1
|
Song J, Zeng J, Chen X, Wang J, Zhang Y, Gao Y, Wang R, Jiang N, Lin Y, Li R. Anti-neuroinflammatory agent rhein lysinate-based self-assembled injectable hydrogel loaded with ZL006 for promoting post-stroke functional recovery. Biomaterials 2025; 318:123124. [PMID: 39884131 DOI: 10.1016/j.biomaterials.2025.123124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/27/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
The therapeutic agent-based self-assembled hydrogel is gaining interest for biomedical applications, because it overcomes the poor biodegradability and low therapeutic agent loading of conventional polymer gelator-based hydrogel. Here, we present rhein lysinate (RHL), a therapeutic agent that self-assembles to form a stable hydrogel through the π-π stacking and hydrogen bonding interactions, while also exerting anti-neuroinflammatory effect. As a small molecular hydrogelator, RHL has significantly improved water solubility and enhanced self-assembly and gelation capabilities compared to the natural anthraquinone rhein. The relaxed gel-forming conditions enhance the practical application potential of self-assembled hydrogel of RHL (RHL gel). The RHL gel can be loaded with the bioactive agents such as 5-Fluorouracil, temozolomide, edaravone, and ZL006, mainly based on efficient stacking between aromatic rings in the bioactive agents and anthraquinone rings in the hydrogel network structure. The pre-gelled RHL gel and ZL006-loaded RHL gel (ZL006-RHL gel) exhibit shear-thinning behavior, flowing like a liquid under high shear stress during injection. Once this shear stress is removal within the body, they rapidly recover to the initial solid-like state. When a single dose of ZL006-RHL gel is administrated to stroke cavity in the subacute phase of stroke, RHL gel matrix effectively reduces post-stroke neuroinflammation, creates a favorable environment for ZL006 to enhance neuroplasticity, and confers a sustained and stable action to ZL006, leading to a long-lasting improvement of motor performance. This study may provide a valuable strategy for therapeutic intervention to promote post-stroke functional recovery, for which there are no clinically available drugs.
Collapse
Affiliation(s)
- Jiamei Song
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jiaqi Zeng
- The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, China
| | - Xi Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jiayu Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ying Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yuhao Gao
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ruiqi Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Nan Jiang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; National Vaccine Innovation Platform, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Yuhui Lin
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Rui Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
2
|
Jiao H, Kalsbeek A, Yi CX. Microglia, circadian rhythm and lifestyle factors. Neuropharmacology 2024; 257:110029. [PMID: 38852838 DOI: 10.1016/j.neuropharm.2024.110029] [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: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Microglia, a vital homeostasis-keeper of the central nervous system, perform critical functions such as synaptic pruning, clearance of cellular debris, and participation in neuroinflammatory processes. Recent research has shown that microglia exhibit strong circadian rhythms that not only actively regulate their own immune activity, but also affect neuronal function. Disruptions of the circadian clock have been linked to a higher risk of developing a variety of diseases. In this article we will provide an overview of how lifestyle factors impact microglial function, with a focus on disruptions caused by irregular sleep-wake patterns, reduced physical activity, and eating at the wrong time-of-day. We will also discuss the potential connection between these lifestyle factors, disrupted circadian rhythms, and the role of microglia in keeping brain health. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Han Jiao
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Kim J, Sullivan O, Lee K, Jao J, Tamayo J, Madany AM, Wong B, Ashwood P, Ciernia AV. Repeated LPS induces training and tolerance of microglial responses across brain regions. J Neuroinflammation 2024; 21:233. [PMID: 39304952 PMCID: PMC11414187 DOI: 10.1186/s12974-024-03198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Neuroinflammation is involved in the pathogenesis of almost every central nervous system disorder. As the brain's innate immune cells, microglia fine tune their activity to a dynamic brain environment. Previous studies have shown that repeated bouts of peripheral inflammation can trigger long-term changes in microglial gene expression and function, a form of innate immune memory. METHODS AND RESULTS In this study, we used multiple low-dose lipopolysaccharide (LPS) injections in adult mice to study the acute cytokine, transcriptomic, and microglia morphological changes that contribute to the formation of immune memory in the frontal cortex, hippocampus, and striatum, as well as the long-term effects of these changes on behavior. Training and tolerance of gene expression was shared across regions, and we identified 3 unique clusters of DEGs (2xLPS-sensitive, 4xLPS-sensitive, LPS-decreased) enriched for different biological functions. 2xLPS-sensitive DEG promoters were enriched for binding sites for IRF and NFkB family transcription factors, two key regulators of innate immune memory. We quantified shifts in microglia morphological populations and found that while the proportion of ramified and rod-like microglia mostly remained consistent within brain regions and sexes with LPS treatment, there was a shift from ameboid towards hypertrophic morphological states across immune memory states and a dynamic emergence and resolution of events of microglia aligning end-to-end with repeated LPS. CONCLUSIONS Together, findings support the dynamic regulation of microglia during the formation of immune memories in the brain and support future work to exploit this model in brain disease contexts.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Justin Jao
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Juan Tamayo
- MIND Institute, University of California Davis, Davis, USA
| | | | - Brandon Wong
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Paul Ashwood
- MIND Institute, University of California Davis, Davis, USA
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.
| |
Collapse
|
4
|
Retinasamy T, Lee ALY, Lee HS, Lee VLL, Shaikh MF, Yeong KY. Repurposing Anakinra for Alzheimer's Disease: The In Vitro and In Vivo Effects of Anakinra on LPS- and AC-Induced Neuroinflammation. ACS Chem Neurosci 2024; 15:3298-3310. [PMID: 39213521 DOI: 10.1021/acschemneuro.4c00205] [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] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease is a significant global health issue, and studies suggest that neuroinflammation plays a vital role in the advancement of this disease. In this study, anakinra has been shown to display a time- and concentration-dependent antineuroinflammatory effect. In the in vitro studies, it diminished the gene expressions of tumor necrosis factor-alpha (TNF-α) and nitric oxide (NO) synthase 2 stimulated by lipopolysaccharide (LPS). Anakinra also reduced the LPS-induced production of NO and reactive oxygen species. Thus, the hypertrophic state of LPS-activated BV2 microglial cells was reversed by anakinra. Furthermore, acrylamide (ACR)-induced activation of nuclear transcription factor-κB, TNF-α, and interleukin-1β was downregulated, while cAMP response element binding protein and brain-derived neurotrophic factor expression levels were markedly enhanced in ACR-treated zebrafish larvae. It was also observed that anakinra improved the uncoordinated swimming behaviors in ACR-exposed zebrafish larvae. Overall, anakinra demonstrated potential antineuroinflammatory and antioxidative effects.
Collapse
Affiliation(s)
- Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Amber Lot Yee Lee
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia
| | - Hsien Siang Lee
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Vanessa Lin Lin Lee
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange 2795, NSW, Australia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia
| |
Collapse
|
5
|
Casadei M, Miguel B, Rubione J, Fiore E, Mengelle D, Guerri-Guttenberg RA, Montaner A, Villar MJ, Constandil-Córdova L, Romero-Sandoval AE, Brumovsky PR. Mesenchymal Stem Cell Engagement Modulates Neuroma Microenviroment in Rats and Humans and Prevents Postamputation Pain. THE JOURNAL OF PAIN 2024; 25:104508. [PMID: 38484854 PMCID: PMC11283994 DOI: 10.1016/j.jpain.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/11/2024]
Abstract
Postamputation pain is currently managed unsatisfactorily with neuron-targeted pharmacological and interventional therapies. Non-neuronal pain mechanisms have emerged as crucial factors in the development and persistence of postamputation pain. Consequently, these mechanisms offer exciting prospects as innovative therapeutic targets. We examined the hypothesis that engaging mesenchymal stem cells (MSCs) would foster local neuroimmune interactions, leading to a potential reduction in postamputation pain. We utilized an ex vivo neuroma model from a phantom limb pain patient to uncover that the oligodeoxynucleotide IMT504 engaged human primary MSCs to promote an anti-inflammatory microenvironment. Reverse translation experiments recapitulated these effects. Thus, in an in vivo rat model, IMT504 exhibited strong efficacy in preventing autotomy (self-mutilation) behaviors. This effect was linked to a substantial accumulation of MSCs in the neuroma and associated dorsal root ganglia and the establishment of an anti-inflammatory phenotype in these compartments. Centrally, this intervention reduced glial reactivity in the dorsal horn spinal cord, demonstrating diminished nociceptive activity. Accordingly, the exogenous systemic administration of MSCs phenocopied the behavioral effects of IMT504. Our findings underscore the mechanistic relevance of MSCs and the translational therapeutic potential of IMT504 to engage non-neuronal cells for the prevention of postamputation pain. PERSPECTIVE: The present study suggests that IMT504-dependent recruitment of endogenous MSCs within severely injured nerves may prevent post-amputation pain by modifying the inflammatory scenario at relevant sites in the pain pathway. Reinforcing data in rat and human tissues supports the potential therapeutic value of IMT504 in patients suffering postamputation pain.
Collapse
Affiliation(s)
- Mailín Casadei
- Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| | - Bernardo Miguel
- Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| | - Julia Rubione
- Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| | - Esteban Fiore
- Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| | - Diego Mengelle
- Hospital Universitario Austral, Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| | | | - Alejandro Montaner
- Instituto de Ciencia y Tecnología “César Milstein”, CONICET-Fundación Pablo Cassará, Buenos Aires, Argentina, C1440FFX
| | - Marcelo J. Villar
- Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| | | | | | - Pablo R. Brumovsky
- Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina, B1629AHJ
| |
Collapse
|
6
|
Boese M, Berman RY, Qiu J, Spencer HF, Radford KD, Choi KH. Effects of Mild Closed-Head Injury and Subanesthetic Ketamine Infusion on Microglia, Axonal Injury, and Synaptic Density in Sprague-Dawley Rats. Int J Mol Sci 2024; 25:4287. [PMID: 38673871 PMCID: PMC11050690 DOI: 10.3390/ijms25084287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Mild traumatic brain injury (mTBI) affects millions of people in the U.S. Approximately 20-30% of those individuals develop adverse symptoms lasting at least 3 months. In a rat mTBI study, the closed-head impact model of engineered rotational acceleration (CHIMERA) produced significant axonal injury in the optic tract (OT), indicating white-matter damage. Because retinal ganglion cells project to the lateral geniculate nucleus (LGN) in the thalamus through the OT, we hypothesized that synaptic density may be reduced in the LGN of rats following CHIMERA injury. A modified SEQUIN (synaptic evaluation and quantification by imaging nanostructure) method, combined with immunofluorescent double-labeling of pre-synaptic (synapsin) and post-synaptic (PSD-95) markers, was used to quantify synaptic density in the LGN. Microglial activation at the CHIMERA injury site was determined using Iba-1 immunohistochemistry. Additionally, the effects of ketamine, a potential neuroprotective drug, were evaluated in CHIMERA-induced mTBI. A single-session repetitive (ssr-) CHIMERA (3 impacts, 1.5 joule/impact) produced mild effects on microglial activation at the injury site, which was significantly enhanced by post-injury intravenous ketamine (10 mg/kg) infusion. However, ssr-CHIMERA did not alter synaptic density in the LGN, although ketamine produced a trend of reduction in synaptic density at post-injury day 4. Further research is necessary to characterize the effects of ssr-CHIMERA and subanesthetic doses of intravenous ketamine on different brain regions and multiple time points post-injury. The current study demonstrates the utility of the ssr-CHIMERA as a rodent model of mTBI, which researchers can use to identify biological mechanisms of mTBI and to develop improved treatment strategies for individuals suffering from head trauma.
Collapse
Affiliation(s)
- Martin Boese
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Rina Y. Berman
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Jennifer Qiu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA;
| | - Haley F. Spencer
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Kennett D. Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Kwang H. Choi
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA;
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
- Department of Psychiatry, F. E. Hébert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
7
|
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
|
8
|
Virtuoso A, Galanis C, Lenz M, Papa M, Vlachos A. Regional Microglial Response in Entorhino-Hippocampal Slice Cultures to Schaffer Collateral Lesion and Metalloproteinases Modulation. Int J Mol Sci 2024; 25:2346. [PMID: 38397023 PMCID: PMC10889226 DOI: 10.3390/ijms25042346] [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: 01/12/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Microglia and astrocytes are essential in sustaining physiological networks in the central nervous system, with their ability to remodel the extracellular matrix, being pivotal for synapse plasticity. Recent findings have challenged the traditional view of homogenous glial populations in the brain, uncovering morphological, functional, and molecular heterogeneity among glial cells. This diversity has significant implications for both physiological and pathological brain states. In the present study, we mechanically induced a Schaffer collateral lesion (SCL) in mouse entorhino-hippocampal slice cultures to investigate glial behavior, i.e., microglia and astrocytes, under metalloproteinases (MMPs) modulation in the lesioned area, CA3, and the denervated region, CA1. We observed distinct response patterns in the microglia and astrocytes 3 days after the lesion. Notably, GFAP-expressing astrocytes showed no immediate changes post-SCL. Microglia responses varied depending on their anatomical location, underscoring the complexity of the hippocampal neuroglial network post-injury. The MMPs inhibitor GM6001 did not affect microglial reactions in CA3, while increasing the number of Iba1-expressing cells in CA1, leading to a withdrawal of their primary branches. These findings highlight the importance of understanding glial regionalization following neural injury and MMPs modulation and pave the way for further research into glia-targeted therapeutic strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Neuronal Morphology Networks and Systems Biology Laboratory, Division of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (C.G.); (A.V.)
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (C.G.); (A.V.)
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (C.G.); (A.V.)
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, 30625 Hannover, Germany
| | - Michele Papa
- Neuronal Morphology Networks and Systems Biology Laboratory, Division of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (C.G.); (A.V.)
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center BrainLinks–BrainTools, University of Freiburg, 79110 Freiburg, Germany
| |
Collapse
|
9
|
Nagata S, Yamasaki R, Takase EO, Iida K, Watanabe M, Masaki K, Wijering MHC, Yamaguchi H, Kira JI, Isobe N. Iguratimod Ameliorates the Severity of Secondary Progressive Multiple Sclerosis in Model Mice by Directly Inhibiting IL-6 Production and Th17 Cell Migration via Mitigation of Glial Inflammation. BIOLOGY 2023; 12:1217. [PMID: 37759616 PMCID: PMC10525689 DOI: 10.3390/biology12091217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
We previously reported a novel secondary progressive multiple sclerosis (SPMS) model, progressive experimental autoimmune encephalomyelitis (pEAE), in oligodendroglia-specific Cx47-inducible conditional knockout (Cx47 icKO) mice. Based on our prior study showing the efficacy of iguratimod (IGU), an antirheumatic drug, for acute EAE treatment, we aimed to elucidate the effect of IGU on the SPMS animal model. We induced pEAE by immunizing Cx47 icKO mice with myelin oligodendrocyte glycoprotein peptide 35-55. IGU was orally administered from 17 to 50 days post-immunization. We also prepared a primary mixed glial cell culture and measured cytokine levels in the culture supernatant after stimulation with designated cytokines (IL-1α, C1q, TNF-α) and lipopolysaccharide. A migration assay was performed to evaluate the effect of IGU on the migration ability of T cells toward mixed glial cell cultures. IGU treatment ameliorated the clinical signs of pEAE, decreased the demyelinated area, and attenuated glial inflammation on immunohistochemical analysis. Additionally, IGU decreased the intrathecal IL-6 level and infiltrating Th17 cells. The migration assay revealed reduced Th17 cell migration and IL-6 levels in the culture supernatant after IGU treatment. Collectively, IGU successfully mitigated the clinical signs of pEAE by suppressing Th17 migration through inhibition of IL-6 production by proinflammatory-activated glial cells.
Collapse
Affiliation(s)
- Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ezgi Ozdemir Takase
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kotaro Iida
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Marion Heleen Cathérine Wijering
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen (UMCG), MS Center Noord Nederland, 9713 AV Groningen, The Netherlands
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka 811-0213, Japan
| | - Jun-ichi Kira
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka 831-8501, Japan
- Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, Fukuoka 810-0022, Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
10
|
Szabo M, Lajkó N, Dulka K, Barczánfalvi G, Lőrinczi B, Szatmári I, Mihály A, Vécsei L, Gulya K. The kynurenic acid analog SZR104 induces cytomorphological changes associated with the anti-inflammatory phenotype in cultured microglia. Sci Rep 2023; 13:11328. [PMID: 37443330 PMCID: PMC10344911 DOI: 10.1038/s41598-023-38107-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
We previously showed the anti-inflammatory effects of kynurenic acid (KYNA) and its brain-penetrable analog N-(2-(dimethylamino)ethyl)-3-(morpholinomethyl)-4-hydroxyquinoline-2-carboxamide (SZR104) both in vivo and in vitro. Here, we identified the cytomorphological effects of KYNA and SZR104 in secondary microglial cultures established from newborn rat forebrains. We quantitatively analyzed selected morphological aspects of microglia in control (unchallenged), lipopolysaccharide (LPS)-treated (challenged), KYNA- or SZR104-treated, and LPS + KYNA or LPS + SZR104-treated cultures. Multicolor immunofluorescence labeling followed by morphometric analysis (area, perimeter, transformation index, lacunarity, density, span ratio, maximum span across the convex hull, hull circularity, hull area, hull perimeter, max/min radii, mean radius, diameter of bounding circle, fractal dimension, roughness, circularity) on binary (digital) silhouettes of the microglia revealed their morphological plasticity under experimental conditions. SZR104 and, to a lesser degree, KYNA inhibited proinflammatory phenotypic changes. For example, SZR104 treatment resulted in hypertrophied microglia characterized by a swollen cell body, enlarged perimeter, increased transformation index/decreased circularity, increased convex hull values (area, perimeter, mean radius, maximum span, diameter of the bounding circle and hull circularity), altered box-counting parameters (such as fractal dimension), and increased roughness/decreased density. Taken together, analysis of cytomorphological features could contribute to the characterization of the anti-inflammatory activity of SZR104 on cultured microglia.
Collapse
Affiliation(s)
- Melinda Szabo
- Department of Cell Biology and Molecular Medicine, University of Szeged, Somogyi utca 4., 6720, Szeged, Hungary
| | - Noémi Lajkó
- Department of Cell Biology and Molecular Medicine, University of Szeged, Somogyi utca 4., 6720, Szeged, Hungary
| | - Karolina Dulka
- Department of Cell Biology and Molecular Medicine, University of Szeged, Somogyi utca 4., 6720, Szeged, Hungary
| | - Gábor Barczánfalvi
- Department of Cell Biology and Molecular Medicine, University of Szeged, Somogyi utca 4., 6720, Szeged, Hungary
| | - Bálint Lőrinczi
- ELKH-SZTE Stereochemistry Research Group, Institute of Pharmaceutical Chemistry, University of Szeged, 6720, Szeged, Hungary
- Institute of Pharmaceutical Chemistry and Interdisciplinary Excellence Center, University of Szeged, 6720, Szeged, Hungary
| | - István Szatmári
- ELKH-SZTE Stereochemistry Research Group, Institute of Pharmaceutical Chemistry, University of Szeged, 6720, Szeged, Hungary
- Institute of Pharmaceutical Chemistry and Interdisciplinary Excellence Center, University of Szeged, 6720, Szeged, Hungary
| | - András Mihály
- Department of Anatomy, University of Szeged, 6724, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, University of Szeged, 6725, Szeged, Hungary
- ELKH-SZTE Neuroscience Research Group, Department of Neurology, Interdisciplinary Excellence Center, University of Szeged, 6725, Szeged, Hungary
| | - Karoly Gulya
- Department of Cell Biology and Molecular Medicine, University of Szeged, Somogyi utca 4., 6720, Szeged, Hungary.
| |
Collapse
|
11
|
Shi C, Zhang J, Wang H, Chen C, Han M, Gao L, Tang C, Sun P, Zhao X, Guo F, Wang Z, Abdalla M, Yang Z, Liu Y, Li A, Zhang C, Jiang X. Trojan Horse Nanocapsule Enabled In Situ Modulation of the Phenotypic Conversion of Th17 Cells to Treg Cells for the Treatment of Multiple Sclerosis in Mice. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210262. [PMID: 36575563 DOI: 10.1002/adma.202210262] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/19/2022] [Indexed: 06/17/2023]
Abstract
Th17/Treg imbalance is closely related to the occurrence and development of multiple sclerosis (MS), and the transdifferentiation of Th17 cells into Treg cells may contribute to the resolution of inflammation, presenting a therapeutic strategy for MS. To modulate this phenotypic shift in situ, a "Trojan horse"-like hybrid system, nanocapsule-coupled Th17 cells, is reported for MS treatment. Following intravenous injection into MS mice, the hybrid system efficiently transmigrates across the blood-brain barrier and homes to the inflamed MS niche. (Aminooxy)-acetic acid, a transdifferentiation inducer, is locally released upon the production of ROS and in turn taken up by Th17 cells. It is demonstrated that the Trojan horse hybrid system enables in situ phenotypic transdifferentiation of Th17 cells into anti-inflammatory Treg cells. This phenotypic conversion leads to a domino-like immune response that is conducive to MS therapy. Overall, this work highlights a new pathway for accurate modulation of the phenotypes of adoptively transferred cells in situ, from proinflammatory to anti-inflammatory for MS therapy, and may be broadly applicable for patients suffering from other autoimmune diseases.
Collapse
Affiliation(s)
- Chongdeng Shi
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Jing Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Huijun Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Chen Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Maosen Han
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Lin Gao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Chunwei Tang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Peng Sun
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250355, P. R. China
| | - Xiaotian Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Feiyue Guo
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Zhaozhong Wang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Mohnad Abdalla
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Zhenmei Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Ying Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Anning Li
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Cai Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| | - Xinyi Jiang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province, 250012, P. R. China
| |
Collapse
|
12
|
Reactive Microgliosis in Sepsis-Associated and Acute Hepatic Encephalopathies: An Ultrastructural Study. Int J Mol Sci 2022; 23:ijms232214455. [PMID: 36430933 PMCID: PMC9696099 DOI: 10.3390/ijms232214455] [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: 10/12/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
Sepsis and acute liver failure are associated with severe endogenous intoxication. Microglia, which are the resident immune brain cells, play diverse roles in central nervous system development, surveillance, and defense, as well as contributing to neuroinflammatory reactions. In particular, microglia are fundamental to the pathophysiology of reactive toxic encephalopathies. We analyzed microglial ultrastructure, morphotypes, and phagocytosis in the sensorimotor cortex of cecal ligation and puncture (CLP) and acetaminophen-induced liver failure (AILF) Wistar rats. A CLP model induced a gradual shift of ~50% of surveillant microglia to amoeboid hypertrophic-like and gitter cell-like reactive phenotypes with active phagocytosis and frequent contacts with damaged neurons. In contrast, AILF microglia exhibited amoeboid, rod-like, and hypertrophic-like reactive morphotypes with minimal indications for efficient phagocytosis, and were mostly in contact with edematous astrocytes. Close interactions of reactive microglia with neurons, astrocytes, and blood-brain barrier components reflect an active contribution of these cells to the tissue adaptation and cellular remodeling to toxic brain damage. Partial disability of reactive microglia may affect the integrity and metabolism in all tissue compartments, leading to failure of the compensatory mechanisms in acute endogenous toxic encephalopathies.
Collapse
|
13
|
Persistent muscle hyperalgesia after adolescent stress is exacerbated by a mild-nociceptive input in adulthood and is associated with microglia activation. Sci Rep 2022; 12:18324. [PMID: 36316425 PMCID: PMC9622712 DOI: 10.1038/s41598-022-21808-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
Non-specific low back pain (LBP) is a major global disease burden and childhood adversity predisposes to its development. The mechanisms are largely unknown. Here, we investigated if adversity in young rats augments mechanical hyperalgesia and how spinal cord microglia contribute to this. Adolescent rats underwent restraint stress, control animals were handled. In adulthood, all rats received two intramuscular injections of NGF/saline or both into the lumbar multifidus muscle. Stress induced in rats at adolescence lowered low back pressure pain threshold (PPT; p = 0.0001) and paw withdrawal threshold (PWT; p = 0.0007). The lowered muscle PPT persisted throughout adulthood (p = 0.012). A subsequent NGF in adulthood lowered only PPT (d = 0.87). Immunohistochemistry revealed changes in microglia morphology: stress followed by NGF induced a significant increase in ameboid state (p < 0.05). Repeated NGF injections without stress showed significantly increased cell size in surveilling and bushy states (p < 0.05). Thus, stress in adolescence induced persistent muscle hyperalgesia that can be enhanced by a mild-nociceptive input. The accompanying morphological changes in microglia differ between priming by adolescent stress and by nociceptive inputs. This novel rodent model shows that adolescent stress is a risk factor for the development of LBP in adulthood and that morphological changes in microglia are signs of spinal mechanisms involved.
Collapse
|
14
|
Sánchez KE, Bhaskar K, Rosenberg GA. Apoptosis-associated speck-like protein containing a CARD-mediated release of matrix metalloproteinase 10 stimulates a change in microglia phenotype. Front Mol Neurosci 2022; 15:976108. [PMID: 36305000 PMCID: PMC9595131 DOI: 10.3389/fnmol.2022.976108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammation contributes to amyloid-β and tau pathology in Alzheimer's disease (AD). Microglia facilitate an altered immune response that includes microgliosis, upregulation of inflammasome proteins, and elevation of matrix-metalloproteinases (MMPs). Studies of cerebrospinal fluid (CSF) and blood in dementia patients show upregulation of two potential biomarkers of inflammation at the cellular level, MMP10 and apoptosis-associated speck-like protein containing a CARD (ASC). However, little is known about their relationship in the context of brain inflammation. Therefore, we stimulated microglia cultures with purified insoluble ASC speck aggregates and MMP10 to elucidate their role. We found that ASC specks altered microglia shape and stimulated the release of MMP3 and MMP10. Furthermore, MMP10 stimulated microglia released additional MMP10 along with the inflammatory cytokines, tumor-necrosis factor-α (TNFα), Interleukin 6 (IL-6), and CXCL1 CXC motif chemokine ligand 1 (CXCL1). A broad-spectrum MMP inhibitor, GM6001, prevented TNFα release. With these results, we conclude that MMP10 and ASC specks act on microglial cells to propagate inflammation.
Collapse
Affiliation(s)
- Kathryn E. Sánchez
- Center for Memory and Aging, University of New Mexico, Albuquerque, NM, United States
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, United States
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Gary A. Rosenberg
- Center for Memory and Aging, University of New Mexico, Albuquerque, NM, United States
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
15
|
Bosch LFP, Kierdorf K. The Shape of μ—How Morphological Analyses Shape the Study of Microglia. Front Cell Neurosci 2022; 16:942462. [PMID: 35846562 PMCID: PMC9276927 DOI: 10.3389/fncel.2022.942462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Microglia, the innate immune cells of the CNS parenchyma, serve as the first line of defense in a myriad of neurodevelopmental, neurodegenerative, and neuroinflammatory conditions. In response to the peripheral inflammation, circulating mediators, and other external signals that are produced by these conditions, microglia dynamically employ different transcriptional programs as well as morphological adaptations to maintain homeostasis. To understand these cells’ function, the field has established a number of essential analysis approaches, such as gene expression, cell quantification, and morphological reconstruction. Although high-throughput approaches are becoming commonplace in regard to other types of analyses (e.g., single-cell scRNA-seq), a similar standard for morphological reconstruction has yet to be established. In this review, we offer an overview of microglial morphological analysis methods, exploring the advantages and disadvantages of each, highlighting a number of key studies, and emphasizing how morphological analysis has significantly contributed to our understanding of microglial function in the CNS parenchyma. In doing so, we advocate for the use of unbiased, automated morphological reconstruction approaches in future studies, in order to capitalize on the valuable information embedded in the cellular structures microglia inhabit.
Collapse
Affiliation(s)
- Lance Fredrick Pahutan Bosch
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS–Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- *Correspondence: Katrin Kierdorf,
| |
Collapse
|
16
|
Gelibter S, Marostica G, Mandelli A, Siciliani S, Podini P, Finardi A, Furlan R. The impact of storage on extracellular vesicles: A systematic study. J Extracell Vesicles 2022; 11:e12162. [PMID: 35102719 PMCID: PMC8804350 DOI: 10.1002/jev2.12162] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 08/31/2021] [Accepted: 10/08/2021] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence suggests that storage has an impact on extracellular vesicles (EVs) properties. While -80°C storage is a widespread approach, some authors proposed improved storage strategies with conflicting results. Here, we designed a systematic study to assess the impact of -80°C storage and freeze-thaw cycles on EVs. We tested the differences among eight storage strategies and investigated the possible fusion phenomena occurring during storage. EVs were collected from human plasma and murine microglia culture by size exclusion chromatography and ultracentrifugation, respectively. The analysis included: concentration, size and zeta potential (tunable resistive pulse sensing), contaminant protein assessment; flow cytometry for the analysis of two single fluorescent-tagged EVs populations (GFP and mCherry), mixed before preservation. We found that -80°C storage reduces EVs concentration and sample purity in a time-dependent manner. Furthermore, it increases the particle size and size variability and modifies EVs zeta potential, with a shift of EVs in size-charge plots. None of the tested conditions prevented the observed effects. Freeze-thaw cycles lead to an EVs reduction after the first cycle and to a cycle-dependent increase in particle size. With flow cytometry, after storage, we observed a significant population of double-positive EVs (GFP+ -mCherry+ ). This observation may suggest the occurrence of fusion phenomena during storage. Our findings show a significant impact of storage on EVs samples in terms of particle loss, purity reduction and fusion phenomena leading to artefactual particles. Depending on downstream analyses and experimental settings, EVs should probably be processed from fresh, non-archival, samples in majority of cases.
Collapse
Affiliation(s)
- Stefano Gelibter
- Clinical Neuroimmunology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Giulia Marostica
- Clinical Neuroimmunology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Alessandra Mandelli
- Clinical Neuroimmunology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Stella Siciliani
- Neuroimmunology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Paola Podini
- Neuropathology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Annamaria Finardi
- Clinical Neuroimmunology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Roberto Furlan
- Clinical Neuroimmunology UnitInstitute of Experimental NeurologyDivision of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| |
Collapse
|
17
|
Yuan T, Orock A, Greenwood-VanMeerveld B. An enriched environment reduces chronic stress-induced visceral pain through modulating microglial activity in the central nucleus of the amygdala. Am J Physiol Gastrointest Liver Physiol 2022; 322:G223-G233. [PMID: 34877892 PMCID: PMC8793868 DOI: 10.1152/ajpgi.00307.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cognitive behavioral therapy (CBT) improves the quality of life for patients with brain-gut disorders; however, the underlying mechanisms of CBT remain to be explored. Previously, we showed that environmental enrichment (EE), an experimental paradigm that mirrors positive behavioral intervention, ameliorates chronic stress-induced visceral hypersensitivity in a rodent model via mechanisms involving altered activity in the central nucleus of amygdala (CeA). In the present study, we investigated whether microglia-mediated synaptic plasticity in the CeA is a potential mechanism underlying the protective effects of EE against stress-induced visceral hypersensitivity. We stereotaxically implanted corticosterone (CORT) micropellets onto the dorsal margin of the CeA shown previously to induce colonic hypersensitivity. Animals were housed in EE cages or standard cages for 14 days after CORT implantation. Visceral sensitivity was assessed via visceromotor behavioral response to colorectal distension. Microglial morphology, microglia-mediated synaptic engulfment, and the expression of synaptic pruning-related signals complement component 1q (C1q), complement component 3 (C3), and C3 receptor (C3R) were measured using immunofluorescence and RNAscope assay. We found that housing CORT implanted rats in EE cages for 14 days attenuated visceral hypersensitivity in both male and female rats as compared with control rats maintained in standard housing. EE reduced CORT-induced microglial remodeling and microglia-mediated synaptic pruning with reduced C1q and CR3, but not C3, expression. Our data suggest that exposure to EE is sufficient to ameliorate stress-induced visceral pain via reducing amygdala microglia-modulated neuronal plasticity.NEW & NOTEWORTHY Clinical studies show that cognitive behavioral therapy (CBT) is effective in ameliorating visceral pain in patient with irritable bowel syndrome (IBS), yet the underlying mechanisms remain unexplored. By using environmental enrichment (EE), an experimental paradigm that mirrors positive behavioral intervention, we demonstrated that microglia-mediated synaptic plasticity in the CeA explains, plays a role, at least in part, in the positive effects of EE to reduce visceral hypersensitivity.
Collapse
Affiliation(s)
- Tian Yuan
- 1Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Albert Orock
- 1Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Beverley Greenwood-VanMeerveld
- 1Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,2Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma
| |
Collapse
|
18
|
Han TH, Lee HW, Kang EA, Song MS, Lee SY, Ryu PD. Microglial activation induced by LPS mediates excitation of neurons in the hypothalamic paraventricular nucleus projecting to the rostral ventrolateral medulla. BMB Rep 2021. [PMID: 34814975 PMCID: PMC8728541 DOI: 10.5483/bmbrep.2021.54.12.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Microglia are known to be activated in the hypothalamic para-ventricular nucleus (PVN) of rats with cardiovascular diseases. However, the exact role of microglial activation in the plasticity of presympathetic PVN neurons associated with the modulation of sympathetic outflow remains poorly investigated. In this study, we analyzed the direct link between microglial activation and spontaneous firing rate along with the underlying synaptic mechanisms in PVN neurons projecting to the rostral ventrolateral medulla (RVLM). Systemic injection of LPS induced microglial activation in the PVN, increased the frequency of spontaneous firing activity of PVN-RVLM neurons, reduced GABAergic inputs into these neurons, and increased plasma NE levels and heart rate. Systemic minocycline injection blocked all the observed LPS-induced effects. Our results indicate that LPS increases the firing rate and decreases GABAergic transmission in PVN-RVLM neurons associated with sympathetic outflow and the alteration is largely attributed to the activation of microglia. Our findings provide some insights into the role of microglial activation in regulating the activity of PVN-RVLM neurons associated with modulation of sympathetic outflow in cardiovascular diseases.
Collapse
Affiliation(s)
- Tae Hee Han
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| | - Heow Won Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| | - Eun A Kang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| | - Min Seok Song
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| | - Pan Dong Ryu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
19
|
Wang XL, Li L. Microglia Regulate Neuronal Circuits in Homeostatic and High-Fat Diet-Induced Inflammatory Conditions. Front Cell Neurosci 2021; 15:722028. [PMID: 34720877 PMCID: PMC8549960 DOI: 10.3389/fncel.2021.722028] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are brain resident macrophages, which actively survey the surrounding microenvironment and promote tissue homeostasis under physiological conditions. During this process, microglia participate in synaptic remodeling, neurogenesis, elimination of unwanted neurons and cellular debris. The complex interplay between microglia and neurons drives the formation of functional neuronal connections and maintains an optimal neural network. However, activation of microglia induced by chronic inflammation increases synaptic phagocytosis and leads to neuronal impairment or death. Microglial dysfunction is implicated in almost all brain diseases and leads to long-lasting functional deficiency, such as hippocampus-related cognitive decline and hypothalamus-associated energy imbalance (i.e., obesity). High-fat diet (HFD) consumption triggers mediobasal hypothalamic microglial activation and inflammation. Moreover, HFD-induced inflammation results in cognitive deficits by triggering hippocampal microglial activation. Here, we have summarized the current knowledge of microglial characteristics and biological functions and also reviewed the molecular mechanism of microglia in shaping neural circuitries mainly related to cognition and energy balance in homeostatic and diet-induced inflammatory conditions.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianjian Li
- Department of Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.,Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
20
|
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: 63] [Impact Index Per Article: 15.8] [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
|
21
|
Neurotoxic Effects of Neonicotinoids on Mammals: What Is There beyond the Activation of Nicotinic Acetylcholine Receptors?-A Systematic Review. Int J Mol Sci 2021; 22:ijms22168413. [PMID: 34445117 PMCID: PMC8395098 DOI: 10.3390/ijms22168413] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Neonicotinoids are a class of insecticides that exert their effect through a specific action on neuronal nicotinic acetylcholine receptors (nAChRs). The success of these insecticides is due to this mechanism of action, since they act as potent agonists of insect nAChRs, presenting low affinity for vertebrate nAChRs, which reduces potential toxic risk and increases safety for non-target species. However, although neonicotinoids are considered safe, their presence in the environment could increase the risk of exposure and toxicity. On the other hand, although neonicotinoids have low affinity for mammalian nAChRs, the large quantity, variety, and ubiquity of these receptors, combined with its diversity of functions, raises the question of what effects these insecticides can produce in non-target species. In the present systematic review, we investigate the available evidence on the biochemical and behavioral effects of neonicotinoids on the mammalian nervous system. In general, exposure to neonicotinoids at an early age alters the correct neuronal development, with decreases in neurogenesis and alterations in migration, and induces neuroinflammation. In adulthood, neonicotinoids induce neurobehavioral toxicity, these effects being associated with their modulating action on nAChRs, with consequent neurochemical alterations. These alterations include decreased expression of nAChRs, modifications in acetylcholinesterase activity, and significant changes in the function of the nigrostriatal dopaminergic system. All these effects can lead to the activation of a series of intracellular signaling pathways that generate oxidative stress, neuroinflammation and, finally, neuronal death. Neonicotinoid-induced changes in nAChR function could be responsible for most of the effects observed in the different studies.
Collapse
|
22
|
da Silva Creão LS, Neto JBT, de Lima CM, dos Reis RR, de Sousa AA, dos Santos ZA, Diniz JAP, Diniz DG, Diniz CWP. Microglial Metamorphosis in Three Dimensions in Virus Limbic Encephalitis: An Unbiased Pictorial Representation Based on a Stereological Sampling Approach of Surveillant and Reactive Microglia. Brain Sci 2021; 11:brainsci11081009. [PMID: 34439628 PMCID: PMC8393838 DOI: 10.3390/brainsci11081009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 12/03/2022] Open
Abstract
Microglia influence pathological progression in neurological diseases, reacting to insults by expressing multiple morphofunctional phenotypes. However, the complete morphological spectrum of reactive microglia, as revealed by three-dimensional microscopic reconstruction, has not been detailed in virus limbic encephalitis. Here, using an anatomical series of brain sections, we expanded on an earlier Piry arbovirus encephalitis study to include CA1/CA2 and assessed the morphological response of homeostatic and reactive microglia at eight days post-infection. Hierarchical cluster and linear discriminant function analyses of multimodal morphometric features distinguished microglial morphology between infected animals and controls. For a broad representation of the spectrum of microglial morphology in each defined cluster, we chose representative cells of homeostatic and reactive microglia, using the sum of the distances of each cell in relation to all the others. Based on multivariate analysis, reactive microglia of infected animals showed more complex trees and thicker branches, covering a larger volume of tissue than in control animals. This approach offers a reliable representation of microglia dispersion in the Euclidean space, revealing the morphological kaleidoscope of surveillant and reactive microglia morphotypes. Because form precedes function in nature, our findings offer a starting point for research using integrative methods to understand microglia form and function.
Collapse
Affiliation(s)
- Leonardo Sávio da Silva Creão
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Brazil; (L.S.d.S.C.); (C.W.P.D.)
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - João Bento Torres Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
- Faculdade de Fisioterapia e Terapia Ocupacional, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Camila Mendes de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - Renata Rodrigues dos Reis
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - Aline Andrade de Sousa
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - Zaire Alves dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | | | - Daniel Guerreiro Diniz
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Brazil; (L.S.d.S.C.); (C.W.P.D.)
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66093-020, Brazil;
- Correspondence:
| | - Cristovam Wanderley Picanço Diniz
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Brazil; (L.S.d.S.C.); (C.W.P.D.)
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| |
Collapse
|
23
|
Tarudji AW, Gee CC, Romereim SM, Convertine AJ, Kievit FM. Antioxidant thioether core-crosslinked nanoparticles prevent the bilateral spread of secondary injury to protect spatial learning and memory in a controlled cortical impact mouse model of traumatic brain injury. Biomaterials 2021; 272:120766. [PMID: 33819812 DOI: 10.1016/j.biomaterials.2021.120766] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/04/2021] [Accepted: 03/14/2021] [Indexed: 01/19/2023]
Abstract
The secondary phase of traumatic brain injury (TBI) is partly caused by the release of excess reactive oxygen species (ROS) from the primary injury. However, there are currently no therapies that have been shown to reduce the secondary spread of injury beyond the primary insult. Nanoparticles offer the ability to rapidly accumulate and be retained in injured brain for improved target engagement. Here, we utilized systemically administered antioxidant thioether core-cross-linked nanoparticles (NP1) that scavenge and inactivate ROS to reduce this secondary spread of injury in a mild controlled cortical impact (CCI) mouse model of TBI. We found that NP1 treatment protected CCI mice from injury induced learning and memory deficits observed in the Morris water maze (MWM) test at 1-month post-CCI. This protection was likely a result of NP1-mediated reduction in oxidative stress in the ipsilateral hemisphere as determined by immunofluorescence imaging of markers of oxidative stress and the spread of neuroinflammation into the contralateral hippocampus as determined by immunofluorescence imaging of activated microglia and neuron-astrocyte-microglia triad formation. These data suggest NP1-mediated reduction in post-traumatic oxidative stress correlates with the reduction in the spread of injury to the contralateral hippocampus to protect spatial memory and learning in CCI mice. Therefore, these materials may offer an improved treatment strategy to reduce the secondary spread of TBI.
Collapse
Affiliation(s)
- Aria W Tarudji
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA
| | - Connor C Gee
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA
| | - Sarah M Romereim
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA
| | - Anthony J Convertine
- Department of Materials Science and Engineering, Missouri University of Science and Technology, 223 McNutt Hall, Rolla, MO, 65409, USA
| | - Forrest M Kievit
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA.
| |
Collapse
|
24
|
Age-dependent and region-specific alteration of parvalbumin neurons, perineuronal nets and microglia in the mouse prefrontal cortex and hippocampus following obesogenic diet consumption. Sci Rep 2021; 11:5593. [PMID: 33692414 PMCID: PMC7970944 DOI: 10.1038/s41598-021-85092-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/18/2021] [Indexed: 02/08/2023] Open
Abstract
Emergent evidence demonstrates that excessive consumption of high fat and high sugar (HFHS) diets has negative consequences on hippocampal and prefrontal cortex (PFC) function. Moreover, the delayed maturation of the PFC including the late development of parvalbumin-expressing (PV) interneurons and perineuronal nets (PNNs) may promote vulnerability to HFHS diet-induced nutritional stress. However, the young brain may have some resistance to diet-induced neuroinflammation. Thus, we examined the impact of a HFHS diet commencing either in adolescence or adulthood in male mice. PV interneurons, PNNs and microglia were assessed using immunohistochemistry. We observed greater numbers of PV neurons and PNNs in the hippocampus and the prelimbic and infralimbic PFC in adult mice in comparison to our younger cohort. Mice that consumed HFHS diet as adults had reduced numbers of hippocampal PV neurons and PNNs, which correlated with adiposity. However, we saw no effects of diet on PV and PNNs in the PFC. HFHS diet increased microgliosis in the adult cohort, and morphological changes to microglia were observed in the PFC and hippocampus of the adolescent cohort, with a shift to activated microglia phenotypes. Taken together, these findings demonstrate different regional and age-specific effects of obesogenic diets on PV neurons, PNNs and microglia.
Collapse
|
25
|
Leiguarda C, Villarreal A, Potilinski C, Pelissier T, Coronel MF, Bayo J, Ramos AJ, Montaner A, Villar MJ, Constandil L, Brumovsky PR. Intrathecal Administration of an Anti-nociceptive Non-CpG Oligodeoxynucleotide Reduces Glial Activation and Central Sensitization. J Neuroimmune Pharmacol 2021; 16:818-834. [PMID: 33502706 DOI: 10.1007/s11481-021-09983-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/12/2021] [Indexed: 01/18/2023]
Abstract
Inflammatory pain associates with spinal glial activation and central sensitization. Systemic administration of IMT504, a non-CpG oligodeoxynucleotide originally designed as an immunomodulator, exerts remarkable anti-allodynic effects in rats with complete Freund´s adjuvant (CFA)-induced hindpaw inflammation. However, the anti-nociceptive mechanisms of IMT504 remain unknown. Here we evaluated whether IMT504 blocks inflammatory pain-like behavior by modulation of spinal glia and central sensitization. The study was performed in Sprague Dawley rats with intraplantar CFA, and a single lumbosacral intrathecal (i.t.) administration of IMT504 or vehicle was chosen to address if changes in glial activation and spinal sensitization relate to the pain-like behavior reducing effects of the ODN. Naïve rats were also included. Von Frey and Randall-Selitto tests, respectively, exposed significant reductions in allodynia and mechanical hypersensitivity, lasting at least 24 h after i.t. IMT504. Analysis of electromyographic responses to electrical stimulation of C fibers showed progressive reductions in wind-up responses. Accordingly, IMT504 significantly downregulated spinal glial activation, as shown by reductions in the protein expression of glial fibrillary acidic protein, CD11b/c, Toll-like receptor 4 (TLR4) and the phosphorylated p65 subunit of NFκB, evaluated by immunohistochemistry and western blot. In vitro experiments using early post-natal cortical glial cultures provided further support to in vivo data and demonstrated IMT504 internalization into microglia and astrocytes. Altogether, our study provides new evidence on the central mechanisms of anti-nociception by IMT504 upon intrathecal application, and further supports its value as a novel anti-inflammatory ODN with actions upon glial cells and the TLR4/NFκB pathway. Intrathecal administration of the non-CpG ODN IMT504 fully blocks CFA-induced mechanical allodynia and hypersensitivity, in association with reduced spinal sensitization. Administration of the ODN also results in downregulated gliosis and reduced TLR4-NF-κB pathway activation. IMT504 uptake into astrocytes and microglia support the concept of direct modulation of CFA-induced glial activation.
Collapse
Affiliation(s)
- C Leiguarda
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Av. Juan D. Perón 1500, Pilar, Buenos Aires, B1629AHJ, Argentina
| | - A Villarreal
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, 1121, Argentina
| | - C Potilinski
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Av. Juan D. Perón 1500, Pilar, Buenos Aires, B1629AHJ, Argentina
| | - T Pelissier
- Laboratorio de Neurobiología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, 8320000, Chile
| | - M F Coronel
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Av. Juan D. Perón 1500, Pilar, Buenos Aires, B1629AHJ, Argentina
| | - J Bayo
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Av. Juan D. Perón 1500, Pilar, Buenos Aires, B1629AHJ, Argentina
| | - A J Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, 1121, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, 1121, Argentina
| | - A Montaner
- Instituto de Ciencia y Tecnología "Dr. César Milstein", CONICET, Fundación Pablo Cassará, Buenos Aires, C1440FFX, Argentina
| | - M J Villar
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Av. Juan D. Perón 1500, Pilar, Buenos Aires, B1629AHJ, Argentina
| | - L Constandil
- Laboratorio de Neurobiología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, 8320000, Chile
| | - Pablo R Brumovsky
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral-CONICET, Av. Juan D. Perón 1500, Pilar, Buenos Aires, B1629AHJ, Argentina.
| |
Collapse
|
26
|
Dukay B, Walter FR, Vigh JP, Barabási B, Hajdu P, Balassa T, Migh E, Kincses A, Hoyk Z, Szögi T, Borbély E, Csoboz B, Horváth P, Fülöp L, Penke B, Vígh L, Deli MA, Sántha M, Tóth ME. Neuroinflammatory processes are augmented in mice overexpressing human heat-shock protein B1 following ethanol-induced brain injury. J Neuroinflammation 2021; 18:22. [PMID: 33423680 PMCID: PMC7798334 DOI: 10.1186/s12974-020-02070-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022] Open
Abstract
Background Heat-shock protein B1 (HSPB1) is among the most well-known and versatile member of the evolutionarily conserved family of small heat-shock proteins. It has been implicated to serve a neuroprotective role against various neurological disorders via its modulatory activity on inflammation, yet its exact role in neuroinflammation is poorly understood. In order to shed light on the exact mechanism of inflammation modulation by HSPB1, we investigated the effect of HSPB1 on neuroinflammatory processes in an in vivo and in vitro model of acute brain injury. Methods In this study, we used a transgenic mouse strain overexpressing the human HSPB1 protein. In the in vivo experiments, 7-day-old transgenic and wild-type mice were treated with ethanol. Apoptotic cells were detected using TUNEL assay. The mRNA and protein levels of cytokines and glial cell markers were examined using RT-PCR and immunohistochemistry in the brain. We also established primary neuronal, astrocyte, and microglial cultures which were subjected to cytokine and ethanol treatments. TNFα and hHSPB1 levels were measured from the supernates by ELISA, and intracellular hHSPB1 expression was analyzed using fluorescent immunohistochemistry. Results Following ethanol treatment, the brains of hHSPB1-overexpressing mice showed a significantly higher mRNA level of pro-inflammatory cytokines (Tnf, Il1b), microglia (Cd68, Arg1), and astrocyte (Gfap) markers compared to wild-type brains. Microglial activation, and 1 week later, reactive astrogliosis was higher in certain brain areas of ethanol-treated transgenic mice compared to those of wild-types. Despite the remarkably high expression of pro-apoptotic Tnf, hHSPB1-overexpressing mice did not exhibit higher level of apoptosis. Our data suggest that intracellular hHSPB1, showing the highest level in primary astrocytes, was responsible for the inflammation-regulating effects. Microglia cells were the main source of TNFα in our model. Microglia isolated from hHSPB1-overexpressing mice showed a significantly higher release of TNFα compared to wild-type cells under inflammatory conditions. Conclusions Our work provides novel in vivo evidence that hHSPB1 overexpression has a regulating effect on acute neuroinflammation by intensifying the expression of pro-inflammatory cytokines and enhancing glial cell activation, but not increasing neuronal apoptosis. These results suggest that hHSPB1 may play a complex role in the modulation of the ethanol-induced neuroinflammatory response. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02070-2.
Collapse
Affiliation(s)
- Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary. .,Doctoral School in Biology, University of Szeged, Szeged, Hungary.
| | - Fruzsina R Walter
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Judit P Vigh
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Beáta Barabási
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Doctoral School in Theoretical Medicine, University of Szeged, Szeged, Hungary
| | - Petra Hajdu
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary
| | - Tamás Balassa
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary.,Doctoral School of Informatics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Ede Migh
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Titanilla Szögi
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Emőke Borbély
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Bálint Csoboz
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary.,Institute of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Péter Horváth
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Lívia Fülöp
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Botond Penke
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, Szeged, H-6726, Hungary.
| |
Collapse
|
27
|
Primary cilia safeguard cortical neurons in neonatal mouse forebrain from environmental stress-induced dendritic degeneration. Proc Natl Acad Sci U S A 2020; 118:2012482118. [PMID: 33443207 DOI: 10.1073/pnas.2012482118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The developing brain is under the risk of exposure to a multitude of environmental stressors. While perinatal exposure to excessive levels of environmental stress is responsible for a wide spectrum of neurological and psychiatric conditions, the developing brain is equipped with intrinsic cell protection, the mechanisms of which remain unknown. Here we show, using neonatal mouse as a model system, that primary cilia, hair-like protrusions from the neuronal cell body, play an essential role in protecting immature neurons from the negative impacts of exposure to environmental stress. More specifically, we found that primary cilia prevent the degeneration of dendritic arbors upon exposure to alcohol and ketamine, two major cell stressors, by activating cilia-localized insulin-like growth factor 1 receptor and downstream Akt signaling. We also found that activation of this pathway inhibits Caspase-3 activation and caspase-mediated cleavage/fragmentation of cytoskeletal proteins in stress-exposed neurons. These results indicate that primary cilia play an integral role in mitigating adverse impacts of environmental stressors such as drugs on perinatal brain development.
Collapse
|
28
|
Ward H, West SJ. Microglia: sculptors of neuropathic pain? ROYAL SOCIETY OPEN SCIENCE 2020; 7:200260. [PMID: 32742693 PMCID: PMC7353970 DOI: 10.1098/rsos.200260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/01/2020] [Indexed: 05/02/2023]
Abstract
Neuropathic pain presents a huge societal and individual burden. The limited efficacy of current analgesics, diagnostic markers and clinical trial outcome measures arises from an incomplete understanding of the underlying mechanisms. A large and growing body of evidence has established the important role of microglia in the onset and possible maintenance of neuropathic pain, and these cells may represent an important target for future therapy. Microglial research has further revealed their important role in structural remodelling of the nervous system. In this review, we aim to explore the evidence for microglia in sculpting nervous system structure and function, as well as their important role in neuropathic pain, and finally integrate these studies to synthesize a new model for microglia in somatosensory circuit remodelling, composed of six key and inter-related mechanisms. Summarizing the mechanisms through which microglia modulate nervous system structure and function helps to frame a better understanding of neuropathic pain, and provide a clear roadmap for future research.
Collapse
Affiliation(s)
- Harry Ward
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Steven J. West
- Sainsbury Wellcome Centre, University College London, 25 Howland St, London WC1E 6BT, UK
- Author for correspondence: Steven J. West e-mail:
| |
Collapse
|
29
|
Fernández-Arjona MDM, Grondona JM, Fernández-Llebrez P, López-Ávalos MD. Microglial Morphometric Parameters Correlate With the Expression Level of IL-1β, and Allow Identifying Different Activated Morphotypes. Front Cell Neurosci 2019; 13:472. [PMID: 31708746 PMCID: PMC6824358 DOI: 10.3389/fncel.2019.00472] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022] Open
Abstract
Microglia are the resident macrophages in the brain. Traditionally, two forms of microglia have been described: one considered as a resting/surveillant state in which cells have a highly branched morphology, and another considered as an activated state in which they acquire a de-ramified or amoeboid form. However, many studies describe intermediate microglial morphologies which emerge during pathological processes. Since microglial form and function are closely related, it is of interest to correlate microglial morphology with the extent of its activation. To address this issue, we used a rat model of neuroinflammation consisting in a single injection of the enzyme neuraminidase (NA) within the lateral ventricle. Sections from NA-injected animals were co-immunolabeled with the microglial marker IBA1 and the cytokine IL-1β, which highlight features of the cell’s shape and inflammatory activation, respectively. Activated (IL-1β positive) microglial cells were sampled from the dorsal hypothalamus nearby the third ventricle. Images of single microglial cells were processed in two different ways to obtain (1) an accurate measure of the level of expression of IL-1β (indicating the degree of activation), and (2) a set of 15 morphological parameters to quantitatively and objectively describe the cell’s shape. A simple regression analysis revealed a dependence of most of the morphometric parameters on IL-1β expression, demonstrating that the morphology of microglial cells changes progressively with the degree of activation. Moreover, a hierarchical cluster analysis pointed out four different morphotypes of activated microglia, which are characterized not only by morphological parameters values, but also by specific IL-1β expression levels. Thus, these results demonstrate in an objective manner that the activation of microglial cells is a gradual process, and correlates with their morphological change. Even so, it is still possible to categorize activated cells according to their morphometric parameters, each category presenting a different activation degree. The physiological relevance of those activated morphotypes is an issue worth to be assessed in the future.
Collapse
Affiliation(s)
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Pedro Fernández-Llebrez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| |
Collapse
|
30
|
Kim S, Kim YE, Hong S, Kim KT, Sung DK, Lee Y, Park WS, Chang YS, Song MR. Reactive microglia and astrocytes in neonatal intraventricular hemorrhage model are blocked by mesenchymal stem cells. Glia 2019; 68:178-192. [PMID: 31441125 DOI: 10.1002/glia.23712] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/08/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022]
Abstract
Severe intraventricular hemorrhage (IVH) in premature infants triggers reactive gliosis, causing acute neuronal death and glial scar formation. Transplantation of mesenchymal stem cells (MSCs) has often showed improved CNS recovery in an IVH model, but whether this response is related to reactive glial cells is still unclear. Herein, we suggest that MSCs impede the response of reactive microglia rather than astrocytes, thereby blocking neuronal damage. Astrocytes alone showed mild reactiveness under hemorrhagic conditions mimicked by thrombin treatment, and this was not blocked by MSC-conditioned medium (MSC-CM) in vitro. In contrast, thrombin-induced microglial activation and release of proinflammatory cytokines were inhibited by MSC-CM. Interestingly, astrocytes showed greater reactive response when co-cultured with microglia, and this was abolished in the presence of MSC-CM. Gene expression profiles in microglia revealed that transcript levels of genes for immune response and proinflammatory cytokines were altered by thrombin treatment. This result coincided with the robust phosphorylation of STAT1 and p38 MAPK, which might be responsible for the production and release of proinflammatory cytokines. Furthermore, application of MSC-CM diminished thrombin-mediated phosphorylation of STAT1 and p38 MAPK, supporting the acute anti-inflammatory role of MSCs under hemorrhagic conditions. In line with this, activation of microglia and consequent cytokine release were impaired in Stat1-null mice. However, reactive response in Stat1-deficient astrocytes was maintained. Taken together, our results demonstrate that MSCs mainly block the activation of microglia involving STAT1-mediated cytokine release and subsequent reduction of reactive astrocytes.
Collapse
Affiliation(s)
- Seojeong Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Young Eun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Sujeong Hong
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Kyung-Tai Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yunjeong Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
31
|
Riazifar M, Mohammadi MR, Pone EJ, Yeri A, Lässer C, Segaliny AI, McIntyre LL, Shelke GV, Hutchins E, Hamamoto A, Calle EN, Crescitelli R, Liao W, Pham V, Yin Y, Jayaraman J, Lakey JRT, Walsh CM, Van Keuren-Jensen K, Lotvall J, Zhao W. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS NANO 2019; 13:6670-6688. [PMID: 31117376 PMCID: PMC6880946 DOI: 10.1021/acsnano.9b01004] [Citation(s) in RCA: 392] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
To dissect therapeutic mechanisms of transplanted stem cells and develop exosome-based nanotherapeutics in treating autoimmune and neurodegenerative diseases, we assessed the effect of exosomes secreted from human mesenchymal stem cells (MSCs) in treating multiple sclerosis using an experimental autoimmune encephalomyelitis (EAE) mouse model. We found that intravenous administration of exosomes produced by MSCs stimulated by IFNγ (IFNγ-Exo) (i) reduced the mean clinical score of EAE mice compared to PBS control, (ii) reduced demyelination, (iii) decreased neuroinflammation, and (iv) upregulated the number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords of EAE mice. Co-culture of IFNγ-Exo with activated peripheral blood mononuclear cells (PBMCs) cells in vitro reduced PBMC proliferation and levels of pro-inflammatory Th1 and Th17 cytokines including IL-6, IL-12p70, IL-17AF, and IL-22 yet increased levels of immunosuppressive cytokine indoleamine 2,3-dioxygenase. IFNγ-Exo could also induce Tregs in vitro in a murine splenocyte culture, likely mediated by a third-party accessory cell type. Further, IFNγ-Exo characterization by deep RNA sequencing suggested that IFNγ-Exo contains anti-inflammatory RNAs, where their inactivation partially hindered the exosomes potential to induce Tregs. Furthermore, we found that IFNγ-Exo harbors multiple anti-inflammatory and neuroprotective proteins. These results not only shed light on stem cell therapeutic mechanisms but also provide evidence that MSC-derived exosomes can potentially serve as cell-free therapies in creating a tolerogenic immune response to treat autoimmune and central nervous system disorders.
Collapse
Affiliation(s)
- Milad Riazifar
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - M. Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Egest J. Pone
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Department of Physiology and Biophysics, Vaccine Research and Development Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ashish Yeri
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Cecilia Lässer
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Aude I. Segaliny
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Laura L. McIntyre
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Ganesh Vilas Shelke
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
- Department of Surgery, Institute of Clinical Sciences, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg 41345, Sweden
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Ashley Hamamoto
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Erika N. Calle
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Rossella Crescitelli
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Wenbin Liao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Victor Pham
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Yanan Yin
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jayapriya Jayaraman
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Jonathan R. T. Lakey
- Department of Surgery, University of California, Irvine, Orange, California 92868, United States
| | - Craig M. Walsh
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, California 92697, United States
| | - Kendall Van Keuren-Jensen
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, United States
| | - Jan Lotvall
- Krefting Research Center, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 40530, Sweden
| | - Weian Zhao
- Department of Pharmaceutical Sciences, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, Department of Biomedical Engineering, and Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Corresponding Author:
| |
Collapse
|
32
|
Seasonal adaptations of the hypothalamo-neurohypophyseal system of the dromedary camel. PLoS One 2019; 14:e0216679. [PMID: 31211771 PMCID: PMC6581255 DOI: 10.1371/journal.pone.0216679] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
The “ship” of the Arabian and North African deserts, the one-humped dromedary camel (Camelus dromedarius) has a remarkable capacity to survive in conditions of extreme heat without needing to drink water. One of the ways that this is achieved is through the actions of the antidiuretic hormone arginine vasopressin (AVP), which is made in a specialised part of the brain called the hypothalamo-neurohypophyseal system (HNS), but exerts its effects at the level of the kidney to provoke water conservation. Interestingly, our electron microscopy studies have shown that the ultrastructure of the dromedary HNS changes according to season, suggesting that in the arid conditions of summer the HNS is in an activated state, in preparation for the likely prospect of water deprivation. Based on our dromedary genome sequence, we have carried out an RNAseq analysis of the dromedary HNS in summer and winter. Amongst the 171 transcripts found to be significantly differentially regulated (>2 fold change, p value <0.05) there is a significant over-representation of neuropeptide encoding genes, including that encoding AVP, the expression of which appeared to increase in summer. Identification of neuropeptides in the HNS and analysis of neuropeptide profiles in extracts from individual camels using mass spectrometry indicates that overall AVP peptide levels decreased in the HNS during summer compared to winter, perhaps due to increased release during periods of dehydration in the dry season.
Collapse
|
33
|
Gyengesi E, Rangel A, Ullah F, Liang H, Niedermayer G, Asgarov R, Venigalla M, Gunawardena D, Karl T, Münch G. Chronic Microglial Activation in the GFAP-IL6 Mouse Contributes to Age-Dependent Cerebellar Volume Loss and Impairment in Motor Function. Front Neurosci 2019; 13:303. [PMID: 31001075 PMCID: PMC6456818 DOI: 10.3389/fnins.2019.00303] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/18/2019] [Indexed: 11/15/2022] Open
Abstract
Chronic microglial activation is a prominent feature of many chronic neurodegenerative diseases, including Parkinson’s and Alzheimer’s disease. To investigate the effects of chronic microglial activation on cerebellar structure and motor function throughout the lifespan, the transgenic GFAP-IL6 mouse model was used. The aim of the study was to examine inflammatory markers and neuronal degeneration while simultaneously characterizing the motor performance of GFAP-IL6 mice at 3, 6, 14, and 24 months of age in comparison to WT (C57BL/6) mice. In respect to markers of neuroinflammation in the cerebellum, increased numbers of Iba1+ microglia were observed as early as at 3 months of age. In addition, TNF-α levels proved to be significantly higher in the GFAP-IL6 compared to WT mice at all time points. A difference in cerebellar volume between the GFAP-IL6 and WT mice was observed later in life, starting at 6 months and increasing to a loss of about 50% in aged (24 months old) GFAP-IL6 mice. Synaptic deficits were also assessed by using pre- (synaptophysin) and post-synaptic (PSD95) markers. While synaptophysin levels remained unchanged, PSD95 levels decreased in the aging GFAP-IL6 mice compared to their WT littermates from 14 months onward. To assess the effect of microglia activation and neurodegeneration on behavior, a variety of motor function tests, semi-quantitative cerebellar ataxia score, accelerod, beam walking, and open field tests were performed. An age-dependent difference between the genotypes was observed in many of the motor function tests. For example, reduced performance on the accelerod and higher ataxia scores were observed at 6 months of age, followed by the beam walking test showing differences at 14 months of age. In summary, this study constitutes a comprehensive, age-dependent examination of inflammatory, synaptic and neurodegenerative changes in the brains of GFAP-IL6 mice leading to a deterioration in motor performance. The results also indicate that early chronic microglia activation in the GFAP-IL6 mouse leads to observable cerebellar volume loss and motor deficits later in life.
Collapse
Affiliation(s)
- Erika Gyengesi
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Alejandra Rangel
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Faheem Ullah
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Huazheng Liang
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,Department of Neurology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Garry Niedermayer
- School of Science and Health, Western Sydney University, Penrith, NSW, Australia
| | - Rustam Asgarov
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Madhuri Venigalla
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Dhanushka Gunawardena
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Tim Karl
- Behavioral Neuroscience Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Gerald Münch
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
34
|
Moser VA, Christensen A, Liu J, Zhou A, Yagi S, Beam CR, Galea L, Pike CJ. Effects of aging, high-fat diet, and testosterone treatment on neural and metabolic outcomes in male brown Norway rats. Neurobiol Aging 2019; 73:145-160. [PMID: 30359877 PMCID: PMC6252085 DOI: 10.1016/j.neurobiolaging.2018.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 07/01/2018] [Accepted: 09/13/2018] [Indexed: 12/23/2022]
Abstract
Risk for Alzheimer's disease (AD) is affected by multiple factors, including aging, obesity, and low testosterone. We previously showed that obesity and low testosterone independently and interactively exacerbate AD-related outcomes in young adult rodents. The goals of the present study are two-fold: to examine whether the effects of an obesogenic diet differ with increasing age and to determine if testosterone treatment in middle-aged and aged animals mitigates negative effects of the diet. Male brown Norway rats were maintained on control or high-fat diets for 12 weeks beginning in young adulthood, middle age, or advanced age. Separate cohorts of middle-aged and aged animals were treated with testosterone during dietary manipulations. Endpoints included metabolic indices, inflammation, cognitive performance, and neural health outcomes. Aging was associated with poorer outcomes that were generally exacerbated by high-fat diet, especially at middle age. Testosterone treatment was largely without benefit, exerting only subtle effects on a select number of measures. Understanding how the deleterious effects of obesity are affected by advancing age and the ability of protective strategies such as testosterone to reduce these effects may provide significant insight into both the development and prevention of age-related cognitive decline and AD.
Collapse
Affiliation(s)
- V Alexandra Moser
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Amy Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jiahui Liu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amanda Zhou
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Shunya Yagi
- Djavad Mowafaghian Centre for Brain Health, Department of Psychology, University of British Columbia, Vancouver BC, Canada
| | - Christopher R Beam
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Liisa Galea
- Djavad Mowafaghian Centre for Brain Health, Department of Psychology, University of British Columbia, Vancouver BC, Canada
| | - Christian J Pike
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Moser VA, Uchoa MF, Pike CJ. TLR4 inhibitor TAK-242 attenuates the adverse neural effects of diet-induced obesity. J Neuroinflammation 2018; 15:306. [PMID: 30396359 PMCID: PMC6217784 DOI: 10.1186/s12974-018-1340-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/22/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Obesity exerts negative effects on brain health, including decreased neurogenesis, impaired learning and memory, and increased risk for Alzheimer's disease and related dementias. Because obesity promotes glial activation, chronic neuroinflammation, and neural injury, microglia are implicated in the deleterious effects of obesity. One pathway that is particularly important in mediating the effects of obesity in peripheral tissues is toll-like receptor 4 (TLR4) signaling. The potential contribution of TLR4 pathways in mediating adverse neural outcomes of obesity has not been well addressed. To investigate this possibility, we examined how pharmacological inhibition of TLR4 affects the peripheral and neural outcomes of diet-induced obesity. METHODS Male C57BL6/J mice were maintained on either a control or high-fat diet for 12 weeks in the presence or absence of the specific TLR4 signaling inhibitor TAK-242. Outcomes examined included metabolic indices, a range of behavioral assessments, microglial activation, systemic and neuroinflammation, and neural health endpoints. RESULTS Peripherally, TAK-242 treatment was associated with partial inhibition of inflammation in the adipose tissue but exerted no significant effects on body weight, adiposity, and a range of metabolic measures. In the brain, obese mice treated with TAK-242 exhibited a significant reduction in microglial activation, improved levels of neurogenesis, and inhibition of Alzheimer-related amyloidogenic pathways. High-fat diet and TAK-242 were associated with only very modest effects on a range of behavioral measures. CONCLUSIONS These results demonstrate a significant protective effect of TLR4 inhibition on neural consequences of obesity, findings that further define the role of microglia in obesity-mediated outcomes and identify a strategy for improving brain health in obese individuals.
Collapse
Affiliation(s)
- V. Alexandra Moser
- 0000 0001 2156 6853grid.42505.36Neuroscience Graduate Program, University of Southern California, 3641 Watt Way, HNB 120, Los Angeles, CA 90089 USA
| | - Mariana F. Uchoa
- 0000 0001 2156 6853grid.42505.36Neuroscience Graduate Program, University of Southern California, 3641 Watt Way, HNB 120, Los Angeles, CA 90089 USA
| | - Christian J. Pike
- 0000 0001 2156 6853grid.42505.36Neuroscience Graduate Program, University of Southern California, 3641 Watt Way, HNB 120, Los Angeles, CA 90089 USA ,0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| |
Collapse
|
36
|
Ueda Y, Bando Y, Misumi S, Ogawa S, Ishida A, Jung CG, Shimizu T, Hida H. Alterations of Both Dendrite Morphology and Weaker Electrical Responsiveness in the Cortex of Hip Area Occur Before Rearrangement of the Motor Map in Neonatal White Matter Injury Model. Front Neurol 2018; 9:443. [PMID: 29971036 PMCID: PMC6018077 DOI: 10.3389/fneur.2018.00443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022] Open
Abstract
Hypoxia-ischemia (H-I) in rats at postnatal day 3 causes disorganization of oligodendrocyte development in layers II/III of the sensorimotor cortex without apparent neuronal loss, and shows mild hindlimb dysfunction with imbalanced motor coordination. However, the mechanisms by which mild motor dysfunction is induced without loss of cortical neurons are currently unclear. To reveal the mechanisms underlying mild motor dysfunction in neonatal H-I model, electrical responsiveness and dendrite morphology in the sensorimotor cortex were investigated at 10 weeks of age. Responses to intracortical microstimulation (ICMS) revealed that the cortical motor map was significantly changed in this model. The cortical area related to hip joint movement was reduced, and the area related to trunk movement was increased. Sholl analysis in Golgi staining revealed that layer I–III neurons on the H-I side had more dendrite branches compared with the contralateral side. To investigate whether changes in the motor map and morphology appeared at earlier stages, ICMS and Sholl analysis were also performed at 5 weeks of age. The minimal ICMS current to evoke twitches of the hip area was higher on the H-I side, while the motor map was unchanged. Golgi staining revealed more dendrite branches in layer I–III neurons on the H-I side. These results revealed that alterations of both dendrite morphology and ICMS threshold of the hip area occurred before the rearrangement of the motor map in the neonatal H-I model. They also suggest that altered dendritic morphology and altered ICMS responsiveness may be related to mild motor dysfunction in this model.
Collapse
Affiliation(s)
- Yoshitomo Ueda
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshio Bando
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Japan
| | - Sachiyo Misumi
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shino Ogawa
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimasa Ishida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Cha-Gyun Jung
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takeshi Shimizu
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
37
|
Li G, Yamasaki R, Fang M, Masaki K, Ochi H, Matsushita T, Kira JI. Novel disease-modifying anti-rheumatic drug iguratimod suppresses chronic experimental autoimmune encephalomyelitis by down-regulating activation of macrophages/microglia through an NF-κB pathway. Sci Rep 2018; 8:1933. [PMID: 29386552 PMCID: PMC5792543 DOI: 10.1038/s41598-018-20390-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 01/17/2018] [Indexed: 12/28/2022] Open
Abstract
We aimed to elucidate the effects of iguratimod, a widely used anti-rheumatic drug with no severe side effects, on chronic experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Iguratimod was orally administered to mice immunised with myelin oligodendrocyte glycoprotein peptide 35–55. Preventive administration of iguratimod from the time of immunisation was found to markedly reduce the clinical severity of acute and chronic EAE. Pathologically, iguratimod treatment significantly reduced demyelination and infiltration of CD3+ T, F4/80+, and CD169+ cells into the spinal cord, and suppressed macrophage/microglia activation in the parenchyma at the acute and chronic stages compared with vehicle treatment. Therapeutic administration of iguratimod after the onset of clinical symptoms significantly ameliorated the clinical severity of chronic EAE and reduced demyelination, T helper (Th)1/Th17 cell infiltration, macrophage/microglia activation, and nuclear factor (NF)-κB p65 and cyclooxygenase-2 expression in the spinal cord. In vitro, iguratimod treatment inhibited nuclear translocation of NF-κB p65 and down-regulated pro-inflammatory responses in macrophages and microglia. Our results suggest that iguratimod ameliorates acute and chronic EAE by suppressing inflammatory cell infiltration and immune cell activation, partly through inhibition of NF-κB p65, supporting the therapeutic potential of this drug for not only acute, but also chronic MS.
Collapse
Affiliation(s)
- Guangrui Li
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Mei Fang
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hirofumi Ochi
- Department of Geriatric Medicine and Neurology, Ehime University Graduate School of Medicine, Matsuyama, 791-0295, Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
38
|
Nguyen T, Mao Y, Sutherland T, Gorrie CA. Neural progenitor cells but not astrocytes respond distally to thoracic spinal cord injury in rat models. Neural Regen Res 2017; 12:1885-1894. [PMID: 29239336 PMCID: PMC5745844 DOI: 10.4103/1673-5374.219051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a detrimental condition that causes loss of sensory and motor function in an individual. Many complex secondary injury cascades occur after SCI and they offer great potential for therapeutic targeting. In this study, we investigated the response of endogenous neural progenitor cells, astrocytes, and microglia to a localized thoracic SCI throughout the neuroaxis. Twenty-five adult female Sprague-Dawley rats underwent mild-contusion thoracic SCI (n = 9), sham surgery (n = 8), or no surgery (n = 8). Spinal cord and brain tissues were fixed and cut at six regions of the neuroaxis. Immunohistochemistry showed increased reactivity of neural progenitor cell marker nestin in the central canal at all levels of the spinal cord. Increased reactivity of astrocyte-specific marker glial fibrillary acidic protein was found only at the lesion epicenter. The number of activated microglia was significantly increased at the lesion site, and activated microglia extended to the lumbar enlargement. Phagocytic microglia and macrophages were significantly increased only at the lesion site. There were no changes in nestin, glial fibrillary acidic protein, microglia and macrophage response in the third ventricle of rats subjected to mild-contusion thoracic SCI compared to the sham surgery or no surgery. These findings indicate that neural progenitor cells, astrocytes and microglia respond differently to a localized SCI, presumably due to differences in inflammatory signaling. These different cellular responses may have implications in the way that neural progenitor cells can be manipulated for neuroregeneration after SCI. This needs to be further investigated.
Collapse
Affiliation(s)
- Tara Nguyen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia
| | - Yilin Mao
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia
| | - Theresa Sutherland
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia
| | - Catherine Anne Gorrie
- School of Life Sciences, Faculty of Science, University of Technology Sydney, New South Wales, Australia
| |
Collapse
|
39
|
The phenotype of the RABV glycoprotein determines cellular and global virus load in the brain and is decisive for the pace of the disease. Virology 2017; 511:82-94. [DOI: 10.1016/j.virol.2017.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/21/2017] [Accepted: 08/14/2017] [Indexed: 11/18/2022]
|
40
|
Evaluation of the neonatal streptozotocin model of diabetes in rats: Evidence for a model of neuropathic pain. Pharmacol Rep 2017; 70:294-303. [PMID: 29477037 DOI: 10.1016/j.pharep.2017.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/01/2017] [Accepted: 09/12/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate the participation of satellite glial cells (SGC), microglia and astrocytes in a model of streptozotocin-induced diabetes initiated in neonatal rats (nSTZ) and to determine the pharmacological profile for pain relief. METHODS nSTZ was used to induce experimental diabetes. Von Frey filaments were used to assess tactile allodynia. Drugs were given by systemic administration. Western blotting and immunohistochemistry were used to determine protein expression and cellular localization. RESULTS nSTZ produced mild hyperglycemia, weight loss, glucose intolerance, and reduction of nerve conduction velocity of C fibers. Moreover, nSTZ enhanced activating transcription factor 3 (ATF3) immunoreactivity in dorsal root ganglia (DRG) and sciatic nerve of adult rats. ATF3 was found in SGC (GFAP+ cells) surrounding DRG at week 16. Late changes in ATF3 immunoreactivity in DRG correlated with up-regulation of ATF3 and GFAP protein expression. nSTZ increased GFAP and OX-42 immunoreactivity and percentage of hypertrophied and ameboid microglia in the spinal dorsal horn. These changes correlated with the presence of mechanical hypersensitivity (tactile allodynia). Administration of gabapentin (30-100mg/kg, po) and metformin (200mg/kg/day, po for 2 weeks) alleviated tactile allodynia, whereas morphine (1-3mg/kg, ip) had a modest effect. CONCLUSIONS Results suggest that nSTZ leads to activation of SGC, microglia and astrocytes in DRG and spinal cord. Pharmacological profile in the nSTZ model resembles diabetic neuropathic pain in humans. Our findings support the conclusion that the nSTZ rat model has utility for the study of a long-lasting diabetic neuropathic pain.
Collapse
|
41
|
Fernández-Arjona MDM, Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. Microglia Morphological Categorization in a Rat Model of Neuroinflammation by Hierarchical Cluster and Principal Components Analysis. Front Cell Neurosci 2017; 11:235. [PMID: 28848398 PMCID: PMC5550745 DOI: 10.3389/fncel.2017.00235] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022] Open
Abstract
It is known that microglia morphology and function are closely related, but only few studies have objectively described different morphological subtypes. To address this issue, morphological parameters of microglial cells were analyzed in a rat model of aseptic neuroinflammation. After the injection of a single dose of the enzyme neuraminidase (NA) within the lateral ventricle (LV) an acute inflammatory process occurs. Sections from NA-injected animals and sham controls were immunolabeled with the microglial marker IBA1, which highlights ramifications and features of the cell shape. Using images obtained by section scanning, individual microglial cells were sampled from various regions (septofimbrial nucleus, hippocampus and hypothalamus) at different times post-injection (2, 4 and 12 h). Each cell yielded a set of 15 morphological parameters by means of image analysis software. Five initial parameters (including fractal measures) were statistically different in cells from NA-injected rats (most of them IL-1β positive, i.e., M1-state) compared to those from control animals (none of them IL-1β positive, i.e., surveillant state). However, additional multimodal parameters were revealed more suitable for hierarchical cluster analysis (HCA). This method pointed out the classification of microglia population in four clusters. Furthermore, a linear discriminant analysis (LDA) suggested three specific parameters to objectively classify any microglia by a decision tree. In addition, a principal components analysis (PCA) revealed two extra valuable variables that allowed to further classifying microglia in a total of eight sub-clusters or types. The spatio-temporal distribution of these different morphotypes in our rat inflammation model allowed to relate specific morphotypes with microglial activation status and brain location. An objective method for microglia classification based on morphological parameters is proposed. Main pointsMicroglia undergo a quantifiable morphological change upon neuraminidase induced inflammation. Hierarchical cluster and principal components analysis allow morphological classification of microglia. Brain location of microglia is a relevant factor.
Collapse
Affiliation(s)
- María Del Mar Fernández-Arjona
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - Pablo Granados-Durán
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - Pedro Fernández-Llebrez
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| |
Collapse
|
42
|
Teo JD, Morris MJ, Jones NM. Maternal obesity increases inflammation and exacerbates damage following neonatal hypoxic-ischaemic brain injury in rats. Brain Behav Immun 2017; 63:186-196. [PMID: 27746186 DOI: 10.1016/j.bbi.2016.10.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE In humans, maternal obesity is associated with an increase in the incidence of birth related difficulties. However, the impact of maternal obesity on the severity of brain injury in offspring is not known. Recent studies have found evidence of increased glial response and inflammatory mediators in the brains as a result of obesity in humans and rodents. We hypothesised that hypoxic-ischaemic (HI) brain injury is greater in neonatal offspring from obese rat mothers compared to lean controls. METHODS Female Sprague Dawley rats were randomly allocated to high fat (HFD, n=8) or chow (n=4) diet and mated with lean male rats. On postnatal day 7 (P7), male and female pups were randomly assigned to HI injury or control (C) groups. HI injury was induced by occlusion of the right carotid artery followed by 3h exposure to 8% oxygen, at 37°C. Control pups were removed from the mother for the same duration under ambient conditions. Righting behaviour was measured on day 1 and 7 following HI. The extent of brain injury was quantified in brain sections from P14 pups using cresyl violet staining and the difference in volume between brain hemispheres was measured. RESULTS Before mating, HFD mothers were 11% heavier than Chow mothers (p<0.05, t-test). Righting reflex was delayed in offspring from HFD-fed mothers compared to the Chow mothers. The Chow-HI pups showed a loss in ipsilateral brain tissue, while the HFD-HI group had significantly greater loss. No significant difference was detected in brain volume between the HFD-C and Chow-C pups. When analysed on a per litter basis, the size of the injury was significantly correlated with maternal weight. Similar observations were made with neuronal staining showing a greater loss of neurons in the brain of offspring from HFD-mothers following HI compared to Chow. Astrocytes appeared to more hypertrophic and a greater number of microglia were present in the injured hemisphere in offspring from mothers on HFD. HI caused an increase in the proportion of amoeboid microglia and exposure to maternal HFD exacerbated this response. In the contralateral hemisphere, offspring exposed to maternal HFD displayed a reduced proportion of ramified microglia. CONCLUSIONS Our data clearly demonstrate that maternal obesity can exacerbate the severity of brain damage caused by HI in neonatal offspring. Given that previous studies have shown enhanced inflammatory responses in offspring of obese mothers, these factors including gliosis and microglial infiltration are likely to contribute to enhanced brain injury.
Collapse
Affiliation(s)
- Jonathan D Teo
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, New South Wales, Australia
| | - Margaret J Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, New South Wales, Australia
| | - Nicole M Jones
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, New South Wales, Australia.
| |
Collapse
|
43
|
Villapol S, Loane DJ, Burns MP. Sexual dimorphism in the inflammatory response to traumatic brain injury. Glia 2017; 65:1423-1438. [PMID: 28608978 DOI: 10.1002/glia.23171] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 12/19/2022]
Abstract
The activation of resident microglial cells, alongside the infiltration of peripheral macrophages, are key neuroinflammatory responses to traumatic brain injury (TBI) that are directly associated with neuronal death. Sexual disparities in response to TBI have been previously reported; however it is unclear whether a sex difference exists in neuroinflammatory progression after TBI. We exposed male and female mice to moderate-to-severe controlled cortical impact injury and studied glial cell activation in the acute and chronic stages of TBI using immunofluorescence and in situ hybridization analysis. We found that the sex response was completely divergent up to 7 days postinjury. TBI caused a rapid and pronounced cortical microglia/macrophage activation in male mice with a prominent activated phenotype that produced both pro- (IL-1β and TNFα) and anti-inflammatory (Arg1 and TGFβ) cytokines with a single-phase, sustained peak from 1 to 7 days. In contrast, TBI caused a less robust microglia/macrophage phenotype in females with biphasic pro-inflammatory response peaks at 4 h and 7 days, and a delayed anti-inflammatory mRNA peak at 30 days. We further report that female mice were protected against acute cell loss after TBI, with male mice demonstrating enhanced astrogliosis, neuronal death, and increased lesion volume through 7 days post-TBI. Collectively, these findings indicate that TBI leads to a more aggressive neuroinflammatory profile in male compared with female mice during the acute and subacute phases postinjury. Understanding how sex affects the course of neuroinflammation following brain injury is a vital step toward developing personalized and effective treatments for TBI.
Collapse
Affiliation(s)
- Sonia Villapol
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, District of Columbia
| | - David J Loane
- Department of Anesthesiology, Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark P Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, District of Columbia
| |
Collapse
|
44
|
Age-dependent regulation of obesity and Alzheimer-related outcomes by hormone therapy in female 3xTg-AD mice. PLoS One 2017; 12:e0178490. [PMID: 28575011 PMCID: PMC5456100 DOI: 10.1371/journal.pone.0178490] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/14/2017] [Indexed: 12/31/2022] Open
Abstract
Depletion of ovarian hormones at menopause is associated with increased Alzheimer's disease (AD) risk. Hormone loss also increases central adiposity, which promotes AD development. One strategy to improve health outcomes in postmenopausal women is estrogen-based hormone therapy (HT), though its efficacy is controversial. The window of opportunity hypothesis posits that HT is beneficial only if initiated near the onset of menopause. Here, we tested this hypothesis by assessing the efficacy of HT against diet-induced obesity and AD-related pathology in female 3xTg-AD mice at early versus late middle-age. HT protected against obesity and reduced β-amyloid burden only at early middle-age. One mechanism that contributes to AD pathogenesis is microglial activation, which is increased by obesity and reduced by estrogens. In parallel to its effects on β-amyloid accumulation, we observed that HT reduced morphological evidence of microglial activation in early but not late middle-age. These findings suggest that HT may be effective during human perimenopause in reducing indices of obesity and AD-related pathology, a conclusion consistent with the window of opportunity hypothesis.
Collapse
|
45
|
Leaw B, Zhu D, Tan J, Muljadi R, Saad MI, Mockler JC, Wallace EM, Lim R, Tolcos M. Human amnion epithelial cells rescue cell death via immunomodulation of microglia in a mouse model of perinatal brain injury. Stem Cell Res Ther 2017; 8:46. [PMID: 28241859 PMCID: PMC5330154 DOI: 10.1186/s13287-017-0496-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/19/2017] [Accepted: 02/09/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Human amnion epithelial cells (hAECs) are clonogenic and have been proposed to reduce inflammatory-induced tissue injury. Perturbation of the immune response is implicated in the pathogenesis of perinatal brain injury; modulating this response could thus be a novel therapy for treating or preventing such injury. The immunomodulatory properties of hAECs have been shown in other animal models, but a detailed investigation of the effects on brain immune cells following injury has not been undertaken. Here, we investigate the effects of hAECs on microglia, the first immune responders to injury within the brain. METHODS We generated a mouse model combining neonatal inflammation and perinatal hyperoxia, both of which are risk factors associated with perinatal brain injury. On embryonic day 16 we administered lipopolysaccharide (LPS), or saline (control), intra-amniotically to C57Bl/6 J mouse pups. On postnatal day (P)0, LPS pups were placed in hyperoxia (65% oxygen) and control pups in normoxia for 14 days. Pups were given either hAECs or saline intravenously on P4. RESULTS At P14, relative to controls, LPS and hyperoxia pups had reduced body weight, increased density of apoptotic cells (TUNEL) in the cortex, striatum and white matter, astrocytes (GFAP) in the white matter and activated microglia (CD68) in the cortex and striatum, but no change in total microglia density (Iba1). hAEC administration rescued the decreased body weight and reduced apoptosis and astrocyte areal coverage in the white matter, but increased the density of total and activated microglia. We then stimulated primary microglia (CD45lowCD11b+) with LPS for 24 h, followed by co-culture with hAEC conditioned medium for 48 h. hAEC conditioned medium increased microglial phagocytic activity, decreased microglia apoptosis and decreased M1 activation markers (CD86). Stimulating hAECs for 24 h with LPS did not alter release of cytokines known to modulate microglia activity. CONCLUSIONS These data demonstrate that hAECs can directly immunomodulate brain microglia, probably via release of trophic factors. This observation offers promise that hAECs may afford therapeutic utility in the management of perinatal brain injury.
Collapse
Affiliation(s)
- Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Dandan Zhu
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Jean Tan
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Ruth Muljadi
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Mohamed I. Saad
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Joanne C. Mockler
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
| | - Euan M. Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
| | - Mary Tolcos
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| |
Collapse
|
46
|
Li L, Wu Y, Bai Z, Hu Y, Li W. Blockade of NMDA receptors decreased spinal microglia activation in bee venom induced acute inflammatory pain in rats. Neurol Res 2017; 39:271-280. [DOI: 10.1080/01616412.2017.1281198] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Li Li
- Central Laboratory, Department of Science and Technology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongfang Wu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Zhifeng Bai
- Department of Pediatrics, Xingtai Medical College, Xingtai, China
| | - Yuyan Hu
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Wenbin Li
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
47
|
Monif M, Reid CA, Powell KL, Drummond KJ, O'Brien TJ, Williams DA. Interleukin-1β has trophic effects in microglia and its release is mediated by P2X7R pore. J Neuroinflammation 2016; 13:173. [PMID: 27364756 PMCID: PMC4929731 DOI: 10.1186/s12974-016-0621-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 06/10/2016] [Indexed: 11/21/2022] Open
Abstract
Background Enhanced expression of the purinergic P2X7 receptor (P2X7R) occurs in several neuroinflammatory conditions where increased microglial activation is a co-existing feature. P2X7 receptors can function either as a cation channel or, upon continued stimulation, a large pore. P2X7R-over-expression alone is sufficient to drive microglial activation and proliferation in a process that is P2X7R pore dependent, although the biological signaling pathway through which this occurs remains unclear. Once activated, microglia are known to release a number of bioactive substances that include the proinflammatory cytokine interleukin-1β (IL-1β). Previous studies have linked P2X7R stimulation to the processing and release of IL-1β, but whether the channel or pore state of P2X7R is predominant in driving IL-1β release is unknown and is a major aim of this study. In addition, we will determine whether IL-1β has trophic effects on surrounding microglia. Methods Electron microscopy and immunohistochemistry were used to delineate the sub-cellular localization of P2X7R and IL-1β in primary hippocampal rat cultures. FM1-43 fluorescent dye and confocal microscopy were used to quantify vesicular exocytosis from microglia expressing the pore-forming P2X7R versus a non-pore-forming point mutant, P2X7RG345Y. IL-1β in culture was quantified with an enzyme-linked immunosorbent assay (ELISA). IL-1β intracellular processing was blocked with inhibition of caspase 1 (with a synthetic peptide antagonist), and its extracellular form was neutralized with an IL-1β neutralizing antibody. Microglial activation and proliferation was quantified immunohistochemically with confocal microscopy. Results P2X7R and IL-1β were co-localized in lysosomes. Vesicular exocytosis was higher in microglia expressing the pore-forming P2X7R compared to those expressing the non-pore-forming mutant. There was increased IL-1β in cultures expressing the pore-forming P2X7R, and this proinflammatory cytokine was found to mediate the trophic effects of P2X7R pore in microglia. Inhibition of IL-1β production and function resulted in a significant decrease in P2X7R-mediated microglial activation and proliferation. Conclusions IL-1β is a mediator of microglial activation and proliferation, and its release/production is P2X7R pore dependent. Blockade of P2X7R pore could serve as a therapeutic target in alleviating the degree of inflammation seen in neurodegenerative and neoplastic conditions. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0621-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mastura Monif
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, 3010, Australia.,The Department of Neurology, The Royal Melbourne Hospital, Parkville, Victoria, 3052, Australia
| | - Christopher A Reid
- Howard Florey Institute, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Kim L Powell
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Katherine J Drummond
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Terrence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - David A Williams
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
48
|
Bhandare AM, Kapoor K, Farnham MM, Pilowsky PM. Microglia PACAP and glutamate: Friends or foes in seizure-induced autonomic dysfunction and SUDEP? Respir Physiol Neurobiol 2016; 226:39-50. [DOI: 10.1016/j.resp.2016.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/18/2016] [Accepted: 01/21/2016] [Indexed: 12/18/2022]
|
49
|
Sant'Anna MB, Kusuda R, Bozzo TA, Bassi GS, Alves-Filho JC, Cunha FQ, Ferreira SH, Souza GR, Cunha TM. Medial plantar nerve ligation as a novel model of neuropathic pain in mice: pharmacological and molecular characterization. Sci Rep 2016; 6:26955. [PMID: 27230787 PMCID: PMC4882539 DOI: 10.1038/srep26955] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 05/11/2016] [Indexed: 12/30/2022] Open
Abstract
Peripheral neuropathic pain is a consequence of an injury/disease of the peripheral nerves. The mechanisms involved in its pathophysiology are not entirely understood. To better understand the mechanisms involved in the development of peripheral nerve injury-induced neuropathic pain, more experimental models are required. Here, we developed a novel peripheral neuropathic pain model in mice by using a minimally invasive surgery and medial plantar nerve ligation (MPNL). After MPNL, mechanical allodynia was established, and mice quickly recovered from the surgery without any significant motor impairment. MPNL causes an increased expression of ATF-3 in the sensory neurons. At 14 days after surgery, gabapentin was capable of reversing the mechanical allodynia, whereas anti-inflammatory drugs and opioids were ineffective. MPNL-induced neuropathic pain was mediated by glial cells activation and the production of TNF-α and IL-6 in the spinal cord. These results indicate MPNL as a reasonable animal model for the study of peripheral neuropathic pain, presenting analgesic pharmacological predictivity to clinically used drugs. The results also showed molecular phenotypic changes similar to other peripheral neuropathic pain models, with the advantage of a lack of motor impairment. These features indicate that MPNL might be more appropriate for the study of neuropathic pain than classical models.
Collapse
Affiliation(s)
- Morena B Sant'Anna
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Ricardo Kusuda
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Tiago A Bozzo
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Gabriel S Bassi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil.,Graduation Program in Basic and Apply Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Sergio H Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Guilherme R Souza
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
50
|
Ohgomori T, Yamada J, Takeuchi H, Kadomatsu K, Jinno S. Comparative morphometric analysis of microglia in the spinal cord of SOD1(G93A) transgenic mouse model of amyotrophic lateral sclerosis. Eur J Neurosci 2016; 43:1340-51. [PMID: 26946061 DOI: 10.1111/ejn.13227] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 12/13/2022]
Abstract
It has long been recognized that reactive microglia undergo a series of phenotypic changes accompanying morphological transformation. However, the morphological classification of microglia has not yet been achieved. To address this issue, here we morphometrically analysed three-dimensionally reconstructed ionized calcium binding adaptor molecule 1-immunoreactive (Iba1(+) ) microglia in the ventral horn of the lumbar spinal cord of SOD1(G93A) transgenic mice, a model of amyotrophic lateral sclerosis. The hierarchical cluster analysis revealed that microglia were objectively divided into four groups: type S (named after surveillant microglia) and types R1, R2 and R3 (named after reactive microglia). For the purpose of comparative morphometry, we also analysed two pharmacological disease models using wild-type mice: 3,3'-iminodipropionitrile (IDPN)-induced axonopathy and lipopolysaccharide (LPS)-induced neuroinflammation. Type S microglia showed a typical ramified morphology of surveillant microglia, and were mostly observed in wild-type controls. Type R1 microglia were seen at the early stage of disease in SOD1(G93A) mice, and also frequently occurred in IDPN-treated mice. They exhibited small cell bodies with shorter and simple processes. Type R2 microglia were morphologically similar to type R1 microglia, but only transiently occurred in the middle stage of disease in SOD1(G93A) mice and in IDPN-treated mice. Type R3 microglia exhibited a bushy shape, and were observed in the end stage of disease in SOD1(G93A) mice and in LPS-treated mice. These findings indicate that microglia of SOD1(G93A) mice can be classified into four types, and also suggest that the phenotypic changes may be induced by the events related to axonopathy and neuroinflammation.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|