1
|
El-Shetry ES, Ibrahim IA, Kamel AM, Abdelwahab OA. Quercetin mitigates doxorubicin-induced neurodegenerative changes in the cerebral cortex and hippocampus of rats; insights to DNA damage, inflammation, synaptic plasticity. Tissue Cell 2024; 87:102313. [PMID: 38286061 DOI: 10.1016/j.tice.2024.102313] [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: 08/15/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Doxorubicin (Dox) is one of the most effective anti-neoplastic agents. Quercetin (QE) exhibits antioxidant and anti-inflammatory properties. AIM To detect neuroprotective properties of quercetin in rats exposed to doxorubicin-induced brain injury. MATERIAL AND METHODS 48 rats were allocated equally into four groups: control group: (given normal saline), QE group: (given 80 mg/kg of QE orally daily for 2 weeks), Dox group: (received 2.5 mg/kg of Dox every other day for a total of seven intraperitoneal injections), and Dox+QE group: (received 2.5 mg/kg of Dox every other day for a total of seven intraperitoneal injections and 80 mg/kg of QE orally daily for 2 weeks). Subsequently, biochemical analyses were carried out along with histopathological (light and electron microscopic) and immunohistochemical examinations of the cerebral cortex and hippocampus. RESULTS The Dox group revealed a decline in the activities of superoxide dismutase, catalase, and glutathione peroxidase, along with an increase in malondialdehyde and an increase in DNA damage. Furthermore, sections of the cerebral cortex and hippocampus revealed neurodegenerative changes, decreased synaptophysin, and increased Interleukin-1 beta expressions. Biochemical and histopathological results were markedly improved by QE administration. CONCLUSIONS It can be concluded that QE induces protective effects against Dox-induced neurotoxicity.
Collapse
Affiliation(s)
- Eman S El-Shetry
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt; Department of Anatomy, College of Medicine, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Ibrahim Amin Ibrahim
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Asmaa Mahde Kamel
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Ola Ali Abdelwahab
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
2
|
Zhao ZA, Yan L, Wen J, Satyanarayanan SK, Yu F, Lu J, Liu YU, Su H. Cellular and molecular mechanisms in vascular repair after traumatic brain injury: a narrative review. BURNS & TRAUMA 2023; 11:tkad033. [PMID: 37675267 PMCID: PMC10478165 DOI: 10.1093/burnst/tkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/01/2023] [Accepted: 05/26/2023] [Indexed: 09/08/2023]
Abstract
Traumatic brain injury (TBI) disrupts normal brain function and is associated with high morbidity and fatality rates. TBI is characterized as mild, moderate or severe depending on its severity. The damage may be transient and limited to the dura matter, with only subtle changes in cerebral parenchyma, or life-threatening with obvious focal contusions, hematomas and edema. Blood vessels are often injured in TBI. Even in mild TBI, dysfunctional cerebral vascular repair may result in prolonged symptoms and poor outcomes. Various distinct types of cells participate in vascular repair after TBI. A better understanding of the cellular response and function in vascular repair can facilitate the development of new therapeutic strategies. In this review, we analyzed the mechanism of cerebrovascular impairment and the repercussions following various forms of TBI. We then discussed the role of distinct cell types in the repair of meningeal and parenchyma vasculature following TBI, including endothelial cells, endothelial progenitor cells, pericytes, glial cells (astrocytes and microglia), neurons, myeloid cells (macrophages and monocytes) and meningeal lymphatic endothelial cells. Finally, possible treatment techniques targeting these unique cell types for vascular repair after TBI are discussed.
Collapse
Affiliation(s)
- Zi-Ai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
- Department of Neurology, General Hospital of Northern Theater Command, 83# Wen-Hua Road, Shenyang 110840, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yong U Liu
- Laboratory of Neuroimmunology in Health and Disease Institute, Guangzhou First People’s Hospital School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 511400, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
3
|
Lopez AJ, ElSaadani M, Culkin MC, Jacovides CL, Georges AP, Song H, Kaplan LJ, Kumar MA, Smith DH, Pascual JL. Persistent Blunting of Penumbral Leukocyte Mobilization by Beta Blockade Administered for Two Weeks After Traumatic Brain Injury. J Surg Res 2022; 280:196-203. [PMID: 35994981 DOI: 10.1016/j.jss.2022.06.069] [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: 02/15/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Beta-blockers (BB) after traumatic brain injury (TBI) accelerate cognitive recovery weeks after injury. BBs also inhibit leukocyte (LEU) mobilization to the penumbral blood brain barrier (BBB) 48-h after TBI. It is unclear whether the latter effects persist longer and accompany the persistent cognitive improvement. We hypothesized that 2 wk of BB after TBI reduce penumbral BBB leukocyte-endothelial interactions. METHODS Thirty CD1 mice underwent TBI (controlled cortical impact, CCI: 6 m/s velocity, 1 mm depth, 3 mm diameter) or sham craniotomy followed by i.p. saline (NS) or propranolol (1, 2, 4 mg/kg) every 12 h for 14 d. On day 14, in vivo pial intravital microscopy visualized endothelial-LEU interactions and BBB microvascular leakage. Day 14 Garcia neurological test scores and animal weights were compared to preinjury levels reflecting concurrent clinical recovery. RESULTS LEU rolling was greatest in CCI + NS when compared to sham (P = 0.03). 4 mg/kg propranolol significantly reduced postCCI LEU rolling down to uninjured sham levels (P = 0.03). LEU adhesion and microvascular permeability were not impacted at this time interval. Untreated injured animals (CCI + NS) scored lower Garcia neurological test and greater weight loss recovery at day 14 when compared to preinjury (P < 0.05). Treatment with higher doses of propranolol (2, 4 mg/kg), improved weight loss recovery (P < 0.001). CONCLUSIONS LEU rolling alone, was influenced by BB therapy 14 d after TBI suggesting that certain penumbral neuroinflammatory cellular effects of BB therapy after TBI persist up to 2 wk after injury potentially explaining the pervasive beneficial effects of BBs on learning and memory.
Collapse
Affiliation(s)
- Alfonso J Lopez
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mohamed ElSaadani
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matthew C Culkin
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christina L Jacovides
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anastasia P Georges
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hailong Song
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lewis J Kaplan
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Monisha A Kumar
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jose L Pascual
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
4
|
Savya SP, Li F, Lam S, Wellman SM, Stieger KC, Chen K, Eles JR, Kozai TDY. In vivo spatiotemporal dynamics of astrocyte reactivity following neural electrode implantation. Biomaterials 2022; 289:121784. [PMID: 36103781 PMCID: PMC10231871 DOI: 10.1016/j.biomaterials.2022.121784] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/02/2022]
Abstract
Brain computer interfaces (BCIs), including penetrating microelectrode arrays, enable both recording and stimulation of neural cells. However, device implantation inevitably causes injury to brain tissue and induces a foreign body response, leading to reduced recording performance and stimulation efficacy. Astrocytes in the healthy brain play multiple roles including regulating energy metabolism, homeostatic balance, transmission of neural signals, and neurovascular coupling. Following an insult to the brain, they are activated and gather around the site of injury. These reactive astrocytes have been regarded as one of the main contributors to the formation of a glial scar which affects the performance of microelectrode arrays. This study investigates the dynamics of astrocytes within the first 2 weeks after implantation of an intracortical microelectrode into the mouse brain using two-photon microscopy. From our observation astrocytes are highly dynamic during this period, exhibiting patterns of process extension, soma migration, morphological activation, and device encapsulation that are spatiotemporally distinct from other glial cells, such as microglia or oligodendrocyte precursor cells. This detailed characterization of astrocyte reactivity will help to better understand the tissue response to intracortical devices and lead to the development of more effective intervention strategies to improve the functional performance of neural interfacing technology.
Collapse
Affiliation(s)
- Sajishnu P Savya
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Northwestern University, USA
| | - Fan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Computational Modeling & Simulation PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephanie Lam
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Synaptamide Modulates Astroglial Activity in Mild Traumatic Brain Injury. Mar Drugs 2022; 20:md20080538. [PMID: 36005540 PMCID: PMC9410022 DOI: 10.3390/md20080538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
At present, the study of the neurotropic activity of polyunsaturated fatty acid ethanolamides (N-acylethanolamines) is becoming increasingly important. N-docosahexaenoylethanolamine (synaptamide, DHEA) is a highly active metabolite of docosahexaenoic acid (DHA) with neuroprotective, synaptogenic, neuritogenic, and anti-inflammatory properties in the nervous system. Synaptamide tested in the present study was obtained using a chemical modification of DHA isolated from squid Berryteuthis magister liver. The results of this study demonstrate the effects of synaptamide on the astroglial response to injury in the acute (1 day) and chronic (7 days) phases of mild traumatic brain injury (mTBI) development. HPLC-MS study revealed several times increase of synaptamide concentration in the cerebral cortex and serum of experimental animals after subcutaneous administration (10 mg/kg/day). Using immunohistochemistry, it was shown that synaptamide regulates the activation of GFAP- and S100β-positive astroglia, reduce nNOS-positive immunostaining, and stimulates the secretion of neurotrophin BDNF. Dynamics of superoxide dismutase production in synaptamide treatment confirm the antioxidant efficacy of the test compound. We found a decrease in TBI biomarkers such as GFAP, S100β, and IL-6 in the blood serum of synaptamide-treated experimental animals using Western blot analysis. The results indicate the high therapeutic potential of synaptamide in reducing the severity of the brain damage consequences.
Collapse
|
6
|
A single factor elicits multilineage reprogramming of astrocytes in the adult mouse striatum. Proc Natl Acad Sci U S A 2022; 119:e2107339119. [PMID: 35254903 PMCID: PMC8931246 DOI: 10.1073/pnas.2107339119] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Outside the neurogenic niches, the adult brain lacks multipotent progenitor cells. In this study, we performed a series of in vivo screens and reveal that a single factor can induce resident brain astrocytes to become induced neural progenitor cells (iNPCs), which then generate neurons, astrocytes, and oligodendrocytes. Such a conclusion is supported by single-cell RNA sequencing and multiple lineage-tracing experiments. Our discovery of iNPCs is fundamentally important for regenerative medicine since neural injuries or degeneration often lead to loss/dysfunction of all three neural lineages. Our findings also provide insights into cell plasticity in the adult mammalian brain, which has largely lost the regenerative capacity. Astrocytes in the adult brain show cellular plasticity; however, whether they have the potential to generate multiple lineages remains unclear. Here, we perform in vivo screens and identify DLX2 as a transcription factor that can unleash the multipotentiality of adult resident astrocytes. Genetic lineage tracing and time-course analyses reveal that DLX2 enables astrocytes to rapidly become ASCL1+ neural progenitor cells, which give rise to neurons, astrocytes, and oligodendrocytes in the adult mouse striatum. Single-cell transcriptomics and pseudotime trajectories further confirm a neural stem cell-like behavior of reprogrammed astrocytes, transitioning from quiescence to activation, proliferation, and neurogenesis. Gene regulatory networks and mouse genetics identify and confirm key nodes mediating DLX2-dependent fate reprogramming. These include activation of endogenous DLX family transcription factors and suppression of Notch signaling. Such reprogramming-induced multipotency of resident glial cells may be exploited for neural regeneration.
Collapse
|
7
|
El-Domiaty HF, El-Roghy ES, Salem HR. Combination of magnesium supplementation with treadmill exercise improves memory deficit in aged rats by enhancing hippocampal neurogenesis and plasticity: a functional and histological study. Appl Physiol Nutr Metab 2022; 47:296-308. [PMID: 35225658 DOI: 10.1139/apnm-2021-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to investigate the possible ameliorative effects of co-supplementation with Mg2+ and treadmill exercise on memory deficit in aged rats. Fifty male albino rats (10 young and 40 aged rats) were divided into 5 groups (10 rats/group): young, aged sedentary, aged exercised, aged Mg2+-supplemented, and aged exercised and Mg2+-supplemented. Memory was assessed using the Y-maze and novel object recognition tests. Plasma samples were collected for measurement of C-reactive protein (CRP). Subsequently, brain malondialdehyde and catalase levels were measured. Histological and immunohistochemical analyses of the hippocampi were performed. Our results showed impaired memory in aged sedentary rats, with significantly elevated plasma CRP and brain malondialdehyde levels and decreased brain catalase. The hippocampus of aged sedentary rats showed cellular degeneration, downregulation of synaptophysin (SYP) and proliferating cell nuclear antigen (PCNA), and upregulation of glial fibrillary acidic protein (GFAP) and caspase-3. Mg2+ supplementation and/or treadmill exercise significantly improved memory tests in aged rats, which could be explained by the upregulation of hippocampal SYP and PCNA expression and downregulation of GFAP and caspase-3 expression with antioxidant and anti-inflammatory mechanisms. The combined therapy had a better effect than both treatments alone, confirming the role of Mg2+ supplementation with physical exercise in enhancing age-related memory deficit. Novelty: Magnesium supplementation with treadmill exercise improves memory deficit in aged rats. The possible mechanisms are upregulation of the hippocampal synaptophysin and PCNA, downregulation of GFAP and caspase-3, the antioxidant and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Heba Fathy El-Domiaty
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia Governorate, Egypt
| | - Eman S El-Roghy
- Histology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia Governorate, Egypt
| | - Heba Rady Salem
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia Governorate, Egypt
| |
Collapse
|
8
|
Cramer SW, Carter RE, Aronson JD, Kodandaramaiah SB, Ebner TJ, Chen CC. Through the looking glass: A review of cranial window technology for optical access to the brain. J Neurosci Methods 2021; 354:109100. [PMID: 33600850 PMCID: PMC8100903 DOI: 10.1016/j.jneumeth.2021.109100] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Deciphering neurologic function is a daunting task, requiring understanding the neuronal networks and emergent properties that arise from the interactions among single neurons. Mechanistic insights into neuronal networks require tools that simultaneously assess both single neuron activity and the consequent mesoscale output. The development of cranial window technologies, in which the skull is thinned or replaced with a synthetic optical interface, has enabled monitoring neuronal activity from subcellular to mesoscale resolution in awake, behaving animals when coupled with advanced microscopy techniques. Here we review recent achievements in cranial window technologies, appraise the relative merits of each design and discuss the future research in cranial window design.
Collapse
Affiliation(s)
- Samuel W Cramer
- Department of Neurosurgery, University of Minnesota, 420 Delaware St SE, Mayo D429, MMC 96, Twin Cities, Minneapolis, MN, 55455, USA
| | - Russell E Carter
- Department of Neuroscience, University of Minnesota, Twin Cities, Room 421, 2001 Sixth Street S.E., Minneapolis, MN, 55455 MN, USA
| | - Justin D Aronson
- Department of Neuroscience, University of Minnesota, Twin Cities, Room 421, 2001 Sixth Street S.E., Minneapolis, MN, 55455 MN, USA
| | - Suhasa B Kodandaramaiah
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, MN, USA; Department of Biomedical Engineering, University of Minnesota, Twin Cities, MN, USA; Graduate Program in Neuroscience, University of Minnesota, Twin Cities, MN, USA
| | - Timothy J Ebner
- Department of Neuroscience, University of Minnesota, Twin Cities, Room 421, 2001 Sixth Street S.E., Minneapolis, MN, 55455 MN, USA.
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, 420 Delaware St SE, Mayo D429, MMC 96, Twin Cities, Minneapolis, MN, 55455, USA.
| |
Collapse
|
9
|
Liang Y, Walczak P. Long term intravital single cell tracking under multiphoton microscopy. J Neurosci Methods 2020; 349:109042. [PMID: 33340557 DOI: 10.1016/j.jneumeth.2020.109042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Visualizing and tracking cells over time in a living organism has been a much-coveted dream before the invention of intravital microscopy. The opaque nature of tissue was a major hurdle that was remedied by the multiphoton microscopy. With the advancement of optical imaging and fluorescent labeling tools, intravital high resolution imaging has become increasingly accessible over the past few years. Long-term intravital tracking of single cells (LIST) under multiphoton microscopy provides a unique opportunity to gain insight into the longitudinal changes in the morphology, migration, or function of cells or subcellular structures. It is particularly suitable for studying slow-evolving cellular and molecular events during normal development or disease progression, without losing the opportunity of catching fast events such as calcium signals. Here, we review the application of LIST under 2-photon microscopy in various fields of neurobiology and discuss challenges and new directions in labeling and imaging methods for LIST. Overall, this review provides an overview of current applications of LIST in mammals, which is an emerging field that will contribute to a better understanding of essential molecular and cellular events in health and disease.
Collapse
Affiliation(s)
- Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Qin J, Li Z, Gong G, Li H, Chen L, Song B, Liu X, Shi C, Yang J, Yang T, Xu Y. Early increased neutrophil-to-lymphocyte ratio is associated with poor 3-month outcomes in spontaneous intracerebral hemorrhage. PLoS One 2019; 14:e0211833. [PMID: 30730945 PMCID: PMC6366889 DOI: 10.1371/journal.pone.0211833] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/21/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to evaluate the association of dynamic neutrophil-to-lymphocyte ratio (NLR) with 3-month functional outcomes in patients with sICH. We retrospectively identified 213 consecutive patients with sICH hospitalized in The First Affiliated Hospital of Zhengzhou University from January 2017 to May 2018. Patients were divided into functional independence (FI) or unfavorable prognosis (UP) groups based on 3-month outcomes. Admission leukocyte counts within 24 hours of symptom onset were obtained, and the recorded fraction, of which the numerator is neutrophil and the denominator is lymphocyte, as NLR0. Determined NLR1, NLR3, NLR7, and NLR14 were recorded on day 1 (n = 77), day 3 (n = 126), day 7 (n = 123), and day 14 (n = 105), respectively. The relationships between dynamic NLR or leukocyte counts and clinical features were evaluated using Spearman’s or Kendall’s correlation analysis. Logistic regression analyses were used to identify the risk factors for unfavorable 3-month prognosis. The patients’ dynamic NLR was positively associated with the National Institutes of Health Stroke Scale, ICH score, and hematoma volume at admission, while inversely correlated to the onset GCS score and FI at 3-month follow-up. Furthermore, higher NLR or lower absolute lymphocyte count obtained at admission was independently risk factor for UP at 3 months (adjusted odds ratio [OR]: 1.06, 95% confidence interval [CI]: 1.003, 1.12; OR: 0.41, 95% CI: 0.18, 0.94, respectively). In conclusion, higher NLR and lower lymphocyte counts at early stages were predictive of 3-month unfavorable outcomes in sICH patients.
Collapse
Affiliation(s)
- Jie Qin
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
- * E-mail: (JQ); (GG)
| | - Zhu Li
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Guangming Gong
- Department of Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
- * E-mail: (JQ); (GG)
| | - Hongwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Ling Chen
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Bo Song
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Xinjing Liu
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Changhe Shi
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Jing Yang
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Ting Yang
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Yuming Xu
- Third Department of Neurology and Key Disciplines Laboratory of Clinical Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| |
Collapse
|
11
|
Inflammation in Traumatic Brain Injury: Roles for Toxic A1 Astrocytes and Microglial-Astrocytic Crosstalk. Neurochem Res 2019; 44:1410-1424. [PMID: 30661228 DOI: 10.1007/s11064-019-02721-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury triggers neuroinflammation that may contribute to progressive neurodegeneration. We investigated patterns of recruitment of astrocytes and microglia to inflammation after brain trauma by firstly characterising expression profiles over time of marker genes following TBI, and secondly by monitoring glial morphologies reflecting inflammatory responses in a rat model of traumatic brain injury (i.e. the lateral fluid percussion injury). Gene expression profiles revealed early elevation of expression of astrocytic marker glial fibrillary acidic protein relative to microglial marker allograft inflammatory factor 1 (also known as ionized calcium-binding adapter molecule 1). Adult rat brains collected at day 7 after injury were processed for immunohistochemistry with allograft inflammatory factor 1, glial fibrillary acidic protein and complement C3 (marker of bad/disruptive astrocytic A1 phenotype). Astrocytes positive for glial fibrillary acidic protein and complement C3 were significant increased in the injured cortex and displayed more complex patterns of arbourisation with significantly increased bifurcations. Our observations suggested that traumatic brain injury changed the phenotype of microglia from a ramified appearance with long, thin, highly branched processes to a swollen amoeboid shape in the injured cortex. These findings suggest differential glial activation with astrocytes likely undergoing strategic changes in morphology and function. Whilst a detailed analysis is needed of temporal patterns of glial activation, ours is the first evidence of a role for the bad/disruptive astrocytic A1 phenotype in an open head model of traumatic brain injury.
Collapse
|
12
|
Martin-Jiménez C, Gaitán-Vaca DM, Areiza N, Echeverria V, Ashraf GM, González J, Sahebkar A, Garcia-Segura LM, Barreto GE. Astrocytes Mediate Protective Actions of Estrogenic Compounds after Traumatic Brain Injury. Neuroendocrinology 2019; 108:142-160. [PMID: 30391959 DOI: 10.1159/000495078] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/02/2018] [Indexed: 11/19/2022]
Abstract
Traumatic brain injury (TBI) is a serious public health problem. It may result in severe neurological disabilities and in a variety of cellular metabolic alterations for which available therapeutic strategies are limited. In the last decade, the use of estrogenic compounds, which activate protective mechanisms in astrocytes, has been explored as a potential experimental therapeutic approach. Previous works have suggested estradiol (E2) as a neuroprotective hormone that acts in the brain by binding to estrogen receptors (ERs). Several steroidal and nonsteroidal estrogenic compounds can imitate the effects of estradiol on ERs. These include hormonal estrogens, phytoestrogens and synthetic estrogens, such as selective ER modulators or tibolone. Current evidence of the role of astrocytes in mediating protective actions of estrogenic compounds after TBI is reviewed in this paper. We conclude that the use of estrogenic compounds to modulate astrocytic properties is a promising therapeutic approach for the treatment of TBI.
Collapse
Affiliation(s)
- Cynthia Martin-Jiménez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Diana Milena Gaitán-Vaca
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Natalia Areiza
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Valentina Echeverria
- Universidad San Sebastián, Fac. Cs de la Salud, Concepción, Chile
- Research and Development Service, Bay Pines VA Healthcare System, Bay Pines, Florida, USA
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias Pontificia Universidad Javeriana, Bogotá, Colombia,
| |
Collapse
|
13
|
Madry C, Arancibia-Cárcamo IL, Kyrargyri V, Chan VTT, Hamilton NB, Attwell D. Effects of the ecto-ATPase apyrase on microglial ramification and surveillance reflect cell depolarization, not ATP depletion. Proc Natl Acad Sci U S A 2018; 115:E1608-E1617. [PMID: 29382767 PMCID: PMC5816168 DOI: 10.1073/pnas.1715354115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Microglia, the brain's innate immune cells, have highly motile processes which constantly survey the brain to detect infection, remove dying cells, and prune synapses during brain development. ATP released by tissue damage is known to attract microglial processes, but it is controversial whether an ambient level of ATP is needed to promote constant microglial surveillance in the normal brain. Applying the ATPase apyrase, an enzyme which hydrolyzes ATP and ADP, reduces microglial process ramification and surveillance, suggesting that ambient ATP/ADP maintains microglial surveillance. However, attempting to raise the level of ATP/ADP by blocking the endogenous ecto-ATPase (termed NTPDase1/CD39), which also hydrolyzes ATP/ADP, does not affect the cells' ramification or surveillance, nor their membrane currents, which respond to even small rises of extracellular [ATP] or [ADP] with the activation of K+ channels. This indicates a lack of detectable ambient ATP/ADP and ecto-ATPase activity, contradicting the results with apyrase. We resolve this contradiction by demonstrating that contamination of commercially available apyrase by a high K+ concentration reduces ramification and surveillance by depolarizing microglia. Exposure to the same K+ concentration (without apyrase added) reduced ramification and surveillance as with apyrase. Dialysis of apyrase to remove K+ retained its ATP-hydrolyzing activity but abolished the microglial depolarization and decrease of ramification produced by the undialyzed enzyme. Thus, applying apyrase affects microglia by an action independent of ATP, and no ambient purinergic signaling is required to maintain microglial ramification and surveillance. These results also have implications for hundreds of prior studies that employed apyrase to hydrolyze ATP/ADP.
Collapse
Affiliation(s)
- Christian Madry
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom;
- Institute of Neurophysiology, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - I Lorena Arancibia-Cárcamo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Vasiliki Kyrargyri
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Victor T T Chan
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Nicola B Hamilton
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom;
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom;
| |
Collapse
|
14
|
Abstract
Neuronal survival, electrical signaling and synaptic activity require a well-balanced micro-environment in the central nervous system. This is achieved by the blood-brain barrier (BBB), an endothelial barrier situated in the brain capillaries, that controls near-to-all passage in and out of the brain. The endothelial barrier function is highly dependent on signaling interactions with surrounding glial, neuronal and vascular cells, together forming the neuro-glio-vascular unit. Within this functional unit, connexin (Cx) channels are of utmost importance for intercellular communication between the different cellular compartments. Connexins are best known as the building blocks of gap junction (GJ) channels that enable direct cell-cell transfer of metabolic, biochemical and electric signals. In addition, beyond their role in direct intercellular communication, Cxs also form unapposed, non-junctional hemichannels in the plasma membrane that allow the passage of several paracrine messengers, complementing direct GJ communication. Within the NGVU, Cxs are expressed in vascular endothelial cells, including those that form the BBB, and are eminent in astrocytes, especially at their endfoot processes that wrap around cerebral vessels. However, despite the density of Cx channels at this so-called gliovascular interface, it remains unclear as to how Cx-based signaling between astrocytes and BBB endothelial cells may converge control over BBB permeability in health and disease. In this review we describe available evidence that supports a role for astroglial as well as endothelial Cxs in the regulation of BBB permeability during development as well as in disease states.
Collapse
|
15
|
FGF-1 Triggers Pannexin-1 Hemichannel Opening in Spinal Astrocytes of Rodents and Promotes Inflammatory Responses in Acute Spinal Cord Slices. J Neurosci 2017; 36:4785-801. [PMID: 27122036 DOI: 10.1523/jneurosci.4195-15.2016] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/16/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED We show here that the growth factor FGF-1 is proinflammatory in the spinal cord and explore the inflammatory mechanisms. FGF-1 applied to rat spinal astrocytes in culture initiates calcium signaling and induces secretion of ATP that within minutes increases membrane permeability to ethidium (Etd(+)) and Ca(2+) by activating P2X7 receptors (P2X7Rs) that open pannexin hemichannels (Px1 HCs) that release further ATP; by 7 h treatment, connexin 43 hemichannels (Cx43 HCs) are also opened. In acute mouse spinal cord slices ex vivo, we found that FGF-1 treatment for 1 h increases the percentage of GFAP-positive astrocytes that show enhanced Px1 HC-mediated Etd(+) uptake. This response to FGF-1 was not observed in astrocytes in slices of cerebral cortex. FGF-1-induced dye uptake by astrocytes is prevented by BAPTA-AM or a phospholipase C (PLC) inhibitor. Furthermore, in spinal cord slices, P2X7R antagonists (BBG and A740003) and Px1 HC blockers ((10)Panx1 and carbenoxolone) prevent the increase in Etd(+) uptake by astrocytes, whereas Gap19, a selective Cx43 HC blocker, has no effect on dye uptake at this time. Microglia are not required for the increase in Etd(+) uptake by astrocytes induced by FGF-1, although they are activated by FGF-1 treatment. The morphological signs of microglia activation are inhibited by P2X7R antagonists and (10)Panx1 and are associated with elevated levels of proinflammatory cytokines in cord slices treated with FGF-1. The FGF-1 initiated cascade may play an important role in spinal cord inflammation in vivo SIGNIFICANCE STATEMENT We find that FGF-1 elevates [Ca(2+)]i in spinal astrocytes, which causes vesicular release of ATP and activation of P2X7Rs to trigger opening of Px1 HCs, which release further ATP. This regenerative response occurs in astrocyte cultures and in acute spinal cord slices. In the latter, FGF-1 application promotes the activation of microglia and increases the production of proinflammatory cytokines through mechanisms depending on P2X7 receptors and Px1 HCs. This proinflammatory microenvironment may favor recruitment of leukocytes into the spinal cord and impacts negatively on neuronal structure and function in vivo Any step in these processes provides a potential therapeutic target for treatment of secondary damage in various spinal cord pathologies.
Collapse
|
16
|
Coles JA, Myburgh E, Brewer JM, McMenamin PG. Where are we? The anatomy of the murine cortical meninges revisited for intravital imaging, immunology, and clearance of waste from the brain. Prog Neurobiol 2017; 156:107-148. [PMID: 28552391 DOI: 10.1016/j.pneurobio.2017.05.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 04/25/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022]
Abstract
Rapid progress is being made in understanding the roles of the cerebral meninges in the maintenance of normal brain function, in immune surveillance, and as a site of disease. Most basic research on the meninges and the neural brain is now done on mice, major attractions being the availability of reporter mice with fluorescent cells, and of a huge range of antibodies useful for immunocytochemistry and the characterization of isolated cells. In addition, two-photon microscopy through the unperforated calvaria allows intravital imaging of the undisturbed meninges with sub-micron resolution. The anatomy of the dorsal meninges of the mouse (and, indeed, of all mammals) differs considerably from that shown in many published diagrams: over cortical convexities, the outer layer, the dura, is usually thicker than the inner layer, the leptomeninx, and both layers are richly vascularized and innervated, and communicate with the lymphatic system. A membrane barrier separates them and, in disease, inflammation can be localized to one layer or the other, so experimentalists must be able to identify the compartment they are studying. Here, we present current knowledge of the functional anatomy of the meninges, particularly as it appears in intravital imaging, and review their role as a gateway between the brain, blood, and lymphatics, drawing on information that is scattered among works on different pathologies.
Collapse
Affiliation(s)
- Jonathan A Coles
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davis Building, University of Glasgow, Glasgow, G12 8TA, United Kingdom.
| | - Elmarie Myburgh
- Centre for Immunology and Infection Department of Biology, University of York, Wentworth Way, Heslington, York YO10 5DD, United Kingdom
| | - James M Brewer
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davis Building, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Paul G McMenamin
- Department of Anatomy & Developmental Biology, School of Biomedical and Psychological Sciences and Monash Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, 10 Chancellor's Walk, Clayton, Victoria, 3800, Australia
| |
Collapse
|
17
|
Sun D, Moore S, Jakobs TC. Optic nerve astrocyte reactivity protects function in experimental glaucoma and other nerve injuries. J Exp Med 2017; 214:1411-1430. [PMID: 28416649 PMCID: PMC5413323 DOI: 10.1084/jem.20160412] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 12/05/2016] [Accepted: 03/08/2017] [Indexed: 01/06/2023] Open
Abstract
Reactive remodeling of optic nerve head astrocytes is consistently observed in glaucoma and other optic nerve injuries. However, it is unknown whether this reactivity is beneficial or harmful for visual function. In this study, we used the Cre recombinase (Cre)-loxP system under regulation of the mouse glial fibrillary acidic protein promoter to knock out the transcription factor signal transducer and activator of transcription 3 (STAT3) from astrocytes and test the effect this has on reactive remodeling, ganglion cell survival, and visual function after experimental glaucoma and nerve crush. After injury, STAT3 knockout mice displayed attenuated astrocyte hypertrophy and reactive remodeling; astrocytes largely maintained their honeycomb organization and glial tubes. These changes were associated with increased loss of ganglion cells and visual function over a 30-day period. Thus, reactive astrocytes play a protective role, preserving visual function. STAT3 signaling is an important mediator of various aspects of the reactive phenotype within optic nerve astrocytes.
Collapse
Affiliation(s)
- Daniel Sun
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - Sara Moore
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
18
|
Hanak BW, Bonow RH, Harris CA, Browd SR. Cerebrospinal Fluid Shunting Complications in Children. Pediatr Neurosurg 2017; 52:381-400. [PMID: 28249297 PMCID: PMC5915307 DOI: 10.1159/000452840] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
Abstract
Although cerebrospinal fluid (CSF) shunt placement is the most common procedure performed by pediatric neurosurgeons, shunts remain among the most failure-prone life-sustaining medical devices implanted in modern medical practice. This article provides an overview of the mechanisms of CSF shunt failure for the 3 most commonly employed definitive CSF shunts in the practice of pediatric neurosurgery: ventriculoperitoneal, ventriculopleural, and ventriculoatrial. The text has been partitioned into the broad modes of shunt failure: obstruction, infection, mechanical shunt failure, overdrainage, and distal catheter site-specific failures. Clinical management strategies for the various modes of shunt failure are discussed as are research efforts directed towards reducing shunt complication rates. As it is unlikely that CSF shunting will become an obsolete procedure in the foreseeable future, it is incumbent on the pediatric neurosurgery community to maintain focused efforts to improve our understanding of and management strategies for shunt failure and shunt-related morbidity.
Collapse
Affiliation(s)
- Brian W. Hanak
- Department of Neurological Surgery, University of Washington and Seattle Children’s Hospital, Seattle, WA
| | - Robert H. Bonow
- Department of Neurological Surgery, University of Washington and Seattle Children’s Hospital, Seattle, WA
| | - Carolyn A. Harris
- Department of Neurosurgery, Wayne State University and Children’s Hospital of Michigan, Detroit, MI, USA
| | - Samuel R. Browd
- Department of Neurological Surgery, University of Washington and Seattle Children’s Hospital, Seattle, WA
| |
Collapse
|
19
|
Hanak BW, Ross EF, Harris CA, Browd SR, Shain W. Toward a better understanding of the cellular basis for cerebrospinal fluid shunt obstruction: report on the construction of a bank of explanted hydrocephalus devices. J Neurosurg Pediatr 2016; 18:213-23. [PMID: 27035548 PMCID: PMC5915300 DOI: 10.3171/2016.2.peds15531] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Shunt obstruction by cells and/or tissue is the most common cause of shunt failure. Ventricular catheter obstruction alone accounts for more than 50% of shunt failures in pediatric patients. The authors sought to systematically collect explanted ventricular catheters from the Seattle Children's Hospital with a focus on elucidating the cellular mechanisms underlying obstruction. METHODS In the operating room, explanted hardware was placed in 4% paraformaldehyde. Weekly, samples were transferred to buffer solution and stored at 4°C. After consent was obtained for their use, catheters were labeled using cell-specific markers for astrocytes (glial fibrillary acidic protein), microglia (ionized calcium-binding adapter molecule 1), and choroid plexus (transthyretin) in conjunction with a nuclear stain (Hoechst). Catheters were mounted in custom polycarbonate imaging chambers. Three-dimensional, multispectral, spinning-disk confocal microscopy was used to image catheter cerebrospinal fluid-intake holes (10× objective, 499.2-μm-thick z-stack, 2.4-μm step size, Olympus IX81 inverted microscope with motorized stage and charge-coupled device camera). Values are reported as the mean ± standard error of the mean and were compared using a 2-tailed Mann-Whitney U-test. Significance was defined at p < 0.05. RESULTS Thirty-six ventricular catheters have been imaged to date, resulting in the following observations: 1) Astrocytes and microglia are the dominant cell types bound directly to catheter surfaces; 2) cellular binding to catheters is ubiquitous even if no grossly visible tissue is apparent; and 3) immunohistochemical techniques are of limited utility when a catheter has been exposed to Bugbee wire electrocautery. Statistical analysis of 24 catheters was performed, after excluding 7 catheters exposed to Bugbee wire cautery, 3 that were poorly fixed, and 2 that demonstrated pronounced autofluorescence. This analysis revealed that catheters with a microglia-dominant cellular response tended to be implanted for shorter durations (24.7 ± 6.7 days) than those with an astrocyte-dominant response (1183 ± 642 days; p = 0.027). CONCLUSIONS Ventricular catheter occlusion remains a significant source of shunt morbidity in the pediatric population, and given their ability to intimately associate with catheter surfaces, astrocytes and microglia appear to be critical to this pathophysiology. Microglia tend to be the dominant cell type on catheters implanted for less than 2 months, while astrocytes tend to be the most prevalent cell type on catheters implanted for longer time courses and are noted to serve as an interface for the secondary attachment of ependymal cells and choroid plexus.
Collapse
Affiliation(s)
- Brian W. Hanak
- Center for Integrative Brain Research, Seattle Children’s Research Institute,Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Emily F. Ross
- Center for Integrative Brain Research, Seattle Children’s Research Institute
| | - Carolyn A. Harris
- Department of Neurosurgery, Wayne State University, Detroit, Michigan
| | - Samuel R. Browd
- Center for Integrative Brain Research, Seattle Children’s Research Institute,Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - William Shain
- Center for Integrative Brain Research, Seattle Children’s Research Institute,Department of Neurological Surgery, University of Washington, Seattle, Washington
| |
Collapse
|
20
|
Eyo UB, Miner SA, Weiner JA, Dailey ME. Developmental changes in microglial mobilization are independent of apoptosis in the neonatal mouse hippocampus. Brain Behav Immun 2016; 55:49-59. [PMID: 26576723 PMCID: PMC4864211 DOI: 10.1016/j.bbi.2015.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/09/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022] Open
Abstract
During CNS development, microglia transform from highly mobile amoeboid-like cells to primitive ramified forms and, finally, to highly branched but relatively stationary cells in maturity. The factors that control developmental changes in microglia are largely unknown. Because microglia detect and clear apoptotic cells, developmental changes in microglia may be controlled by neuronal apoptosis. Here, we assessed the extent to which microglial cell density, morphology, motility, and migration are regulated by developmental apoptosis, focusing on the first postnatal week in the mouse hippocampus when the density of apoptotic bodies peaks at postnatal day 4 and declines sharply thereafter. Analysis of microglial form and distribution in situ over the first postnatal week showed that, although there was little change in the number of primary microglial branches, microglial cell density increased significantly, and microglia were often seen near or engulfing apoptotic bodies. Time-lapse imaging in hippocampal slices harvested at different times over the first postnatal week showed differences in microglial motility and migration that correlated with the density of apoptotic bodies. The extent to which these changes in microglia are driven by developmental neuronal apoptosis was assessed in tissues from BAX null mice lacking apoptosis. We found that apoptosis can lead to local microglial accumulation near apoptotic neurons in the pyramidal cell body layer but, unexpectedly, loss of apoptosis did not alter overall microglial cell density in vivo or microglial motility and migration in ex vivo tissue slices. These results demonstrate that developmental changes in microglial form, distribution, motility, and migration occur essentially normally in the absence of developmental apoptosis, indicating that factors other than neuronal apoptosis regulate these features of microglial development.
Collapse
|
21
|
Immune Surveillance of the CNS following Infection and Injury. Trends Immunol 2016; 36:637-650. [PMID: 26431941 DOI: 10.1016/j.it.2015.08.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/10/2015] [Accepted: 08/10/2015] [Indexed: 12/24/2022]
Abstract
The central nervous system (CNS) contains a sophisticated neural network that must be constantly surveyed in order to detect and mitigate a diverse array of challenges. The innate and adaptive immune systems actively participate in this surveillance, which is critical for the maintenance of CNS homeostasis and can facilitate the resolution of infections, degeneration, and tissue damage. Infections and sterile injuries represent two common challenges imposed on the CNS that require a prompt immune response. While the inducers of these two challenges differ in origin, the resultant responses orchestrated by the CNS share some overlapping features. Here, we review how the CNS immunologically discriminates between pathogens and sterile injuries, mobilizes an immune reaction, and, ultimately, regulates local and peripherally-derived immune cells to provide a supportive milieu for tissue repair.
Collapse
|
22
|
Price L, Wilson C, Grant G. Blood–Brain Barrier Pathophysiology following Traumatic Brain Injury. TRANSLATIONAL RESEARCH IN TRAUMATIC BRAIN INJURY 2015. [DOI: 10.1201/b18959-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
24
|
Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP. Neuroinflammation in Alzheimer's disease. Lancet Neurol 2015; 14:388-405. [PMID: 25792098 DOI: 10.1016/s1474-4422(15)70016-5] [Citation(s) in RCA: 3948] [Impact Index Per Article: 394.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing evidence suggests that Alzheimer's disease pathogenesis is not restricted to the neuronal compartment, but includes strong interactions with immunological mechanisms in the brain. Misfolded and aggregated proteins bind to pattern recognition receptors on microglia and astroglia, and trigger an innate immune response characterised by release of inflammatory mediators, which contribute to disease progression and severity. Genome-wide analysis suggests that several genes that increase the risk for sporadic Alzheimer's disease encode factors that regulate glial clearance of misfolded proteins and the inflammatory reaction. External factors, including systemic inflammation and obesity, are likely to interfere with immunological processes of the brain and further promote disease progression. Modulation of risk factors and targeting of these immune mechanisms could lead to future therapeutic or preventive strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Michael T Heneka
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany; German Center for Neurodegnerative Diseases (DZNE), Bonn, Germany.
| | - Monica J Carson
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California, Riverside, CA, USA
| | - Joseph El Khoury
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Gary E Landreth
- Alzheimer Research Laboratory, Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Andreas H Jacobs
- Department of Geriatrics, Johanniter Hospital, Bonn, Germany; European Institute for Molecular Imaging (EIMI) at the Westfalian Wilhelms University (WWU), Münster, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Javier Vitorica
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Cientificas Universidad de Sevilla, Sevilla, Spain
| | - Richard M Ransohoff
- Department of Neuroscience, Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Karl Herrup
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong
| | - Sally A Frautschy
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, the Geriatric, Research, and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Bente Finsen
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, Basque Foundation for Science (IKERBASQUE), Bilbao, Spain; Department of Neurosciences, University of the Basque Country UPV/EHU (Euskal Herriko Unibertsitatea/Universidad del País Vasco) and CIBERNED (Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas), Leioa, Spain
| | - Koji Yamanaka
- Research Institute of Environmental Medicine, Nagoya University/RIKEN Brain Science Institute, Wako-shi, Japan
| | - Jari Koistinaho
- Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Eicke Latz
- German Center for Neurodegnerative Diseases (DZNE), Bonn, Germany; Institute of Innate Immunity, University of Bonn, Bonn, Germany; Department of InfectiousDiseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Annett Halle
- Max-Planck Research Group Neuroimmunology, Center of Advanced European Studies and Research (CAESAR), Bonn, Germany
| | - Gabor C Petzold
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany; German Center for Neurodegnerative Diseases (DZNE), Bonn, Germany
| | - Terrence Town
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Dave Morgan
- Department of Molecular Pharmacology and Physiology, Byrd Alzheimer's Institute, University of South Florida College of Medicine, Tampa, FL, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - V Hugh Perry
- School of Biological Sciences, Southampton General Hospital, Southampton, UK
| | - Clive Holmes
- Clinical and Experimental Science, University of Southampton, Southampton, UK; Memory Assessment and Research Centre, Moorgreen Hospital, Southern Health Foundation Trust, Southampton, UK
| | - Nicolas G Bazan
- Louisiana State University Neuroscience Center of Excellence, Louisiana State University Health Sciences Center School of Medicine in New Orleans, LA, USA
| | - David J Brooks
- Division of Experimental Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Stéphane Hunot
- Centre National de la Recherche Scientifique (CNRS), UMR 7225, Experimental Therapeutics of Neurodegeneration, Paris, France
| | - Bertrand Joseph
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Nikolaus Deigendesch
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Olga Garaschuk
- Institute of Physiology II, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Erik Boddeke
- Department of Neuroscience, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | | | - John C Breitner
- Centre for Studies on Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, and the McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Greg M Cole
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, the Geriatric, Research, and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Douglas T Golenbock
- Department of InfectiousDiseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Markus P Kummer
- Department of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
25
|
Pozner A, Xu B, Palumbos S, Gee JM, Tvrdik P, Capecchi MR. Intracellular calcium dynamics in cortical microglia responding to focal laser injury in the PC::G5-tdT reporter mouse. Front Mol Neurosci 2015; 8:12. [PMID: 26005403 PMCID: PMC4424843 DOI: 10.3389/fnmol.2015.00012] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/16/2015] [Indexed: 11/13/2022] Open
Abstract
Microglia, the resident immune cells of the brain parenchyma, are highly responsive to tissue injury. Following cell damage, microglial processes redirect their motility from randomly scouting the extracellular space to specifically reaching toward the compromised tissue. While the cell morphology aspects of this defense mechanism have been characterized, the intracellular events underlying these responses remain largely unknown. Specifically, the role of intracellular Ca2+ dynamics has not been systematically investigated in acutely activated microglia due to technical difficulty. Here we used live two-photon imaging of the mouse cortex ubiquitously expressing the genetically encoded Ca2+ indicator GCaMP5G and fluorescent marker tdTomato in central nervous system microglia. We found that spontaneous Ca2+ transients in microglial somas and processes were generally low (only 4% of all microglia showing transients within 20 min), but baseline activity increased about 8-fold when the animals were treated with LPS 12 h before imaging. When challenged with focal laser injury, an additional surge in Ca2+ activity was observed in the somas and protruding processes. Notably, coherent and simultaneous Ca2+ rises in multiple microglial cells were occasionally detected in LPS-treated animals. We show that Ca2+ transients were pre-dominantly mediated via purinergic receptors. This work demonstrates the usefulness of genetically encoded Ca2+ indicators for investigation of microglial physiology.
Collapse
Affiliation(s)
- Amir Pozner
- Department of Human Genetics, University of Utah Salt Lake City, UT, USA ; Department of Chemistry, University of Utah Salt Lake City, UT, USA
| | - Ben Xu
- Department of Human Genetics, University of Utah Salt Lake City, UT, USA ; Howard Hughes Medical Institute Chevy Chase, MD, USA
| | - Sierra Palumbos
- Department of Human Genetics, University of Utah Salt Lake City, UT, USA
| | - J Michael Gee
- Department of Bioengineering, University of Utah Salt Lake City, UT, USA
| | - Petr Tvrdik
- Department of Human Genetics, University of Utah Salt Lake City, UT, USA
| | - Mario R Capecchi
- Department of Human Genetics, University of Utah Salt Lake City, UT, USA ; Howard Hughes Medical Institute Chevy Chase, MD, USA
| |
Collapse
|
26
|
Vázquez C, Tolón RM, Pazos MR, Moreno M, Koester EC, Cravatt BF, Hillard CJ, Romero J. Endocannabinoids regulate the activity of astrocytic hemichannels and the microglial response against an injury: In vivo studies. Neurobiol Dis 2015; 79:41-50. [PMID: 25917763 DOI: 10.1016/j.nbd.2015.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 04/02/2015] [Accepted: 04/19/2015] [Indexed: 10/23/2022] Open
Abstract
Anandamide (AEA) is an endocannabinoid (EC) that modulates multiple functions in the CNS and that is released in areas of injury, exerting putative neuroprotective actions. In the present study, we have used intravital microscopy to analyze the role of the EC system in the glial response against an acute insult. Our data show that AEA modulates astroglial function in vivo by increasing connexin-43 hemichannel (HC) activity. Furthermore, the genetic inactivation of the AEA-degrading enzyme, fatty acid amide hydrolase (FAAH), also increased HC activity and enhanced the microglial response against an acute injury to the brain parenchyma, effects that were mediated by cannabinoid CB1 receptors. The contribution of ATP released through an astrocytic HC was critical for the microglial response, as this was prevented by the use of the HC blocker flufenamic acid and by apyrase. As could be expected, brain concentrations of AEA, palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) were elevated in FAAH-null mice, while 2-arachidonoylglycerol (2-AG) concentrations remained unaltered. In summary, these findings demonstrate that AEA modifies glial functions by promoting an enhanced pro-inflammatory glial response in the brain.
Collapse
Affiliation(s)
- Carmen Vázquez
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Madrid, Spain
| | - Rosa María Tolón
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Madrid, Spain
| | - María Ruth Pazos
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Marta Moreno
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Madrid, Spain
| | - Erin C Koester
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Cecilia J Hillard
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Julián Romero
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, 28922 Alcorcón, Madrid, Spain; School of Biosciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain.
| |
Collapse
|
27
|
Burda JE, Bernstein AM, Sofroniew MV. Astrocyte roles in traumatic brain injury. Exp Neurol 2015; 275 Pt 3:305-315. [PMID: 25828533 DOI: 10.1016/j.expneurol.2015.03.020] [Citation(s) in RCA: 515] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 02/28/2015] [Accepted: 03/08/2015] [Indexed: 01/15/2023]
Abstract
Astrocytes sense changes in neural activity and extracellular space composition. In response, they exert homeostatic mechanisms critical for maintaining neural circuit function, such as buffering neurotransmitters, modulating extracellular osmolarity and calibrating neurovascular coupling. In addition to upholding normal brain activities, astrocytes respond to diverse forms of brain injury with heterogeneous and progressive changes of gene expression, morphology, proliferative capacity and function that are collectively referred to as reactive astrogliosis. Traumatic brain injury (TBI) sets in motion complex events in which noxious mechanical forces cause tissue damage and disrupt central nervous system (CNS) homeostasis, which in turn trigger diverse multi-cellular responses that evolve over time and can lead either to neural repair or secondary cellular injury. In response to TBI, astrocytes in different cellular microenvironments tune their reactivity to varying degrees of axonal injury, vascular disruption, ischemia and inflammation. Here we review different forms of TBI-induced astrocyte reactivity and the functional consequences of these responses for TBI pathobiology. Evidence regarding astrocyte contribution to post-traumatic tissue repair and synaptic remodeling is examined, and the potential for targeting specific aspects of astrogliosis to ameliorate TBI sequelae is considered.
Collapse
Affiliation(s)
- Joshua E Burda
- Department of Neurobiology and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095-1763, USA
| | - Alexander M Bernstein
- Department of Neurobiology and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095-1763, USA
| | - Michael V Sofroniew
- Department of Neurobiology and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095-1763, USA.
| |
Collapse
|
28
|
Schwarzmaier SM, Plesnila N. Contributions of the immune system to the pathophysiology of traumatic brain injury - evidence by intravital microscopy. Front Cell Neurosci 2014; 8:358. [PMID: 25408636 PMCID: PMC4219391 DOI: 10.3389/fncel.2014.00358] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) results in immediate brain damage that is caused by the mechanical impact and is non-reversible. This initiates a cascade of delayed processes which cause additional—secondary—brain damage. Among these secondary mechanisms, the inflammatory response is believed to play an important role, mediating actions that can have both protective and detrimental effects on the progression of secondary brain damage. Histological data generated extensive information; however, this is only a snapshot of processes that are, in fact, very dynamic. In contrast, in vivo microscopy provides detailed insight into the temporal and spatial patterns of cellular dynamics. In this review, we aim to summarize data which was generated by in vivo microscopy, specifically investigating the immune response following brain trauma, and its potential effects on secondary brain damage.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- Department of Anesthesiology, University of Munich Medical Center Munich, Germany ; Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University of Munich Medical Center Munich, Germany ; Munich Cluster of Systems Neurology Munich, Germany
| |
Collapse
|
29
|
Activation of neuronal NMDA receptors triggers transient ATP-mediated microglial process outgrowth. J Neurosci 2014; 34:10511-27. [PMID: 25100586 DOI: 10.1523/jneurosci.0405-14.2014] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Microglia are morphologically dynamic cells that rapidly extend their processes in response to various stimuli including extracellular ATP. In this study, we tested the hypothesis that stimulation of neuronal NMDARs trigger ATP release leading to communication with microglia. We used acute mouse hippocampal brain slices and two-photon laser scanning microscopy to study microglial dynamics and developed a novel protocol for fixation and immunolabeling of microglia processes. Similar to direct topical ATP application in vivo, short multiple applications of NMDA triggered transient microglia process outgrowth that was reversible and repeatable indicating that this was not due to excitotoxic damage. Stimulation of NMDAR was required as NMDAR antagonists, but not blockers of AMPA/kainate receptors or voltage-gated sodium channels, prevented microglial outgrowth. We report that ATP release, secondary to NMDAR activation, was the key mediator of this neuron-microglia communication as both blocking purinergic receptors and inhibiting hydrolysis of ATP to prevent locally generated gradients abolished outgrowth. Pharmacological and genetic analyses showed that the NMDA-triggered microglia process extension was independent of Pannexin 1, the ATP releasing channels, ATP release from astrocytes via connexins, and nitric oxide generation. Finally, using whole-cell patch clamping we demonstrate that activation of dendritic NMDAR on single neurons is sufficient to trigger microglia process outgrowth. Our results suggest that dendritic neuronal NMDAR activation triggers ATP release via a Pannexin 1-independent manner that induces outgrowth of microglia processes. This represents a novel uncharacterized form of neuron-microglial communication mediated by ATP.
Collapse
|
30
|
Salouci M, Antoine N, Shikh Al Sook MK, Piret J, Mignon Y, Kirschvink N, Gabriel A. Developmental profiles of GFAP-positive astrocytes in sheep cerebellum. Vet Res Commun 2014; 38:279-85. [PMID: 25113608 DOI: 10.1007/s11259-014-9614-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/05/2014] [Indexed: 11/27/2022]
Abstract
Astroglial account for the largest glial population in the brain and play a variety of vital functions in the development of the central nervous system (CNS). An immunohistochemical study was performed in 19 ovine foetuses ranging from 2 to 5 months of gestation, one newborn lamb and three adult sheep. Using the anit-glial fibrillary acidic protein (GFAP) marker, several variations were found in the degree of GFAP positive (GFAP+) astrocyte distribution between the different zones in the cerebellum of sheep during brain development. Our study indicates that the first appearance of astrocytes from restricted zones in the cerebellum occurs around the eighth week of gestation. Bergmann cells were found to be present from around the 15th week of gestation onwards. Our findings suggest that the maturation of astrocytes begins in the caudal parts of the cerebellum, developing from their initial ventral regions to spread first to dorsal regions radially within the white matter, then followed by the more rostral parts of the cerebellum. Astrocytes were also found to proliferate in the vermis before appearing in the cerebellar hemispheres.
Collapse
Affiliation(s)
- Moustafa Salouci
- Department of Morphology and Pathology, University of Liège, Boulevard de Colonster 20, 4000, Liège, Sart Tilman, Belgium,
| | | | | | | | | | | | | |
Collapse
|
31
|
De Bock M, Decrock E, Wang N, Bol M, Vinken M, Bultynck G, Leybaert L. The dual face of connexin-based astroglial Ca(2+) communication: a key player in brain physiology and a prime target in pathology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2211-32. [PMID: 24768716 DOI: 10.1016/j.bbamcr.2014.04.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 12/21/2022]
Abstract
For decades, studies have been focusing on the neuronal abnormalities that accompany neurodegenerative disorders. Yet, glial cells are emerging as important players in numerous neurological diseases. Astrocytes, the main type of glia in the central nervous system , form extensive networks that physically and functionally connect neuronal synapses with cerebral blood vessels. Normal brain functioning strictly depends on highly specialized cellular cross-talk between these different partners to which Ca(2+), as a signaling ion, largely contributes. Altered intracellular Ca(2+) levels are associated with neurodegenerative disorders and play a crucial role in the glial responses to injury. Intracellular Ca(2+) increases in single astrocytes can be propagated toward neighboring cells as intercellular Ca(2+) waves, thereby recruiting a larger group of cells. Intercellular Ca(2+) wave propagation depends on two, parallel, connexin (Cx) channel-based mechanisms: i) the diffusion of inositol 1,4,5-trisphosphate through gap junction channels that directly connect the cytoplasm of neighboring cells, and ii) the release of paracrine messengers such as glutamate and ATP through hemichannels ('half of a gap junction channel'). This review gives an overview of the current knowledge on Cx-mediated Ca(2+) communication among astrocytes as well as between astrocytes and other brain cell types in physiology and pathology, with a focus on the processes of neurodegeneration and reactive gliosis. Research on Cx-mediated astroglial Ca(2+) communication may ultimately shed light on the development of targeted therapies for neurodegenerative disorders in which astrocytes participate. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.
Collapse
Affiliation(s)
- Marijke De Bock
- Department of Basic Medical Sciences, Physiology group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Elke Decrock
- Department of Basic Medical Sciences, Physiology group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium.
| | - Nan Wang
- Department of Basic Medical Sciences, Physiology group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Mélissa Bol
- Department of Basic Medical Sciences, Physiology group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Mathieu Vinken
- Department of Toxicology, Center for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, B-1090 Brussels, Belgium
| | - Geert Bultynck
- Department of Cellular and Molecular Medicine, Laboratory of Molecular and Cellular Signalling, KULeuven, Campus Gasthuisberg O/N-I bus 802, B-3000 Leuven, Belgium
| | - Luc Leybaert
- Department of Basic Medical Sciences, Physiology group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
32
|
Morris G, Maes M. Mitochondrial dysfunctions in myalgic encephalomyelitis/chronic fatigue syndrome explained by activated immuno-inflammatory, oxidative and nitrosative stress pathways. Metab Brain Dis 2014; 29:19-36. [PMID: 24557875 DOI: 10.1007/s11011-013-9435-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 08/22/2013] [Indexed: 02/07/2023]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/cfs) is classified by the World Health Organization as a disorder of the central nervous system. ME/cfs is an neuro-immune disorder accompanied by chronic low-grade inflammation, increased levels of oxidative and nitrosative stress (O&NS), O&NS-mediated damage to fatty acids, DNA and proteins, autoimmune reactions directed against neoantigens and brain disorders. Mitochondrial dysfunctions have been found in ME/cfs, e.g. lowered ATP production, impaired oxidative phosphorylation and mitochondrial damage. This paper reviews the pathways that may explain mitochondrial dysfunctions in ME/cfs. Increased levels of pro-inflammatory cytokines, such as interleukin-1 and tumor necrosis factor-α, and elastase, and increased O&NS may inhibit mitochondrial respiration, decrease the activities of the electron transport chain and mitochondrial membrane potential, increase mitochondrial membrane permeability, interfere with ATP production and cause mitochondrial shutdown. The activated O&NS pathways may additionally lead to damage of mitochondrial DNA and membranes thus decreasing membrane fluidity. Lowered levels of antioxidants, zinc and coenzyme Q10, and ω3 polyunsaturated fatty acids in ME/cfs may further aggravate the activated immuno-inflammatory and O&NS pathways. Therefore, it may be concluded that immuno-inflammatory and O&NS pathways may play a role in the mitochondrial dysfunctions and consequently the bioenergetic abnormalities seen in patients with ME/cfs. Defects in ATP production and the electron transport complex, in turn, are associated with an elevated production of superoxide and hydrogen peroxide in mitochondria creating adaptive and synergistic damage. It is argued that mitochondrial dysfunctions, e.g. lowered ATP production, may play a role in the onset of ME/cfs symptoms, e.g. fatigue and post exertional malaise, and may explain in part the central metabolic abnormalities observed in ME/cfs, e.g. glucose hypometabolism and cerebral hypoperfusion.
Collapse
|
33
|
Fenrich KK, Weber P, Rougon G, Debarbieux F. Implanting glass spinal cord windows in adult mice with experimental autoimmune encephalomyelitis. J Vis Exp 2013:e50826. [PMID: 24378439 DOI: 10.3791/50826] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) in adult rodents is the standard experimental model for studying autonomic demyelinating diseases such as multiple sclerosis. Here we present a low-cost and reproducible glass window implantation protocol that is suitable for intravital microscopy and studying the dynamics of spinal cord cytoarchitecture with subcellular resolution in live adult mice with EAE. Briefly, we surgically expose the vertebrae T12-L2 and construct a chamber around the exposed vertebrae using a combination of cyanoacrylate and dental cement. A laminectomy is performed from T13 to L1, and a thin layer of transparent silicone elastomer is applied to the dorsal surface of the exposed spinal cord. A modified glass cover slip is implanted over the exposed spinal cord taking care that the glass does not directly contact the spinal cord. To reduce the infiltration of inflammatory cells between the window and spinal cord, anti-inflammatory treatment is administered every 2 days (as recommended by ethics committee) for the first 10 days after implantation. EAE is induced only 2-3 weeks after the cessation of anti-inflammatory treatment. Using this approach we successfully induced EAE in 87% of animals with implanted windows and, using Thy1-CFP-23 mice (blue axons in dorsal spinal cord), quantified axonal loss throughout EAE progression. Taken together, this protocol may be useful for studying the recruitment of various cell populations as well as their interaction dynamics, with subcellular resolution and for extended periods of time. This intravital imaging modality represents a valuable tool for developing therapeutic strategies to treat autoimmune demyelinating diseases such as EAE.
Collapse
Affiliation(s)
- Keith K Fenrich
- Developmental Biology Institute of Marseille-Luminy (IBDML), Aix Marseille University
| | | | | | | |
Collapse
|
34
|
Pascual JL, Murcy MA, Li S, Gong W, Eisenstadt R, Kumasaka K, Sims C, Smith DH, Browne K, Allen S, Baren J. Neuroprotective effects of progesterone in traumatic brain injury: blunted in vivo neutrophil activation at the blood-brain barrier. Am J Surg 2013; 206:840-5; discussion 845-6. [PMID: 24112683 DOI: 10.1016/j.amjsurg.2013.07.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 07/23/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Progesterone (PRO) may confer a survival advantage in traumatic brain injury (TBI) by reducing cerebral edema. We hypothesized that PRO reduces edema by blocking polymorphonuclear (PMN) interactions with endothelium (EC) in the blood-brain barrier (BBB). METHODS CD1 mice received repeated PRO (16 mg/kg intraperitoneally) or vehicle (cyclodextrin) for 36 hours after TBI. Sham animals underwent craniotomy without TBI. The modified Neurological Severity Score graded neurologic recovery. A second craniotomy allowed in vivo observation of pial EC/PMN interactions and vascular macromolecule leakage. Wet/dry ratios assessed cerebral edema. RESULTS Compared with the vehicle, PRO reduced subjective cerebral swelling (2.9 ± .1 vs 1.2 ± .1, P < .001), PMN rolling (95 ± 1.8 vs 57 ± 2.0 cells/100 μm/min, P < .001), total EC/PMN adhesion (2.0 ± .4 vs .8 ± .1 PMN/100 μm, P < .01), and vascular permeability (51.8% ± 4.9% vs 27.1% ± 4.6%, P < .01). TBI groups had similar a Neurological Severity Score and cerebral wet/dry ratios (P > .05). CONCLUSIONS PRO reduces live pericontusional EC/PMN and BBB macromolecular leakage after TBI. Direct PRO effects on the microcirculation warrant further investigation.
Collapse
Affiliation(s)
- Jose L Pascual
- Department of Surgery, Division of Traumatology, Surgical Critical Care and Emergency Surgery, Perelman School of Medicine at the University of Pennsylvania, The Trauma Center at Penn, 3400 Spruce Street, Maloney Building, 5th Floor, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Wong AD, Ye M, Levy AF, Rothstein JD, Bergles DE, Searson PC. The blood-brain barrier: an engineering perspective. FRONTIERS IN NEUROENGINEERING 2013; 6:7. [PMID: 24009582 PMCID: PMC3757302 DOI: 10.3389/fneng.2013.00007] [Citation(s) in RCA: 403] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 08/07/2013] [Indexed: 12/03/2022]
Abstract
It has been more than 100 years since Paul Ehrlich reported that various water-soluble dyes injected into the circulation did not enter the brain. Since Ehrlich's first experiments, only a small number of molecules, such as alcohol and caffeine have been found to cross the blood-brain barrier, and this selective permeability remains the major roadblock to treatment of many central nervous system diseases. At the same time, many central nervous system diseases are associated with disruption of the blood-brain barrier that can lead to changes in permeability, modulation of immune cell transport, and trafficking of pathogens into the brain. Therefore, advances in our understanding of the structure and function of the blood-brain barrier are key to developing effective treatments for a wide range of central nervous system diseases. Over the past 10 years it has become recognized that the blood-brain barrier is a complex, dynamic system that involves biomechanical and biochemical signaling between the vascular system and the brain. Here we reconstruct the structure, function, and transport properties of the blood-brain barrier from an engineering perspective. New insight into the physics of the blood-brain barrier could ultimately lead to clinical advances in the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Andrew D. Wong
- Department of Materials Science and Engineering, Johns Hopkins UniversityBaltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins UniversityBaltimore, MD, USA
| | - Mao Ye
- Department of Materials Science and Engineering, Johns Hopkins UniversityBaltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins UniversityBaltimore, MD, USA
| | - Amanda F. Levy
- Department of Materials Science and Engineering, Johns Hopkins UniversityBaltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins UniversityBaltimore, MD, USA
| | - Jeffrey D. Rothstein
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimore, MD, USA
- Brain Sciences Institute, Johns Hopkins UniversityBaltimore, MD, USA
| | - Dwight E. Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimore, MD, USA
| | - Peter C. Searson
- Department of Materials Science and Engineering, Johns Hopkins UniversityBaltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins UniversityBaltimore, MD, USA
| |
Collapse
|
36
|
Brawek B, Garaschuk O. Microglial calcium signaling in the adult, aged and diseased brain. Cell Calcium 2013; 53:159-69. [PMID: 23395344 DOI: 10.1016/j.ceca.2012.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/10/2012] [Indexed: 10/27/2022]
Abstract
Microglial cells are the resident immune cells of the CNS. They mediate innate immune response of the brain to injury, inflammation and neurodegenerative diseases. Apart from their role in disease they are critically involved in the development and plasticity-driven reorganization of neuronal networks and the homeostatic maintenance of brain tissue. Accumulating in vitro evidence suggests that executive functions of microglia are coupled to the intracellular Ca(2+) signaling of these cells. So far, however, very little is known about microglial Ca(2+) signaling in situ or in vivo, both in the healthy and in the diseased brain. Here, we summarize the recent in vivo/in situ findings and compare the properties of surveillant microglia in these preparations with those of microglia in vitro. The data suggest that surveillant microglia rarely show spontaneous Ca(2+) transients, express fewer functional receptors directly coupled to changes in the intracellular free Ca(2+) concentration on their surface, but vividly respond with Ca(2+) transients to cell or tissue damage in their microenvironment. Interestingly, some of these properties microglia share with monocytes engrafting in the brain under pathological conditions.
Collapse
Affiliation(s)
- Bianca Brawek
- Institute of Physiology II, Eberhard Karls University of Tuebingen, Keplerstr. 15, 72074 Tuebingen, Germany
| | | |
Collapse
|
37
|
Eyo UB, Dailey ME. Microglia: key elements in neural development, plasticity, and pathology. J Neuroimmune Pharmacol 2013; 8:494-509. [PMID: 23354784 DOI: 10.1007/s11481-013-9434-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 01/14/2013] [Indexed: 12/31/2022]
Abstract
A century after Cajal identified a "third element" of the nervous system, many issues have been clarified about the identity and function of one of its major components, the microglia. Here, we review recent findings by microgliologists, highlighting results from imaging studies that are helping provide new views of microglial behavior and function. In vivo imaging in the intact adult rodent CNS has revolutionized our understanding of microglial behaviors in situ and has raised speculation about their function in the uninjured adult brain. Imaging studies in ex vivo mammalian tissue preparations and in intact model organisms including zebrafish are providing insights into microglial behaviors during brain development. These data suggest that microglia play important developmental roles in synapse remodeling, developmental apoptosis, phagocytic clearance, and angiogenesis. Because microglia also contribute to pathology, including neurodevelopmental and neurobehavioral disorders, ischemic injury, and neuropathic pain, promising new results raise the possibility of leveraging microglia for therapeutic roles. Finally, exciting recent work is addressing unanswered questions regarding the nature of microglial-neuronal communication. While it is now apparent that microglia play diverse roles in neural development, behavior, and pathology, future research using neuroimaging techniques will be essential to more fully exploit these intriguing cellular targets for effective therapeutic intervention applied to a variety of conditions.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
38
|
Lin Y, Wen L. Inflammatory response following diffuse axonal injury. Int J Med Sci 2013; 10:515-21. [PMID: 23532682 PMCID: PMC3607236 DOI: 10.7150/ijms.5423] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 03/06/2013] [Indexed: 12/12/2022] Open
Abstract
DAI is a leading cause of the patient's death or lasting vegetable state following severe TBI, and up to now the detailed mechanism of axonal injury after head trauma is still unclear. Inflammatory responses have been proved to be an important mechanism of neural injury after TBI. However, most of these studies are concerned with focal cerebral injury following head trauma. In contrast to focal injury, studies on the inflammatory reaction following DAI are only beginning. And in this article, we aimed to review such studies. From the studies reviewed, immune response cells would become reactive around the sites of axonal injury after DAI. Besides, the concentrations of several important inflammatory factors, such as IL-1 family, IL-6 and TNF-ɑ, increased after DAI as well, which implies the participation of inflammatory responses. It can be concluded that inflammatory responses probably participate in the neural injury in DAI, but at present the study of inflammatory responses following DAI is still limited and the clear effects of inflammatory response on axonal injury remain to be more explored.
Collapse
Affiliation(s)
- Yu Lin
- School of Medicine, Zhejiang University City College, China
| | | |
Collapse
|
39
|
ROCK/Cdc42-mediated microglial motility and gliapse formation lead to phagocytosis of degenerating dopaminergic neurons in vivo. Sci Rep 2012; 2:809. [PMID: 23139861 PMCID: PMC3492875 DOI: 10.1038/srep00809] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/18/2012] [Indexed: 01/17/2023] Open
Abstract
The role of microglial motility in the context of adult neurodegeneration is poorly understood. In the present work, we investigated the microanatomical details of microglia-neuron interactions in an experimental mouse model of Parkinson's disease following the intraperitoneal injection of MPTP. The specific intoxication of dopaminergic neurons induces the cellular polarization of microglia, leading to the formation of body-to-body neuron-glia contacts, called gliapses, which precede neuron elimination. Inhibiting ROCK/Cdc42-mediated microglial motility in vivo blocks the activating features of microglia, such as increased cell size and number of filopodia and diminishes their phagocyting/secreting domains, as the reduction of the Golgi apparatus and the number of microglia-neuron contacts has shown. High-resolution confocal images and three-dimensional rendering demonstrate that microglia engulf entire neurons at one-to-one ratio, and the microglial cell body participates in the formation of the phagocytic cup, engulfing and eliminating neurons in areas of dopaminergic degeneration in adult mammals.
Collapse
|
40
|
Targeted ablation of oligodendrocytes induces axonal pathology independent of overt demyelination. J Neurosci 2012; 32:8317-30. [PMID: 22699912 DOI: 10.1523/jneurosci.1053-12.2012] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The critical role of oligodendrocytes in producing and maintaining myelin that supports rapid axonal conduction in CNS neurons is well established. More recently, additional roles for oligodendrocytes have been posited, including provision of trophic factors and metabolic support for neurons. To investigate the functional consequences of oligodendrocyte loss, we have generated a transgenic mouse model of conditional oligodendrocyte ablation. In this model, oligodendrocytes are rendered selectively sensitive to exogenously administered diphtheria toxin (DT) by targeted expression of the diphtheria toxin receptor in oligodendrocytes. Administration of DT resulted in severe clinical dysfunction with an ascending spastic paralysis ultimately resulting in fatal respiratory impairment within 22 d of DT challenge. Pathologically, at this time point, mice exhibited a loss of ∼26% of oligodendrocyte cell bodies throughout the CNS. Oligodendrocyte cell-body loss was associated with moderate microglial activation, but no widespread myelin degradation. These changes were accompanied with acute axonal injury as characterized by structural and biochemical alterations at nodes of Ranvier and reduced somatosensory-evoked potentials. In summary, we have shown that a death signal initiated within oligodendrocytes results in subcellular changes and loss of key symbiotic interactions between the oligodendrocyte and the axons it ensheaths. This produces profound functional consequences that occur before the removal of the myelin membrane, i.e., in the absence of demyelination. These findings have clear implications for the understanding of the pathogenesis of diseases of the CNS such as multiple sclerosis in which the oligodendrocyte is potentially targeted.
Collapse
|
41
|
Lu YM, Tao RR, Huang JY, Li LT, Liao MH, Li XM, Fukunaga K, Hong ZH, Han F. P2X7 signaling promotes microsphere embolism-triggered microglia activation by maintaining elevation of Fas ligand. J Neuroinflammation 2012; 9:172. [PMID: 22789015 PMCID: PMC3420259 DOI: 10.1186/1742-2094-9-172] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/20/2012] [Indexed: 11/30/2022] Open
Abstract
Background The cerebral microvascular occlusion elicits microvascular injury which mimics the different degrees of stroke severity observed in patients, but the mechanisms underlying these embolic injuries are far from understood. The Fas ligand (FasL)-Fas system has been implicated in a number of pathogenic states. Here, we examined the contribution of microglia-derived FasL to brain inflammatory injury, with a focus on the potential to suppress the FasL increase by inhibition of the P2X7-FasL signaling with pharmacological or genetic approaches during ischemia. Methods The cerebral microvascular occlusion was induced by microsphere injection in experimental animals. Morphological changes in microglial cells were studied immunohistochemically. The biochemical analyses were used to examine the intracellular changes of P2X7/FasL signaling. The BV-2 cells and primary microglia from mice genetically deficient in P2X7 were used to further establish a linkage between microglia activation and FasL overproduction. Results The FasL expression was continuously elevated and was spatiotemporally related to microglia activation following microsphere embolism. Notably, P2X7 expression concomitantly increased in microglia and presented a distribution pattern that was similar to that of FasL in ED1-positive cells at pathological process of microsphere embolism. Interestingly, FasL generation in cultured microglia cells subjected to oxygen-glucose deprivation-treated neuron-conditioned medium was prevented by the silencing of P2X7. Furthermore, FasL induced the migration of BV-2 microglia, whereas the neutralization of FasL with a blocking antibody was highly effective in inhibiting ischemia-induced microglial mobility. Similar results were observed in primary microglia from wild-type mice or mice genetically deficient in P2X7. Finally, the degrees of FasL overproduction and neuronal death were consistently reduced in P2X7−/− mice compared with wild-type littermates following microsphere embolism insult. Conclusion FasL functions as a key component of an immunoreactive response loop by recruiting microglia to the lesion sites through a P2X7-dependent mechanism. The specific modulation of P2X7/FasL signaling and aberrant microglial activation could provide therapeutic benefits in acute and subacute phase of cerebral microembolic injury.
Collapse
Affiliation(s)
- Ying-mei Lu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ribeiro-Gomes FL, Sacks D. The influence of early neutrophil-Leishmania interactions on the host immune response to infection. Front Cell Infect Microbiol 2012; 2:59. [PMID: 22919650 PMCID: PMC3417510 DOI: 10.3389/fcimb.2012.00059] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 04/16/2012] [Indexed: 11/13/2022] Open
Abstract
Neutrophils are the first cells recruited to the dermal site of Leishmania infection following injection by needle or sand fly bite. The role of neutrophils in either promoting or suppressing host immunity remains controversial. We discuss the events driving neutrophil recruitment, their interaction with the parasite and apoptotic fate, and the nature of their encounters with other innate cells. We suggest that the influence of the neutrophil response on infection outcome critically depends on the timing of their recruitment and the tissue environment in which it occurs.
Collapse
Affiliation(s)
- Flavia L Ribeiro-Gomes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD, USA
| | | |
Collapse
|
43
|
Nimmerjahn A. Optical window preparation for two-photon imaging of microglia in mice. Cold Spring Harb Protoc 2012; 2012:2012/5/pdb.prot069286. [PMID: 22550298 DOI: 10.1101/pdb.prot069286] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Microglia are the primary immune effector cells of the brain parenchyma. They are distributed throughout the brain at various densities. Two-photon fluorescence microscopy, together with expression of fluorescent proteins in microglia, has enabled the study of these fascinating cells in vivo. Imaging studies have shown, for example, that microglia continually survey their cellular environment and immediately respond to injury. However, we still know very little about their roles in various parts of the developing and adult brain or their diverse effector functions in aging and different disease states. Experimental procedures have been developed for minimally invasive short- and long-term two-photon imaging of microglial cells in cortical regions of the intact mouse brain. This protocol presents two methods for the preparation of the optical window that is needed for two-photon imaging of microglia. The thinned skull method should be used whenever possible. Skull thinning enables transcranial two-photon imaging while minimizing external influences that might disturb normal microglia physiology and brain homeostasis. The sealed craniotomy preparation is useful for short-term investigation of microglia.
Collapse
|
44
|
Nimmerjahn A. Two-photon imaging of microglia in the mouse cortex in vivo. Cold Spring Harb Protoc 2012; 2012:2012/5/pdb.prot069294. [PMID: 22550299 DOI: 10.1101/pdb.prot069294] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Microglia are the primary immune effector cells of the brain parenchyma. They are distributed throughout the brain at various densities. Two-photon fluorescence microscopy, together with expression of fluorescent proteins in microglia, has enabled study of these fascinating cells in vivo. Imaging studies have shown, for example, that microglia continually survey their cellular environment and immediately respond to injury. However, we still know very little about their roles in various parts of the developing and adult brain or their diverse effector functions in aging and different disease states. Experimental procedures have been developed for minimally invasive short- and long-term two-photon imaging of microglial cells in cortical regions of the intact mouse brain. This protocol describes two-photon imaging of microglia in the mouse cortex in vivo, using mice which have had a head plate implanted and have been prepared with either a thinned skull or optical window. Technical pitfalls, limitations, and alternative approaches are also discussed.
Collapse
|
45
|
Williams WM, Castellani RJ, Weinberg A, Perry G, Smith MA. Do β-defensins and other antimicrobial peptides play a role in neuroimmune function and neurodegeneration? ScientificWorldJournal 2012; 2012:905785. [PMID: 22606066 PMCID: PMC3346844 DOI: 10.1100/2012/905785] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 10/26/2011] [Indexed: 12/25/2022] Open
Abstract
It is widely accepted that the brain responds to mechanical trauma and development of most neurodegenerative diseases with an inflammatory sequelae that was once thought exclusive to systemic immunity. Mostly cationic peptides, such as the β-defensins, originally assigned an antimicrobial function are now recognized as mediators of both innate and adaptive immunity. Herein supporting evidence is presented for the hypothesis that neuropathological changes associated with chronic disease conditions of the CNS involve abnormal expression and regulatory function of specific antimicrobial peptides. It is also proposed that these alterations exacerbate proinflammatory conditions within the brain that ultimately potentiate the neurodegenerative process.
Collapse
Affiliation(s)
- Wesley M Williams
- Department of Biological Sciences, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | | | | | |
Collapse
|
46
|
Li JL, Goh CC, Keeble JL, Qin JS, Roediger B, Jain R, Wang Y, Chew WK, Weninger W, Ng LG. Intravital multiphoton imaging of immune responses in the mouse ear skin. Nat Protoc 2012; 7:221-34. [PMID: 22240584 DOI: 10.1038/nprot.2011.438] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Multiphoton (MP) microscopy enables the direct in vivo visualization, with high spatial and temporal resolution, of fluorescently tagged immune cells, extracellular matrix and vasculature in tissues. This approach, therefore, represents a powerful alternative to traditional methods of assessing immune cell function in the skin, which are mainly based on flow cytometry and histology. Here we provide a step-by-step protocol describing experimental procedures for intravital MP imaging of the mouse ear skin, which can be easily adapted to address many specific skin-related biological questions. We demonstrate the use of this procedure by characterizing the response of neutrophils during cutaneous inflammation, which can be used to perform in-depth analysis of neutrophil behavior in the context of the skin microanatomy, including the epidermis, dermis and blood vessels. Such experiments are typically completed within 1 d, but as the procedures are minimally invasive, it is possible to perform longitudinal studies through repeated imaging.
Collapse
Affiliation(s)
- Jackson LiangYao Li
- Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Microglia were recently shown to play unexpected roles in normal brain development and adult physiology. This has begun to dramatically change our view of these resident "immune" cells. Here, we briefly review topics covered in our 2011 Society for Neuroscience minisymposium "The Role of Microglia in the Healthy Brain." This summary is not meant to be a comprehensive review of microglia physiology, but rather to share new results and stimulate further research into the cellular and molecular mechanisms by which microglia influence postnatal development, adult neuronal plasticity, and circuit function.
Collapse
|
48
|
Rosidi NL, Zhou J, Pattanaik S, Wang P, Jin W, Brophy M, Olbricht WL, Nishimura N, Schaffer CB. Cortical microhemorrhages cause local inflammation but do not trigger widespread dendrite degeneration. PLoS One 2011; 6:e26612. [PMID: 22028924 PMCID: PMC3197572 DOI: 10.1371/journal.pone.0026612] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 09/29/2011] [Indexed: 11/18/2022] Open
Abstract
Microhemorrhages are common in the aging brain, and their incidence is correlated with increased risk of neurodegenerative disease. Past work has shown that occlusion of individual cortical microvessels as well as large-scale hemorrhages can lead to degeneration of neurons and increased inflammation. Using two-photon excited fluorescence microscopy in anesthetized mice, we characterized the acute and chronic dynamics of vessel bleeding, tissue compression, blood flow change, neural degeneration, and inflammation following a microhemorrhage caused by rupturing a single penetrating arteriole with tightly-focused femtosecond laser pulses. We quantified the extravasation of red blood cells (RBCs) and blood plasma into the brain and determined that the bleeding was limited by clotting. The vascular bleeding formed a RBC-filled core that compressed the surrounding parenchymal tissue, but this compression was not sufficient to crush nearby brain capillaries, although blood flow speeds in these vessels was reduced by 20%. Imaging of cortical dendrites revealed no degeneration of the large-scale structure of the dendritic arbor up to 14 days after the microhemorrhage. Dendrites close to the RBC core were displaced by extravasating RBCs but began to relax back one day after the lesion. Finally, we observed a rapid inflammatory response characterized by morphology changes in microglia/macrophages up to 200 µm from the microhemorrhage as well as extension of cellular processes into the RBC core. This inflammation persisted over seven days. Taken together, our data suggest that a cortical microhemorrhage does not directly cause significant neural pathology but does trigger a sustained, local inflammatory response.
Collapse
Affiliation(s)
- Nathanael L. Rosidi
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Joan Zhou
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Sanket Pattanaik
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Peng Wang
- Department of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, United States of America
| | - Weiyang Jin
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Morgan Brophy
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - William L. Olbricht
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
- Department of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, United States of America
| | - Nozomi Nishimura
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Chris B. Schaffer
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
49
|
Abstract
Viral infections are a major cause of human disease. Although most viruses replicate in peripheral tissues, some have developed unique strategies to move into the nervous system, where they establish acute or persistent infections. Viral infections in the central nervous system (CNS) can alter homeostasis, induce neurological dysfunction and result in serious, potentially life-threatening inflammatory diseases. This Review focuses on the strategies used by neurotropic viruses to cross the barrier systems of the CNS and on how the immune system detects and responds to viral infections in the CNS. A special emphasis is placed on immune surveillance of persistent and latent viral infections and on recent insights gained from imaging both protective and pathogenic antiviral immune responses.
Collapse
|
50
|
Vojdani A, Lambert J. The Role of Th17 in Neuroimmune Disorders: Target for CAM Therapy. Part II. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2011:984965. [PMID: 19622601 PMCID: PMC3137879 DOI: 10.1093/ecam/nep063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 05/22/2009] [Indexed: 12/23/2022]
Abstract
Decades of research went into understanding the role that Th1 autoreactive T-cells play in neuroinflammation. Here we describe another effector population, the IL-17-producing T-helper lineage (Th17), which drives the inflammatory process. Through the recruitment of inflammatory infiltration neutrophils and the activation of matrix metalloproteinases, IL-17, a cytokine secreted by Th17 cells, contributes to blood-brain barrier breakdown and the subsequent attraction of macrophages and monocytes into the nervous system. The entry of cells along with the local production of inflammatory cytokines leads to myelin and axonal damage. This activation of the inflammatory response system is induced by different pathogenic factors, such as gut bacterial endotoxins resulting in progressive neurodegeneration by Th17 cells. Through the understanding of the role of bacterial endotoxins and other pathogenic factors in the induction of autoimmune diseases by Th17 cells, CAM practitioners will be able to design CAM therapies targeting IL-17 activity. Targeted therapy can restore the integrity of the intestinal and blood-brain barriers using probiotics, N-acetyl-cysteine, α-lipoic acid, resveratrol and others for their patients with autoimmunities, in particular those with neuroinflammation and neurodegeneration.
Collapse
|