synaptic compartments by microglia can be proposed as the underlying mechanism for spine loss triggered by H7N7 infection, which was partially modulated by vaccination. Although young mice showed better protection against neuroinflammation and the resulting deleterious neuronal effects upon vaccination, a beneficial role of the vaccine was also observed in the brains of older mice. Therefore, vaccination can be proposed as an important strategy to prevent neurological sequelae of H7N7 infection.
Collapse
Affiliation(s)
- Luisa Demuth
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Melanie Ohm
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kristin Michaelsen-Preusse
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kai Schulze
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Peggy Riese
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Carlos A. Guzmán
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany
- *Correspondence: Shirin Hosseini,
| |
Collapse
|
18
|
Sleep Deprivation Induces Dopamine System Maladaptation and Escalated Corticotrophin-Releasing Factor Signaling in Adolescent Mice. Mol Neurobiol 2023; 60:3190-3209. [PMID: 36813955 DOI: 10.1007/s12035-023-03258-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023]
Abstract
Sleep disruption is highly associated with the pathogenesis and progression of a wild range of psychiatric disorders. Furthermore, appreciable evidence shows that experimental sleep deprivation (SD) on humans and rodents evokes anomalies in the dopaminergic (DA) signaling, which are also implicated in the development of psychiatric illnesses such as schizophrenia or substance abuse. Since adolescence is a vital period for the maturation of the DA system as well as the occurrence of mental disorders, the present studies aimed to investigate the impacts of SD on the DA system of adolescent mice. We found that 72 h SD elicited a hyperdopaminergic status, with increased sensitivity to the novel environment and amphetamine (Amph) challenge. Also, altered neuronal activity and expression of striatal DA receptors were noticed in the SD mice. Moreover, 72 h SD influenced the immune status in the striatum, with reduced microglial phagocytic capacity, primed microglial activation, and neuroinflammation. The abnormal neuronal and microglial activity were putatively provoked by the enhanced corticotrophin-releasing factor (CRF) signaling and sensitivity during the SD period. Together, our findings demonstrated the consequences of SD in adolescents including aberrant neuroendocrine, DA system, and inflammatory status. Sleep insufficiency is a risk factor for the aberration and neuropathology of psychiatric disorders.
Collapse
|
19
|
Abdelsalam SA, Renu K, Zahra HA, Abdallah BM, Ali EM, Veeraraghavan VP, Sivalingam K, Ronsard L, Ammar RB, Vidya DS, Karuppaiya P, Al-Ramadan SY, Rajendran P. Polyphenols Mediate Neuroprotection in Cerebral Ischemic Stroke-An Update. Nutrients 2023; 15:nu15051107. [PMID: 36904106 PMCID: PMC10005012 DOI: 10.3390/nu15051107] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Stroke is one of the main causes of mortality and disability, and it is due to be included in monetary implications on wellbeing frameworks around the world. Ischemic stroke is caused by interference in cerebral blood flow, leading to a deficit in the supply of oxygen to the affected region. It accounts for nearly 80-85% of all cases of stroke. Oxidative stress has a significant impact on the pathophysiologic cascade in brain damage leading to stroke. In the acute phase, oxidative stress mediates severe toxicity, and it initiates and contributes to late-stage apoptosis and inflammation. Oxidative stress conditions occur when the antioxidant defense in the body is unable to counteract the production and aggregation of reactive oxygen species (ROS). The previous literature has shown that phytochemicals and other natural products not only scavenge oxygen free radicals but also improve the expressions of cellular antioxidant enzymes and molecules. Consequently, these products protect against ROS-mediated cellular injury. This review aims to give an overview of the most relevant data reported in the literature on polyphenolic compounds, namely, gallic acid, resveratrol, quercetin, kaempferol, mangiferin, epigallocatechin, and pinocembrin, in terms of their antioxidant effects and potential protective activity against ischemic stroke.
Collapse
Affiliation(s)
- Salaheldin Abdelraouf Abdelsalam
- Department of Biological Sciences, College of Science, King Faisal University, Chennai 31982, Saudi Arabia
- Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College & Hospitals, Saveetha University, Chennai 600077, India
| | - Hamad Abu Zahra
- Department of Biological Sciences, College of Science, King Faisal University, Chennai 31982, Saudi Arabia
| | - Basem M. Abdallah
- Department of Biological Sciences, College of Science, King Faisal University, Chennai 31982, Saudi Arabia
| | - Enas M. Ali
- Department of Biological Sciences, College of Science, King Faisal University, Chennai 31982, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College & Hospitals, Saveetha University, Chennai 600077, India
| | - Kalaiselvi Sivalingam
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Larance Ronsard
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rebai Ben Ammar
- Department of Biological Sciences, College of Science, King Faisal University, Chennai 31982, Saudi Arabia
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cedria, Technopole of Borj-Cedria, P.O. Box 901, Hammam-Lif 2050, Tunisia
| | - Devanathadesikan Seshadri Vidya
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al-Kharj 11942, Saudi Arabia
| | - Palaniyandi Karuppaiya
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
| | - S. Y. Al-Ramadan
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Chennai 31982, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College & Hospitals, Saveetha University, Chennai 600077, India
- Correspondence: ; Tel.: +966-0135899543
| |
Collapse
|
20
|
Wu Q, Wang H, Liu X, Zhao Y, Su P. Microglial activation and over pruning involved in developmental epilepsy. J Neuropathol Exp Neurol 2023; 82:150-159. [PMID: 36453895 DOI: 10.1093/jnen/nlac111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
To understand the potential role of microglia in synaptic pruning following status epilepticus (SE), we examined the time course of expression of Iba-1, and immune and neuroinflammatory regulators, including CD86, CD206, and CX3CR1, and TLR4/NF-κB after SE induced by pilocarpine in rats. Behavioral tests, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, immunohistochemical staining, Western blotting, PCR, and fluorescence double staining assessments were performed. The expression of Iba-1 protein was lowest in the control group, and peaked after 2 days (p < 0.001). CD86 and CD206 mRNA levels increased gradually in the microglia of the epilepsy group after 12 hours, 1 day, 2 days, and 3 days; peak expression was on the second day. The expression of the chemokine receptor CX3CR1 in microglia increased to varying degrees after SE, and expression of the presynaptic protein synapsin decreased. The expression of TLR4/NF-κB in microglia positively correlated with Iba-1 protein expression. These findings indicate that the TLR4/NF-κB signaling pathway may be involved in the activation and polarization of microglia in epilepsy and in excess synaptic pruning, which could lead to an increase in brain injury.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueyan Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yajuan Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Su
- Experimental Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Ng PY, Zhang C, Li H, Baker DJ. Senescent Microglia Represent a Subset of Disease-Associated Microglia in P301S Mice. J Alzheimers Dis 2023; 95:493-507. [PMID: 37545233 PMCID: PMC10848894 DOI: 10.3233/jad-230109] [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] [Indexed: 08/08/2023]
Abstract
BACKGROUND The existence and contribution of microglia with senescent-like alterations in the pathogenesis of age-related neurodegenerative diseases like Alzheimer's disease (AD) have been suggested in recent years. However, the identification of this distinct microglial population in vivo has proven challenging, largely due to overlaps in the inflammatory phenotype of activated and senescent microglia. Furthermore, attempts at recapitulating senescence in microglia in vitro are limited. OBJECTIVE To identify and characterize senescent microglia that occur in vivo in an animal model of neurodegeneration driven by pathologic tau. METHODS We analyzed the RNA expression patterns of individual microglia from normal mice and the pathogenic tau P301 S PS19 mouse model. We have previously demonstrated that p16-expressing senescent microglia occur in these mice when neurodegeneration has occurred. RESULTS Here we identify a subset of disease-associated microglia with senescent features, notably characterized by the expression of Ccl4. This signature overlaps with established markers of senescence from other cell types. CONCLUSION Our characterization of senescent microglia can be used to better understand the role of senescent microglia in various age-related contexts, including whether clearance of senescent microglia represents a viable therapeutic option.
Collapse
Affiliation(s)
- Pei Y. Ng
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Darren J. Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- The Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Imado E, Sun S, Abawa AR, Tahara T, Kochi T, Huynh TNB, Asano S, Hasebe S, Nakamura Y, Hisaoka-Nakashima K, Kotake Y, Irifune M, Tsuga K, Takuma K, Morioka N, Kiguchi N, Ago Y. Prenatal exposure to valproic acid causes allodynia associated with spinal microglial activation. Neurochem Int 2022; 160:105415. [PMID: 36027995 DOI: 10.1016/j.neuint.2022.105415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and social interaction and the presence of restricted, repetitive behaviors. Additionally, difficulties in sensory processing commonly occur in ASD. Sensory abnormalities include heightened or reduced sensitivity to pain, but the mechanism underlying sensory phenotypes in ASD remain unknown. Emerging evidence suggests that microglia play an important role in forming and refining neuronal circuitry, and thus contribute to neuronal plasticity and nociceptive signaling. In the present study, we investigated the age-dependent tactile sensitivity in an animal model of ASD induced by prenatal exposure to valproic acid (VPA) and subsequently assessed the involvement of microglia in the spinal cord in pain processing. Pregnant ICR (CD1) mice were intraperitoneally injected with either saline or VPA (500 mg/kg) on embryonic day 12.5. Male offspring of VPA-treated mothers showed mechanical allodynia at both 4 and 8 weeks of age. In the spinal cord dorsal horn in prenatally VPA-treated mice, the numbers and staining intensities of ionized calcium-binding adapter molecule 1-positive cells were increased and the cell bodies became enlarged, indicating microglial activation. Treatment with PLX3397, a colony-stimulating factor 1 receptor inhibitor, for 10 days resulted in a decreased number of spinal microglia and attenuated mechanical allodynia in adult mice prenatally exposed to VPA. Additionally, intrathecal injection of Mac-1-saporin, a saporin-conjugated anti-CD11b antibody to deplete microglia, abolished mechanical allodynia. These findings suggest that prenatal VPA treatment causes allodynia and that spinal microglia contribute to the increased nociceptive responses.
Collapse
Affiliation(s)
- Eiji Imado
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Samnang Sun
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Faculty of Odonto-Stomatology, University of Health Sciences, #73, Monivong Blvd., Sangkat Sras Chak, Khan Daun Penh, Phnom Penh, 12201, Cambodia
| | - Abrar Rizal Abawa
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Faculty of Dental Medicine, Universitas Airlangga, Jl. Mayjen Prof. Dr. Moestopo No. 47, Surabaya, East Java, 60132, Indonesia
| | - Takeru Tahara
- Department of Neurochemistry and Environmental Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Takahiro Kochi
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Tran Ngoc Bao Huynh
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Faculty of Odonto-Stomatology, Hong Bang International University, 215 Dien Bien Phu Street, Ward 15, Binh Thanh District, Ho Chi Minh City, Viet Nam
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Shigeru Hasebe
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Yaichiro Kotake
- Department of Neurochemistry and Environmental Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Masahiro Irifune
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazuhiro Tsuga
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Department of Advanced Prosthodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, 565-0871, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Wakayama, 640-8156, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan; Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
23
|
Deng P, Wang L, Zhang Q, Chen S, Zhang Y, Xu H, Chen H, Xu Y, He W, Zhang J, Sun H. Therapeutic Potential of a Combination of Electroacupuncture and Human iPSC-Derived Small Extracellular Vesicles for Ischemic Stroke. Cells 2022; 11:820. [PMID: 35269441 PMCID: PMC8909871 DOI: 10.3390/cells11050820] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
This paper aimed to explore the roles of the combination of electroacupuncture (EA) and induced pluripotent stem cell-derived small extracellular vesicles (iPSC-EVs) on mice with ischemic stroke and the underlying mechanisms. A focal cerebral ischemia model was established in C57BL/6 mice through middle cerebral artery occlusion (MCAO). After 3 days, neurological impairment and motor function were examined by performing behavioral tests. The infarct volume and neuronal apoptosis were examined using TTC staining and TUNEL assays. Flow cytometry was performed to assess the proliferation of T lymphocytes. The changes in the interleukin (IL)-33/ST2 axis were evaluated by immunofluorescence and Western blotting. The combination of EA and iPSC-EVs treatment ameliorated neurological impairments and reduced the infarct volume and neuronal apoptosis in MCAO mice. EA plus iPSC-EVs suppressed T helper (Th1) and Th17 responses and promoted the regulatory T cell (Treg) response. In addition, EA plus iPSC-EVs exerted neuroprotective effects by regulating the IL-33/ST2 axis and inhibiting the microglia and astrocyte activation. Taken together, the study shows that EA and iPSC-EVs exerted a synergistic neuroprotective effect in MCAO mice, and this treatment may represent a novel potent therapy for ischemic stroke and damage to other tissues.
Collapse
Affiliation(s)
- Peiying Deng
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Liang Wang
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Qiongqiong Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Suhui Chen
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Yamin Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Hong Xu
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Hui Chen
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Yi Xu
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Wei He
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Jianmin Zhang
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
- Guidon Pharmaceutics, Beijing 100176, China
| | - Hua Sun
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| |
Collapse
|
24
|
Hutchinson E, Osting S, Rutecki P, Sutula T. Diffusion Tensor Orientation as a Microstructural MRI Marker of Mossy Fiber Sprouting After TBI in Rats. J Neuropathol Exp Neurol 2021; 81:27-47. [PMID: 34865073 DOI: 10.1093/jnen/nlab123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diffusion tensor imaging (DTI) metrics are highly sensitive to microstructural brain alterations and are potentially useful imaging biomarkers for underlying neuropathologic changes after experimental and human traumatic brain injury (TBI). As potential imaging biomarkers require direct correlation with neuropathologic alterations for validation and interpretation, this study systematically examined neuropathologic abnormalities underlying alterations in DTI metrics in the hippocampus and cortex following controlled cortical impact (CCI) in rats. Ex vivo DTI metrics were directly compared with a comprehensive histologic battery for neurodegeneration, microgliosis, astrocytosis, and mossy fiber sprouting by Timm histochemistry at carefully matched locations immediately, 48 hours, and 4 weeks after injury. DTI abnormalities corresponded to spatially overlapping but temporally distinct neuropathologic alterations representing an aggregate measure of dynamic tissue damage and reorganization. Prominent DTI alterations of were observed for both the immediate and acute intervals after injury and associated with neurodegeneration and inflammation. In the chronic period, diffusion tensor orientation in the hilus of the dentate gyrus became prominently abnormal and was identified as a reliable structural biomarker for mossy fiber sprouting after CCI in rats, suggesting potential application as a biomarker to follow secondary progression in experimental and human TBI.
Collapse
Affiliation(s)
- Elizabeth Hutchinson
- From the Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA (EH); and Department of Neurology, University of Wisconsin, Madison, Wisconsin, USA (SO, PR, TS)
| | - Susan Osting
- From the Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA (EH); and Department of Neurology, University of Wisconsin, Madison, Wisconsin, USA (SO, PR, TS)
| | - Paul Rutecki
- From the Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA (EH); and Department of Neurology, University of Wisconsin, Madison, Wisconsin, USA (SO, PR, TS)
| | - Thomas Sutula
- From the Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA (EH); and Department of Neurology, University of Wisconsin, Madison, Wisconsin, USA (SO, PR, TS)
| |
Collapse
|
25
|
Augusto-Oliveira M, Arrifano GP, Delage CI, Tremblay MÈ, Crespo-Lopez ME, Verkhratsky A. Plasticity of microglia. Biol Rev Camb Philos Soc 2021; 97:217-250. [PMID: 34549510 DOI: 10.1111/brv.12797] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023]
Abstract
Microglial cells are the scions of foetal macrophages which invade the neural tube early during embryogenesis. The nervous tissue environment instigates the phenotypic metamorphosis of foetal macrophages into idiosyncratic surveilling microglia, which are generally characterised by a small cell body and highly ramified motile processes that constantly scan the nervous tissue for signs of changes in homeostasis and allow microglia to perform crucial homeostatic functions. The surveilling microglial phenotype is evolutionarily conserved from early invertebrates to humans. Despite this evolutionary conservation, microglia show substantial heterogeneity in their gene and protein expression, as well as morphological appearance. These differences are age, region and context specific and reflect a high degree of plasticity underlying the life-long adaptation of microglia, supporting the exceptional adaptive capacity of the central nervous system. Microgliocytes are essential elements of cellular network formation and refinement in the developing nervous tissue. Several distinct patrolling modes of microglial processes contribute to the formation, modification, and pruning of synapses; to the support and protection of neurones through microglial-somatic junctions; and to the control of neuronal and axonal excitability by specific microglia-axonal contacts. In pathology, microglia undergo proliferation and reactive remodelling known as microgliosis, which is context dependent, yet represents an evolutionarily conserved defence response. Microgliosis results in the emergence of multiple disease and context-specific reactive states; in addition, neuropathology is associated with the appearance of specific protective or recovery microglial forms. In summary, the plasticity of microglia supports the development and functional activity of healthy nervous tissue and provides highly sophisticated defences against disease.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Charlotte Isabelle Delage
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, V8P 5C2, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec City, QC, G1V 4G2, Canada.,Neurology and Neurosurgery Department, McGill University, 3801 University Street, Montreal, QC, H3A 2B4, Canada.,Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Bureau 4835, 1050 Avenue de la Médecine, Québec City, QC, G1V 0A6, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, U.K.,Achucarro Center for Neuroscience, IKERBASQUE, 48011, Bilbao, Spain.,Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| |
Collapse
|
26
|
Giordano KR, Denman CR, Dubisch PS, Akhter M, Lifshitz J. An update on the rod microglia variant in experimental and clinical brain injury and disease. Brain Commun 2021; 3:fcaa227. [PMID: 33501429 PMCID: PMC7811762 DOI: 10.1093/braincomms/fcaa227] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Contemporary microglia morphologies include ramified, activated and amoeboid, with the morphology of microglia considered highly coupled to the cellular function. Rod microglia are an additional activated microglia variant observed in the ageing, injured and diseased brain. Rod microglia were reported frequently in the early 1900s by neuropathologists in post-mortem cases of general paresis, Alzheimer's disease and encephalitis, and then remained largely ignored for almost 100 years. Recent reports have renewed interest in rod microglia, most notably after experimental traumatic brain injury. Rod microglia are formed by the narrowing of the soma and retraction of planar processes, which results in the appearance of an elongated, rod-shaped cell. Rod microglia are most commonly observed in the cortex, aligned perpendicular to the dural surface and adjacent to neuronal processes; in the hippocampus, they are aligned perpendicular to hippocampal layers. Furthermore, rod microglia form trains with one another, apical end to basal end. By replicating the process of sketching microscopic observation, rod microglia are re-defined by circumnutation around the long axis. In this update, we summarize the rod microglia variant in clinical and experimental literature and advocate for investigation into mechanisms of rod microglia origin and function.
Collapse
Affiliation(s)
- Katherine R Giordano
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Charlotte R Denman
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Department of Biology and Biochemistry, University of Bath, Bath, UK
| | | | - Murtaza Akhter
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Department of Emergency Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Maricopa Integrated Healthcare System, Phoenix, AZ, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Phoenix VA Health Care System, Phoenix, AZ, USA
| |
Collapse
|
27
|
Hassan QN, Queen NJ, Cao L. Regulation of aging and cancer by enhanced environmental activation of a hypothalamic-sympathoneural-adipocyte axis. Transl Cancer Res 2020; 9:5687-5699. [PMID: 33134111 PMCID: PMC7595574 DOI: 10.21037/tcr.2020.02.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Social and environmental factors impact cancer and energy balance profoundly. Years ago, our lab established the existence of a novel brain-fat interaction we termed the "hypothalamic-sympathoneural-adipocyte (HSA) axis", through which complex environmental stimuli provided by an enriched environment regulate body composition, energy balance, and development of cancer. We have spent a significant portion of the past decade to further characterize the broad health benefits of an enriched environment (for example, leanness, enhanced immune function, and cancer resistance), and to identify mediators in the brain and periphery along the HSA axis. This review summarizes our recent work regarding the interface between endocrinology, immunology, cancer biology, aging, and neuroscience. We will discuss the interplay between these systemic phenomena and how the HSA axis can be targeted for regulation of cancer and aging.
Collapse
Affiliation(s)
- Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
28
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
29
|
Queen NJ, Hassan QN, Cao L. Improvements to Healthspan Through Environmental Enrichment and Lifestyle Interventions: Where Are We Now? Front Neurosci 2020; 14:605. [PMID: 32655354 PMCID: PMC7325954 DOI: 10.3389/fnins.2020.00605] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Environmental enrichment (EE) is an experimental paradigm that is used to explore how a complex, stimulating environment can impact overall health. In laboratory animal experiments, EE housing conditions typically include larger-than-standard cages, abundant bedding, running wheels, mazes, toys, and shelters which are rearranged regularly to further increase stimulation. EE has been shown to improve multiple aspects of health, including but not limited to metabolism, learning and cognition, anxiety and depression, and immunocompetence. Recent advances in lifespan have led some researchers to consider aging as a risk factor for disease. As such, there is a pressing need to understand the processes by which healthspan can be increased. The natural and predictable changes during aging can be reversed or decreased through EE and its underlying mechanisms. Here, we review the use of EE in laboratory animals to understand mechanisms involved in aging, and comment on relative areas of strength and weakness in the current literature. We additionally address current efforts toward applying EE-like lifestyle interventions to human health to extend healthspan. Although increasing lifespan is a clear goal of medical research, improving the quality of this added time also deserves significant attention. Despite hurdles in translating experimental results toward clinical application, we argue there is great potential in using features of EE toward improving human healthy life expectancy or healthspan, especially in the context of increased global longevity.
Collapse
Affiliation(s)
- Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
30
|
Gegunde S, Alfonso A, Alonso E, Alvariño R, Botana LM. Gracilin-Derivatives as Lead Compounds for Anti-inflammatory Effects. Cell Mol Neurobiol 2020; 40:603-615. [PMID: 31729596 DOI: 10.1007/s10571-019-00758-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/05/2019] [Indexed: 01/23/2023]
Abstract
Gracilins are diterpenes derivative, isolated from the marine sponge Spongionella gracilis. Natural gracilins and synthetic derivatives have shown antioxidant, immunosuppressive, and neuroprotective capacities related to the affinity for cyclophilins. The aim of this work was to study anti-inflammatory and immunosuppressive pathways modulated by gracilin L and two synthetic analogues, compound 1 and 2, on a cellular model of inflammation. In this way, the murine BV2 microglia cell line was used. To carry out the experiments, microglia cells were pre-treated with compounds for 1 h and then stimulated with lipopolysaccharide for 24 h to determine reactive oxygen species production, mitochondrial membrane potential, the release of nitric oxide, interleukin-6 and tumor necrosis factor-α and the expression of Nuclear factor-erythroid 2-related factor 2, Nuclear Factor-κB, the inducible nitric oxide synthase, and the cyclophilin A. Finally, a co-culture of neuron SH-SY5Y and microglia BV2 cells was used to check the neuroprotective effect of these compounds. Cyclosporine A was used as a control of effect. The compounds were able to decrease inflammatory mediators, the expression of inflammatory target proteins as well as they activated anti-oxidative mechanism upon inflammatory conditions. For this reason, natural and synthetic gracilins could be interesting for developing anti-inflammatory drugs.
Collapse
Grants
- 2017 GRC GI-1682 Consellería de Cultura, Educación e Ordenación Universitaria, Xunta de Galicia
- AGL2016-78728-R Ministerio de Economía, Industria y Competitividad, Gobierno de España
- ISCIII/PI16/01830 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- RTC-2016-5507-2 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- ITC-20161072 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- 0161-Nanoeaters -1-E-1 European Commission
- Interreg AlertoxNet EAPA-317-2016 European Commission
- Interreg Agritox EAPA-998-2018 European Commission
- 778069-EMERTOX Horizon 2020
Collapse
Affiliation(s)
- Sandra Gegunde
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Amparo Alfonso
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain.
| | - Eva Alonso
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
- Fundación Instituto de Investigación Sanitaria Santiago de Compostela (FIDIS), Hospital Universitaio Lucus Augusti, 27004, Lugo, Spain
| | - Rebeca Alvariño
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| | - Luis M Botana
- Pharmacology Department, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002, Lugo, Spain
| |
Collapse
|
31
|
Diaz-Chávez A, Lajud N, Roque A, Cheng JP, Meléndez-Herrera E, Valdéz-Alarcón JJ, Bondi CO, Kline AE. Early life stress increases vulnerability to the sequelae of pediatric mild traumatic brain injury. Exp Neurol 2020; 329:113318. [PMID: 32305419 DOI: 10.1016/j.expneurol.2020.113318] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Early life stress (ELS) is a risk factor for many psychopathologies that happen later in life. Although stress can occur in cases of child abuse, studies on non-accidental brain injuries in pediatric populations do not consider the possible increase in vulnerability caused by ELS. Hence, we sought to determine whether ELS increases the effects of pediatric mild traumatic brain injury (mTBI) on cognition, hippocampal inflammation, and plasticity. Male rats were subjected to maternal separation for 180 min per day (MS180) or used as controls (CONT) during the first 21 post-natal (P) days. At P21 the rats were anesthetized with isoflurane and subjected to a mild controlled cortical impact or sham injury. At P32 the rats were injected with the cell proliferation marker bromodeoxyuridine (BrdU, 500 mg/kg), then evaluated for spatial learning and memory in a water maze (P35-40) and sacrificed for quantification of Ki67+, BrdU+ and Iba1+ (P42). Neither MS180 nor mTBI impacted cognitive outcome when provided alone but their combination (MS180 + mTBI) decreased spatial learning and memory relative to Sham controls (p < .01). mTBI increased microglial activation and affected BrdU+ cell survival in the ipsilateral hippocampus without affecting proliferation rates. However, only MS180 + mTBI increased microglial activation in the area adjacent to the injury and the contralateral CA1 hippocampal subfield, and decreased cell proliferation in the ipsilateral neurogenic niche. Overall, the data show that ELS increases the vulnerability to the sequelae of pediatric mTBI and may be mediated by increased neuroinflammation.
Collapse
Affiliation(s)
- Arturo Diaz-Chávez
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Angélica Roque
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Esperanza Meléndez-Herrera
- Instituto de Investigaciones sobre los Recursos Naturales - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Juan José Valdéz-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología, Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; Psychology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
32
|
Gilmour A, Poole-Warren L, Green RA. An Improved in vitro Model of Cortical Tissue. Front Neurosci 2019; 13:1349. [PMID: 31920510 PMCID: PMC6928009 DOI: 10.3389/fnins.2019.01349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/02/2019] [Indexed: 11/20/2022] Open
Abstract
Intracortical electrodes for brain-machine interfaces rely on intimate contact with tissues for recording signals and stimulating neurons. However, the long-term viability of intracortical electrodes in vivo is poor, with a major contributing factor being the development of a glial scar. In vivo approaches for evaluating responses to intracortical devices are resource intensive and complex, making statistically significant, high throughput data difficult to obtain. In vitro models provide an alternative to in vivo studies; however, existing approaches have limitations which restrict the translation of the cellular reactions to the implant scenario. Notably, there is no current robust model that includes astrocytes, microglia, oligodendrocytes and neurons, the four principle cell types, critical to the health, function and wound responses of the central nervous system (CNS). In previous research a co-culture of primary mouse mature mixed glial cells and immature neural precursor cells were shown to mimic several key properties of the CNS response to implanted electrode materials. However, the method was not robust and took up to 63 days, significantly affecting reproducibility and widespread use for assessing brain-material interactions. In the current research a new co-culture approach has been developed and evaluated using immunocytochemistry and quantitative polymerase chain reaction (qPCR). The resulting method reduced the time in culture significantly and the culture model was shown to have a genetic signature similar to that of healthy adult mouse brain. This new robust CNS culture model has the potential to significantly improve the capacity to translate in vitro data to the in vivo responses.
Collapse
Affiliation(s)
- Aaron Gilmour
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Clem Jones Centre for Neurobiology and Stem Cell Research, Menzies Health Institute Queensland, Griffiths University, Gold Coast, QLD, Australia
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Rylie A Green
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
33
|
Abstract
Microglia are the primary innate immune cells in the CNS. In the healthy brain, they exhibit a unique molecular homeostatic 'signature', consisting of a specific transcriptional profile and surface protein expression pattern, which differs from that of tissue macrophages. In recent years, there have been a number of important advances in our understanding of the molecular signatures of homeostatic microglia and disease-associated microglia that have provided insight into how these cells are regulated in health and disease and how they contribute to the maintenance of the neural environment.
Collapse
|
34
|
McCarter KD, Li C, Li J, Xu G, Sun H. Influence of low-dose alcohol consumption on post-ischemic inflammation: Role of cystathionine γ-lyase. Alcohol 2019; 76:81-89. [PMID: 30597416 DOI: 10.1016/j.alcohol.2018.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023]
Abstract
Low-dose alcohol consumption (LAC) has been shown to suppress post-ischemic inflammation and alleviate cerebral ischemia/reperfusion (I/R) injury. Cystathionine γ-Lyase (CSE) is one of the enzymes that endogenously produce hydrogen sulfide (H2S), which has an anti-inflammatory property at low concentration. We determined the potential role of CSE in the protective effect of LAC. Male C57BL/6J mice were divided into two groups, an ethanol group and a control group, and gavage fed with 0.7 g/kg/day ethanol or volume-matched water once a day for 8 weeks. Transient focal cerebral ischemia was induced by unilateral middle cerebral artery occlusion (MCAO) for 90 min. CSE inhibitors were intraperitoneally given 30 min prior to the ischemia. Cerebral I/R injury, H2S production, adhesion molecules, IL-1 receptor accessory protein (IL-1RAcP), IL-1β, microglial activation, and neutrophil infiltration were evaluated at 24 h of reperfusion. Eight-week ethanol feeding upregulated CSE in the cerebral cortex and reduced cerebral I/R injury. Moreover, ethanol increased post-ischemic H2S production and alleviated the post-ischemic inflammatory response (expression of adhesion molecules, IL-1RAcP, IL-1β, microglial activation, and neutrophil infiltration) in the peri-infarct cerebral cortex. Both inhibitors of CSE, DL-Propargylglycine (PAG) and β-cyano-L-alanine (BCA), abolished the protective effect of ethanol on cerebral I/R injury. In addition, PAG attenuated the inhibitory effect of ethanol on the post-ischemic inflammation. Thus, LAC may protect against cerebral I/R injury by suppressing post-ischemic inflammation via an upregulated CSE.
Collapse
|
35
|
Ali S, Liu X, Queen NJ, Patel RS, Wilkins R, Mo X, Cao L. Long-term environmental enrichment affects microglial morphology in middle age mice. Aging (Albany NY) 2019; 11:2388-2402. [PMID: 31039130 PMCID: PMC6519992 DOI: 10.18632/aging.101923] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/14/2019] [Indexed: 12/17/2022]
Abstract
Aging is associated with increased central nervous system inflammation, in large part due to dysfunctional microglia. Environmental enrichment (EE) provides a model for studying the dynamics of lifestyle factors in the development of age-related neuroinflammation and microglial dysfunction. EE results in improvements in learning and memory, metabolism, and mental health in a variety of animal models. We recently reported that implementing EE in middle age promotes healthy aging. In the present study, we investigated whether EE influences microglial morphology, and whether EE is associated with changes in expression of microglial and neuroinflammatory markers. Inflammatory cytokines and MHC-II were reduced following 12-month EE in 10-month-old mice. Long-term EE for 7.5 months resulted in broad increases in Iba1 expression in hippocampus, hypothalamus, and amygdala detected by immunohistochemistry. Quantification of microglial morphology reveal both hypertrophy and ramification in these three brain regions, without increases in microglial cell density. These data indicate that long-term EE implemented in middle age results in a microglial state distinct from that of normal aging in standard laboratory housing, in specific brain regions, associated with reduced neuroinflammatory markers and improvement of systemic metabolism.
Collapse
Affiliation(s)
- Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xianglan Liu
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Ripal S. Patel
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Ryan K. Wilkins
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
36
|
Rod microglia and their role in neurological diseases. Semin Cell Dev Biol 2019; 94:96-103. [PMID: 30826549 DOI: 10.1016/j.semcdb.2019.02.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 12/21/2022]
Abstract
The striking morphology of microglia is one of their most prominent characteristics, with many studies categorising microglial function based on morphology e.g. ramified, hyper-ramified, activated, or amoeboid. Communications regarding rod microglia in neurological disease are scant, and where reported, these cells are rarely the focus of discussion. These factors make it difficult to determine how widespread these cells are not only through the brain but also across diseases. Studies in experimental diffuse brain injury are the first reports of not only significant numbers of rod microglia, but distinct arrangements of these cells, reminiscent of carriages of a train. This review summarises the available reports of rod microglia in vivo and rod-like microglia in vitro and eludes to possible functions and signalling cascades that may evoke this distinct morphology. More investigations are required to fully elucidate the function that rod microglia play in neurological diseases.
Collapse
|
37
|
Xu G, Li C, Parsiola AL, Li J, McCarter KD, Shi R, Mayhan WG, Sun H. Dose-Dependent Influences of Ethanol on Ischemic Stroke: Role of Inflammation. Front Cell Neurosci 2019; 13:6. [PMID: 30853895 PMCID: PMC6396710 DOI: 10.3389/fncel.2019.00006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/08/2019] [Indexed: 01/17/2023] Open
Abstract
Chronic ethanol consumption dose-dependently affects both incidence and prognosis of ischemic stroke. Our goal was to determine whether the influence of chronic ethanol consumption on ischemic stroke is related to an altered inflammatory profile in the brain. Male C57BL/6J mice were divided into six groups and gavage fed with 0.175, 0.35, 0.7, 1.4, 2.8 g/kg/day ethanol or volume-matched water once a day for 8 weeks. Adhesion molecules, microglial activation, neutrophil infiltration, pro- and anti-inflammatory cytokines/chemokines, blood-brain barrier (BBB) permeability, and matrix metallopeptidases (MMPs) in the cerebral cortex before and following a 90-min unilateral middle cerebral artery occlusion (MCAO)/24-h reperfusion were evaluated. Brain ischemia/reperfusion (I/R) injury was significantly reduced in 0.7 g/kg/day ethanol group (peak blood ethanol concentration: 9 mM) and worsened in 2.8 g/kg/day ethanol group (peak blood ethanol concentration: 37 mM). Baseline E-selectin was downregulated in all ethanol groups, whereas baseline intercellular adhesion molecule-1 (ICAM-1) was only downregulated in 0.35 and 0.7 g/kg/day ethanol groups. Interestingly, baseline vascular cell adhesion molecule-1 (VCAM-1) was upregulated in 0.35, 0.7, and 1.4 g/kg/day ethanol groups. Post-ischemic upregulation of ICAM-1 and E-selectin were suppressed in all ethanol groups. Post-ischemic neutrophil infiltration and microglial activation were significantly less in the low-moderate (0.175–1.4 g/kg/day) ethanol groups but greater in the 2.8 g/kg/day ethanol group compared to the vehicle group. At basal conditions, ethanol increased one pro- and two anti-inflammatory cytokines/chemokines at the 0.7 g/kg/day dose, and 13 pro- and eight anti-inflammatory cytokines/chemokines at the 2.8 g/kg/day dose. After ischemia, 0.7 g/kg/day ethanol suppressed post-ischemic pro-inflammatory cytokines/chemokines and enhanced post-ischemic anti-inflammatory cytokines/chemokines. Moreover, 0.7 g/kg/day ethanol significantly reduced baseline MMP-9 activity and alleviated post-ischemic BBB breakdown. On the other hand, 2.8 g/kg/day ethanol worsened post-ischemic BBB breakdown. Our findings suggest that low-moderate ethanol consumption may prevent ischemic stroke and reduce brain I/R injury by suppressing inflammation, whereas heavy alcohol consumption may induce ischemic stroke and worsen brain I/R injury by aggravating inflammation.
Collapse
Affiliation(s)
- Guodong Xu
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Chun Li
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Anne L Parsiola
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Jiyu Li
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Kimberly D McCarter
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Runhua Shi
- Department of Medicine/Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - William G Mayhan
- Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD, United States
| | - Hong Sun
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
38
|
Abstract
Neuroinflammation is initiated as a result of traumatic brain injury and can exacerbate evolving tissue pathology. Immune cells respond to acute signals from damaged cells, initiate neuroinflammation, and drive the pathological consequences over time. Importantly, the mechanism(s) of injury, the location of the immune cells within the brain, and the animal species all contribute to immune cell behavior following traumatic brain injury. Understanding the signals that initiate neuroinflammation and the context in which they appear may be critical for understanding immune cell contributions to pathology and regeneration. Within this paper, we review a number of factors that could affect immune cell behavior acutely following traumatic brain injury.
Collapse
Affiliation(s)
- Kathryn L Wofford
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma, and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA; School of Biochemistry and Immunology and Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - D Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
39
|
Webster KM, Sun M, Crack PJ, O'Brien TJ, Shultz SR, Semple BD. Age-dependent release of high-mobility group box protein-1 and cellular neuroinflammation after traumatic brain injury in mice. J Comp Neurol 2018; 527:1102-1117. [DOI: 10.1002/cne.24589] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/15/2018] [Accepted: 11/09/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Kyria M. Webster
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
| | - Mujun Sun
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
| | - Peter J. Crack
- Department of Pharmacology and Therapeutics; The University of Melbourne; Parkville Victoria Australia
| | - Terence J. O'Brien
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
- Department of Neuroscience; Monash University; Melbourne Victoria Australia
| | - Sandy R. Shultz
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
- Department of Neuroscience; Monash University; Melbourne Victoria Australia
| | - Bridgette D. Semple
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
- Department of Neuroscience; Monash University; Melbourne Victoria Australia
| |
Collapse
|
40
|
Khan MT, Liu J, Nerlich J, Tang Y, Franke H, Illes P. Regulation of P2X7 receptor function of neural progenitor cells in the hippocampal subgranular zone by neuronal activity in the dentate gyrus. Neuropharmacology 2018; 140:139-149. [PMID: 30092245 DOI: 10.1016/j.neuropharm.2018.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/09/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022]
Abstract
P2X7 receptors (Rs) mediate apoptosis/necrosis in neuronal and non-neuronal systems. Patch-clamp recordings from dentate gyrus (DG) granule cells in acutely prepared hippocampal slices of mice showed that incubation with 4-aminopyridine (4-AP) causes an excitability increase. This led to an enhanced sensitivity of P2X7Rs of the underlying subgranular zone neural progenitor cells (NPCs) towards dibenzoyl-ATP (Bz-ATP). The glutamatergic agonists NMDA and AMPA, as well as the purinergic agonist ATP also increased the Bz-ATP-induced current amplitudes (IBzATP). Tetrodotoxin as well as the standard antiepileptic drugs phenytoin, valproic acid and gabapentin counteracted the effect of 4-AP, most likely by decreasing the firing rate and/or action potential duration of DG granule cells and in consequence the release of ATP/glutamate onto NPCs. Experiments with organotypic hippocampal slice cultures confirmed these results also under conditions when 4-AP was applied for longer time periods and at much lower concentrations than used in acute slices. It was concluded that pathological firing modelled by 4-AP might trigger a sensitivity increase of P2X7Rs leading to necrosis/apoptosis of NPCs with the subsequent decrease of NPC, and in consequence, granule cell number. Hence, supersensitive P2X7Rs may exert a beneficial counter-regulatory effect by reducing the chances for the evolution of chronic temporal lobe epilepsy by ectopically located granule cells.
Collapse
Affiliation(s)
- Muhammad Tahir Khan
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, 04107, Leipzig, Germany
| | - Juan Liu
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, 04107, Leipzig, Germany; Acupuncture and Tuina School, Chengdu University of TCM, 610075, Chengdu, China
| | - Jana Nerlich
- Carl-Ludwig-Institut für Physiologie, Universität Leipzig, 04103, Leipzig, Germany
| | - Yong Tang
- Acupuncture and Tuina School, Chengdu University of TCM, 610075, Chengdu, China
| | - Heike Franke
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, 04107, Leipzig, Germany
| | - Peter Illes
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, 04107, Leipzig, Germany; Acupuncture and Tuina School, Chengdu University of TCM, 610075, Chengdu, China.
| |
Collapse
|
41
|
Haber M, Amyot F, Kenney K, Meredith-Duliba T, Moore C, Silverman E, Podell J, Chou YY, Pham DL, Butman J, Lu H, Diaz-Arrastia R, Sandsmark D. Vascular Abnormalities within Normal Appearing Tissue in Chronic Traumatic Brain Injury. J Neurotrauma 2018; 35:2250-2258. [PMID: 29609518 DOI: 10.1089/neu.2018.5684] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Magnetic resonance imaging (MRI) is a powerful tool for visualizing traumatic brain injury(TBI)-related lesions. Trauma-induced encephalomalacia is frequently identified by its hyperintense appearance on fluid-attenuated inversion recovery (FLAIR) sequences. In addition to parenchymal lesions, TBI commonly results in cerebral microvascular injury, but its anatomical relationship to parenchymal encephalomalacia is not well characterized. The current study utilized a multi-modal MRI protocol to assess microstructural tissue integrity (by mean diffusivity [MD] and fractional aniosotropy [FA]) and altered vascular function (by cerebral blood flow [CBF] and cerebral vascular reactivity [CVR]) within regions of visible encephalomalacia and normal appearing tissue in 27 chronic TBI (minimum 6 months post-injury) subjects. Fifteen subjects had visible encephalomalacias whereas 12 did not have evident lesions on MRI. Imaging from 14 age-matched healthy volunteers were used as controls. CBF was assessed by arterial spin labeling (ASL) and CVR by measuring the change in blood-oxygen-level-dependent (BOLD) MRI during a hypercapnia challenge. There was a significant reduction in FA, CBF, and CVR with a complementary increase in MD within regions of FLAIR-visible encephalomalacia (p < 0.05 for all comparisons). In normal-appearing brain regions, only CVR was significantly reduced relative to controls (p < 0.05). These findings indicate that vascular dysfunction represents a TBI endophenotype that is distinct from structural injury detected using conventional MRI, may be present even in the absence of visible structural injury, and persists long after trauma. CVR may serve as a useful diagnostic and pharmacodynamic imaging biomarker of traumatic microvascular injury.
Collapse
Affiliation(s)
- Margalit Haber
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Franck Amyot
- 6 National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Kimbra Kenney
- 2 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Tawny Meredith-Duliba
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Carol Moore
- 2 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Erika Silverman
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Jamie Podell
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Yi-Yu Chou
- 3 Center for Neuroscience and Regenerative Medicine , Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | - Dzung L Pham
- 3 Center for Neuroscience and Regenerative Medicine , Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland
| | - John Butman
- 4 National Institutes of Health , Clinical Center, Radiology and Imaging Sciences, Bethesda, Maryland
| | - Hanzhang Lu
- 5 Department of Radiology, Johns Hopkins University Baltimore , Maryland
| | - Ramon Diaz-Arrastia
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Danielle Sandsmark
- 1 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| |
Collapse
|
42
|
Liu X, Quan N. Microglia and CNS Interleukin-1: Beyond Immunological Concepts. Front Neurol 2018; 9:8. [PMID: 29410649 PMCID: PMC5787061 DOI: 10.3389/fneur.2018.00008] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022] Open
Abstract
Activation of microglia and expression of the inflammatory cytokine interleukin-1 (IL-1) in the CNS have become almost synonymous with neuroinflammation. In numerous studies, increased CNS IL-1 expression and altered microglial morphology have been used as hallmarks of CNS inflammation. A central concept of how CNS IL-1 and microglia influence functions of the nervous system was derived from the notion initially generated in the peripheral immune system: IL-1 stimulates monocyte/macrophage (the peripheral counterparts of microglia) to amplify inflammation. It is increasingly clear, however, CNS IL-1 acts on other targets in the CNS and microglia participates in many neural functions that are not related to immunological activities. Further, CNS exhibits immunological privilege (although not as absolute as previously thought), rendering amplification of inflammation within CNS under stringent control. This review will analyze current literature to evaluate the contribution of immunological and non-immunological aspects of microglia/IL-1 interaction in the CNS to gain insights for how these aspects might affect health and disease in the nervous tissue.
Collapse
Affiliation(s)
- Xiaoyu Liu
- College of Medicine, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Ning Quan
- College of Medicine, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
43
|
Effect of Low-Dose Alcohol Consumption on Inflammation Following Transient Focal Cerebral Ischemia in Rats. Sci Rep 2017; 7:12547. [PMID: 28970514 PMCID: PMC5624984 DOI: 10.1038/s41598-017-12720-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/14/2017] [Indexed: 01/14/2023] Open
Abstract
Increasing evidence suggest that low-dose alcohol consumption (LAC) reduces the incidence and improves the functional outcome of ischemic stroke. We determined the influence of LAC on post-ischemic inflammation. Male Sprague-Dawley rats were divided into 3 groups, an ethanol (13.5% alcohol) group, a red wine (Castle Rock Pinot Noir, 13.5% alcohol) group, and a control group. The amount of alcohol given to red wine and ethanol groups was 1.4 g/kg/day. After 8 weeks, the animals were subjected to a 2-hour middle cerebral artery occlusion (MCAO) and sacrificed at 24 hours of reperfusion. Cerebral ischemia/reperfusion (I/R) injury, expression of adhesion molecules and pro- and anti-inflammatory cytokines/chemokines, microglial activation and neutrophil infiltration were evaluated. The total infarct volume and neurological deficits were significantly reduced in red wine- and ethanol-fed rats compared to control rats. Both red wine and ethanol suppressed post-ischemic expression of adhesion molecules and microglial activation. In addition, both red wine and ethanol upregulated expression of tissue inhibitor of metalloproteinases 1 (TIMP-1), downregulated expression of proinflammatory cytokines/chemokines, and significantly alleviated post-ischemic expression of inflammatory mediators. Furthermore, red wine significantly reduced post-ischemic neutrophil infiltration. Our findings suggest that LAC may protect the brain against its I/R injury by suppressing post-ischemic inflammation.
Collapse
|
44
|
Kenzie ES, Parks EL, Bigler ED, Lim MM, Chesnutt JC, Wakeland W. Concussion As a Multi-Scale Complex System: An Interdisciplinary Synthesis of Current Knowledge. Front Neurol 2017; 8:513. [PMID: 29033888 PMCID: PMC5626937 DOI: 10.3389/fneur.2017.00513] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/13/2017] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) has been called "the most complicated disease of the most complex organ of the body" and is an increasingly high-profile public health issue. Many patients report long-term impairments following even "mild" injuries, but reliable criteria for diagnosis and prognosis are lacking. Every clinical trial for TBI treatment to date has failed to demonstrate reliable and safe improvement in outcomes, and the existing body of literature is insufficient to support the creation of a new classification system. Concussion, or mild TBI, is a highly heterogeneous phenomenon, and numerous factors interact dynamically to influence an individual's recovery trajectory. Many of the obstacles faced in research and clinical practice related to TBI and concussion, including observed heterogeneity, arguably stem from the complexity of the condition itself. To improve understanding of this complexity, we review the current state of research through the lens provided by the interdisciplinary field of systems science, which has been increasingly applied to biomedical issues. The review was conducted iteratively, through multiple phases of literature review, expert interviews, and systems diagramming and represents the first phase in an effort to develop systems models of concussion. The primary focus of this work was to examine concepts and ways of thinking about concussion that currently impede research design and block advancements in care of TBI. Results are presented in the form of a multi-scale conceptual framework intended to synthesize knowledge across disciplines, improve research design, and provide a broader, multi-scale model for understanding concussion pathophysiology, classification, and treatment.
Collapse
Affiliation(s)
- Erin S. Kenzie
- Systems Science Program, Portland State University, Portland, OR, United States
| | - Elle L. Parks
- Systems Science Program, Portland State University, Portland, OR, United States
| | - Erin D. Bigler
- Department of Psychology and Neuroscience Center, Brigham Young University, Provo, UT, United States
| | - Miranda M. Lim
- Sleep Disorders Clinic, Division of Hospital and Specialty Medicine, Veterans Affairs Portland Health Care System, Portland, OR, United States
- Departments of Neurology, Medicine, and Behavioral Neuroscience, and Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - James C. Chesnutt
- TBI/Concussion Program, Orthopedics & Rehabilitation and Family Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Wayne Wakeland
- Systems Science Program, Portland State University, Portland, OR, United States
| |
Collapse
|
45
|
Defining an Analytic Framework to Evaluate Quantitative MRI Markers of Traumatic Axonal Injury: Preliminary Results in a Mouse Closed Head Injury Model. eNeuro 2017; 4:eN-NWR-0164-17. [PMID: 28966972 PMCID: PMC5616192 DOI: 10.1523/eneuro.0164-17.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/17/2017] [Accepted: 08/05/2017] [Indexed: 01/11/2023] Open
Abstract
Diffuse axonal injury (DAI) is a hallmark of traumatic brain injury (TBI) pathology. Recently, the Closed Head Injury Model of Engineered Rotational Acceleration (CHIMERA) was developed to generate an experimental model of DAI in a mouse. The characterization of DAI using diffusion tensor magnetic resonance imaging (MRI; diffusion tensor imaging, DTI) may provide a useful set of outcome measures for preclinical and clinical studies. The objective of this study was to identify the complex neurobiological underpinnings of DTI features following DAI using a comprehensive and quantitative evaluation of DTI and histopathology in the CHIMERA mouse model. A consistent neuroanatomical pattern of pathology in specific white matter tracts was identified across ex vivo DTI maps and photomicrographs of histology. These observations were confirmed by voxelwise and regional analysis of DTI maps, demonstrating reduced fractional anisotropy (FA) in distinct regions such as the optic tract. Similar regions were identified by quantitative histology and exhibited axonal damage as well as robust gliosis. Additional analysis using a machine-learning algorithm was performed to identify regions and metrics important for injury classification in a manner free from potential user bias. This analysis found that diffusion metrics were able to identify injured brains almost with the same degree of accuracy as the histology metrics. Good agreement between regions detected as abnormal by histology and MRI was also found. The findings of this work elucidate the complexity of cellular changes that give rise to imaging abnormalities and provide a comprehensive and quantitative evaluation of the relative importance of DTI and histological measures to detect brain injury.
Collapse
|
46
|
Ziebell JM, Ray-Jones H, Lifshitz J. Nogo presence is inversely associated with shifts in cortical microglial morphology following experimental diffuse brain injury. Neuroscience 2017; 359:209-223. [PMID: 28736137 DOI: 10.1016/j.neuroscience.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/28/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
Abstract
Diffuse traumatic brain injury (TBI) initiates secondary pathology, including inflammation and reduced myelination. Considering these injury-related pathologies, the many states of activated microglia as demonstrated by differing morphologies would form, migrate, and function in and through fields of growth-inhibitory myelin byproduct, specifically Nogo. Here we evaluate the relationship between inflammation and reduced myelin antigenicity in the wake of diffuse TBI and present the hypothesis that the Nogo-66 receptor antagonist peptide NEP(1-40) would reverse the injury-induced shift in distribution of microglia morphologies by limiting myelin-based inhibition. Adult male rats were subjected to midline fluid percussion sham or brain injury. At 2h, 6h, 1d, 2d, 7d, and 21d post-injury, immunohistochemical staining was analyzed in sensory cortex (S1BF) for myelin antigens (myelin basic protein; MBP and CNPase), microglia morphology (ionized calcium-binding adapter protein; Iba1), Nogo receptor and Nogo. Pronounced reduction in myelin antigenicity was evident transiently at 1d post-injury, as evidenced by decreased MBP and CNPase staining, as well as loss of white matter organization, compared to sham and later injury time points. Concomitant with reduced myelin antigenicity, injury shifted microglia morphology from the predominantly ramified morphology observed in sham-injured cortex to hyper-ramified, activated, fully activated, or rod. Changes in microglial morphology were evident as early as 2h post-injury, and remained at least until day 21. Additional cohorts of uninjured and brain-injured animals received vehicle or drug (NEP(1-40), i.p., 15min and 19h post-injury) and brains were collected at 2h, 6h, 1d, 2d, or 7d post-injury. NEP(1-40) administration further shifted distributions of microglia away from an injury-induced activated morphology toward greater proportions of rod and macrophage-like morphologies compared to vehicle-treated. By 7d post-injury, no differences in the distributions of microglia were noted between vehicle and NEP(1-40). This study begins to link secondary pathologies of white matter damage and inflammation after diffuse TBI. In the injured brain, secondary pathologies co-occur and likely interact, with consequences for neuronal circuit disruption leading to neurological symptoms.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| | - Helen Ray-Jones
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Department of Biology and Biochemistry, University of Bath, Bath, England, UK
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; VA Healthcare System, Phoenix, AZ, USA; Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
47
|
Hutchinson EB, Schwerin SC, Avram AV, Juliano SL, Pierpaoli C. Diffusion MRI and the detection of alterations following traumatic brain injury. J Neurosci Res 2017; 96:612-625. [PMID: 28609579 PMCID: PMC5729069 DOI: 10.1002/jnr.24065] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 12/18/2022]
Abstract
This article provides a review of brain tissue alterations that may be detectable using diffusion magnetic resonance imaging MRI (dMRI) approaches and an overview and perspective on the modern dMRI toolkits for characterizing alterations that follow traumatic brain injury (TBI). Noninvasive imaging is a cornerstone of clinical treatment of TBI and has become increasingly used for preclinical and basic research studies. In particular, quantitative MRI methods have the potential to distinguish and evaluate the complex collection of neurobiological responses to TBI arising from pathology, neuroprotection, and recovery. dMRI provides unique information about the physical environment in tissue and can be used to probe physiological, architectural, and microstructural features. Although well‐established approaches such as diffusion tensor imaging are known to be highly sensitive to changes in the tissue environment, more advanced dMRI techniques have been developed that may offer increased specificity or new information for describing abnormalities. These tools are promising, but incompletely understood in the context of TBI. Furthermore, model dependencies and relative limitations may impact the implementation of these approaches and the interpretation of abnormalities in their metrics. The objective of this paper is to present a basic review and comparison across dMRI methods as they pertain to the detection of the most commonly observed tissue and cellular alterations following TBI.
Collapse
Affiliation(s)
- Elizabeth B Hutchinson
- Quantitative Medical Imaging Section, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland.,Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland
| | - Susan C Schwerin
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, Maryland.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Alexandru V Avram
- Quantitative Medical Imaging Section, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland.,Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Sharon L Juliano
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Carlo Pierpaoli
- Quantitative Medical Imaging Section, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
48
|
Au NPB, Ma CHE. Recent Advances in the Study of Bipolar/Rod-Shaped Microglia and their Roles in Neurodegeneration. Front Aging Neurosci 2017; 9:128. [PMID: 28522972 PMCID: PMC5415568 DOI: 10.3389/fnagi.2017.00128] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/18/2017] [Indexed: 01/06/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and they contribute to primary inflammatory responses following CNS injuries. The morphology of microglia is closely associated with their functional activities. Most previous research efforts have attempted to delineate the role of ramified and amoeboid microglia in the pathogenesis of neurodegenerative diseases. In addition to ramified and amoeboid microglia, bipolar/rod-shaped microglia were first described by Franz Nissl in 1899 and their presence in the brain was closely associated with the pathology of infectious diseases and sleeping disorders. However, studies relating to bipolar/rod-shaped microglia are very limited, largely due to the lack of appropriate in vitro and in vivo experimental models. Recent studies have reported the formation of bipolar/rod-shaped microglia trains in in vivo models of CNS injury, including diffuse brain injury, focal transient ischemia, optic nerve transection and laser-induced ocular hypertension (OHT). These bipolar/rod-shaped microglia formed end-to-end alignments in close proximity to the adjacent injured axons, but they showed no interactions with blood vessels or other types of glial cell. Recent studies have also reported on a highly reproducible in vitro culture model system to enrich bipolar/rod-shaped microglia that acts as a powerful tool with which to characterize this form of microglia. The molecular aspects of bipolar/rod-shaped microglia are of great interest in the field of CNS repair. This review article focuses on studies relating to the morphology and transformation of microglia into the bipolar/rod-shaped form, along with the differential gene expression and spatial distribution of bipolar/rod-shaped microglia in normal and pathological CNSs. The spatial arrangement of bipolar/rod-shaped microglia is crucial in the reorganization and remodeling of neuronal and synaptic circuitry following CNS injuries. Finally, we discuss the potential neuroprotective roles of bipolar/rod-shaped microglia, and the possibility of transforming ramified/amoeboid microglia into bipolar/rod-shaped microglia. This will be of considerable clinical benefit in the development of novel therapeutic strategies for treating various neurodegenerative diseases and promoting CNS repair after injury.
Collapse
Affiliation(s)
- Ngan Pan Bennett Au
- Department of Biomedical Sciences, City University of Hong KongKowloon Tong, Hong Kong
| | - Chi Him Eddie Ma
- Department of Biomedical Sciences, City University of Hong KongKowloon Tong, Hong Kong.,Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong KongKowloon Tong, Hong Kong.,State Key Laboratory in Marine Pollution, City University of Hong KongKowloon Tong, Hong Kong
| |
Collapse
|
49
|
Simon DW, McGeachy M, Bayır H, Clark RS, Loane DJ, Kochanek PM. The far-reaching scope of neuroinflammation after traumatic brain injury. Nat Rev Neurol 2017; 13:171-191. [PMID: 28186177 PMCID: PMC5675525 DOI: 10.1038/nrneurol.2017.13] [Citation(s) in RCA: 625] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The 'silent epidemic' of traumatic brain injury (TBI) has been placed in the spotlight as a result of clinical investigations and popular press coverage of athletes and veterans with single or repetitive head injuries. Neuroinflammation can cause acute secondary injury after TBI, and has been linked to chronic neurodegenerative diseases; however, anti-inflammatory agents have failed to improve TBI outcomes in clinical trials. In this Review, we therefore propose a new framework of targeted immunomodulation after TBI for future exploration. Our framework incorporates factors such as the time from injury, mechanism of injury, and secondary insults in considering potential treatment options. Structuring our discussion around the dynamics of the immune response to TBI - from initial triggers to chronic neuroinflammation - we consider the ability of soluble and cellular inflammatory mediators to promote repair and regeneration versus secondary injury and neurodegeneration. We summarize both animal model and human studies, with clinical data explicitly defined throughout this Review. Recent advances in neuroimmunology and TBI-responsive neuroinflammation are incorporated, including concepts of inflammasomes, mechanisms of microglial polarization, and glymphatic clearance. Moreover, we highlight findings that could offer novel therapeutic targets for translational and clinical research, assimilate evidence from other brain injury models, and identify outstanding questions in the field.
Collapse
Affiliation(s)
- Dennis W. Simon
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mandy McGeachy
- Department of Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Hülya Bayır
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Robert S.B. Clark
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Anesthesiology, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MA 21201, USA
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Anesthesiology, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; The Children’s Hospital of Pittsburgh of UPMC, and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
50
|
Does time heal all wounds? Experimental diffuse traumatic brain injury results in persisting histopathology in the thalamus. Behav Brain Res 2016; 340:137-146. [PMID: 28042008 DOI: 10.1016/j.bbr.2016.12.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/30/2016] [Accepted: 12/28/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Thalamic dysfunction has been implicated in overall chronic neurological dysfunction after traumatic brain injury (TBI), however little is known about the underlying histopathology. In experimental diffuse TBI (dTBI), we hypothesize that persisting histopathological changes in the ventral posteromedial (VPM) nucleus of the thalamus is indicative of progressive circuit reorganization. Since circuit reorganization in the VPM impacts the whisker sensory system, the histopathology could explain the development of hypersensitivity to whisker stimulation by 28days post-injury; similar to light and sound hypersensitivity in human TBI survivors. METHODS Adult, male Sprague-Dawley rats underwent craniotomy and midline fluid percussion injury (FPI) (moderate severity; 1.8-2.0atm) or sham surgery. At 1d, 7d, and 28days post-FPI (d FPI) separate experiments confirmed the cytoarchitecture (Giemsa stain) and evaluated neuropathology (silver stain), activated astrocytes (GFAP), neuron morphology (Golgi stain) and microglial morphology (Iba-1) in the VPM. RESULTS Cytoarchitecture was unchanged throughout the time course, similar to previously published data; however, neuropathology and astrocyte activation were significantly increased at 7d and 28d and activated microglia were present at all time points. Neuron morphology was dynamic over the time course with decreased dendritic complexity (fewer branch points; decreased length of processes) at 7d FPI and return to sham values by 28d FPI. CONCLUSIONS These data indicate that dTBI results in persisting thalamic histopathology out to a chronic time point. While these changes can be indicative of either adaptive (recovery) or maladaptive (neurological dysfunction) circuit reorganization, they also provide a potential mechanism by which maladaptive circuit reorganization could contribute to the development of chronic neurological dysfunction. Understanding the processes that mediate circuit reorganization is critical to the development of future therapies for TBI patients.
Collapse
|