1
|
Wang P, Li J, Li S, Liu Y, Gong J, He S, Wu W, Tan G, Liu S. Palladium-reduced graphene oxide nanocomposites enhance neurite outgrowth and protect neurons from Ishemic stroke. Mater Today Bio 2024; 28:101184. [PMID: 39221214 PMCID: PMC11364903 DOI: 10.1016/j.mtbio.2024.101184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/09/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024] Open
Abstract
Currently, the construction of novel biomimetic reduced graphene oxide (RGO)-based nanocomposites to induce neurite sprouting and repair the injured neurons represents a promising strategy in promoting neuronal development or treatment of cerebral anoxia or ischemia. Here, we present an effective method for constructing palladium-reduced graphene oxide (Pd-RGO) nanocomposites by covalently bonding Pd onto RGO surfaces to enhance neurite sprouting of cultured neurons. As described, the Pd-RGO nanocomposites exhibit the required physicochemical features for better biocompatibility without impacting cell viability. Primary neurons cultured on Pd-RGO nanocomposites had significantly increased number and length of neuronal processes, including both axons and dendrites, compared with the control. Western blotting showed that Pd-RGO nanocomposites improved the expression levels of growth associate protein-43 (GAP-43), as well as β-III tubulin, Tau-1, microtubule-associated protein-2 (MAP2), four proteins that are involved in regulating neurite sprouting and outgrowth. Importantly, Pd-RGO significantly promoted neurite length and complexity under oxygen-glucose deprivation/re-oxygenation (OGD/R) conditions, an in vitro cellular model of ischemic brain damage, that closely relates to neuronal GAP-43 expression. Furthermore, using the middle cerebral artery occlusion (MCAO) model in rats, we found Pd-RGO effectively reduced the infarct area, decreased neuronal apoptosis in the brain, and improved the rats' behavioral outcomes after MCAO. Together, these results indicate the great potential of Pd-RGO nanocomposites as a novel excellent biomimetic material for neural interfacing that shed light on its applications in brain injuries.
Collapse
Affiliation(s)
- Ping Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuntang Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanyuan Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiangu Gong
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shipei He
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Weifeng Wu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guohe Tan
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Pan YW, Wu DP, Liang HF, Tang GY, Fan CL, Shi L, Ye WC, Li MM. Total Saponins of Panax notoginseng Activate Akt/mTOR Pathway and Exhibit Neuroprotection in vitro and in vivo against Ischemic Damage. Chin J Integr Med 2021; 28:410-418. [PMID: 34581940 DOI: 10.1007/s11655-021-3454-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To reveal the neuroprotective effect and the underlying mechanisms of a mixture of the main components of Panax notoginseng saponins (TSPN) on cerebral ischemia-reperfusion injury and oxygen-glucose deprivation/reoxygenation (OGD/R) of cultured cortical neurons. METHODS The neuroprotective effect of TSPN was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, flow cytometry and live/dead cell assays. The morphology of dendrites was detected by immunofluorescence. Middle cerebral artery occlusion (MCAO) was developed in rats as a model of cerebral ischemia-reperfusion. The neuroprotective effect of TSPN was evaluated by neurological scoring, tail suspension test, 2,3,5-triphenyltetrazolium chloride (TTC) and Nissl stainings. Western blot analysis, immunohistochemistry and immunofluorescence were used to measure the changes in the Akt/mammalian target of rapamycin (mTOR) signaling pathway. RESULTS MTT showed that TSPN (50, 25 and 12.5 µ g/mL) protected cortical neurons after OGD/R treatment (P<0.01 or P<0.05). Flow cytometry and live/dead cell assays indicated that 25 µ g/mL TSPN decreased neuronal apoptosis (P<0.05), and immunofluorescence showed that 25 µ g/mL TSPN restored the dendritic morphology of damaged neurons (P<0.05). Moreover, 12.5 µ g/mL TSPN downregulated the expression of Beclin-1, Cleaved-caspase 3 and LC3B-II/LC3B-I, and upregulated the levels of phosphorylated (p)-Akt and p-mTOR (P<0.01 or P<0.05). In the MCAO model, 50 µ g/mL TSPN improved defective neurological behavior and reduced infarct volume (P<0.05). Moreover, the expression of Beclin-1 and LC3B in cerebral ischemic penumbra was downregulated after 50 µ g/mL TSPN treatment, whereas the p-mTOR level was upregulated (P<0.05 or P<0.01). CONCLUSION TSPN promoted neuronal survival and protected dendrite integrity after OGD/R and had a potential therapeutic effect by alleviating neurological deficits and reversing neuronal loss. TSPN promoted p-mTOR and inhibited Beclin-1 to alleviate ischemic damage, which may be the mechanism that underlies the neuroprotective activity of TSPN.
Collapse
Affiliation(s)
- Yu-Wei Pan
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, 510632, China.,Department of TCM Preventive Medicine, Tianhe District Hospital of Traditional Chinese Medicine, Guangzhou, 510632, China
| | - Dong-Ping Wu
- Institute of Traditional Chinese Medicine and Natural Products, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Hua-Feng Liang
- Institute of Traditional Chinese Medicine and Natural Products, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Gen-Yun Tang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, 510632, China
| | - Chun-Lin Fan
- Institute of Traditional Chinese Medicine and Natural Products, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, 510632, China
| | - Wen-Cai Ye
- Institute of Traditional Chinese Medicine and Natural Products, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Man-Mei Li
- Institute of Traditional Chinese Medicine and Natural Products, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
3
|
Growth Hormone (GH) Enhances Endogenous Mechanisms of Neuroprotection and Neuroplasticity after Oxygen and Glucose Deprivation Injury (OGD) and Reoxygenation (OGD/R) in Chicken Hippocampal Cell Cultures. Neural Plast 2021; 2021:9990166. [PMID: 34567109 PMCID: PMC8461227 DOI: 10.1155/2021/9990166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/14/2021] [Indexed: 11/18/2022] Open
Abstract
As a classical growth promoter and metabolic regulator, growth hormone (GH) is involved in development of the central nervous system (CNS). This hormone might also act as a neurotrophin, since GH is able to induce neuroprotection, neurite growth, and synaptogenesis during the repair process that occurs in response to neural injury. After an ischemic insult, the neural tissue activates endogenous neuroprotective mechanisms regulated by local neurotrophins that promote tissue recovery. In this work, we investigated the neuroprotective effects of GH in cultured hippocampal neurons exposed to hypoxia-ischemia injury and further reoxygenation. Hippocampal cell cultures obtained from chick embryos were incubated under oxygen-glucose deprivation (OGD, <5% O2, 1 g/L glucose) conditions for 24 h and simultaneously treated with GH. Then, cells were either collected for analysis or submitted to reoxygenation and normal glucose incubation conditions (OGD/R) for another 24 h, in the presence of GH. Results showed that OGD injury significantly reduced cell survival, the number of cells, dendritic length, and number of neurites, whereas OGD/R stage restored most of those adverse effects. Also, OGD/R increased the mRNA expression of several synaptogenic markers (i.e., NRXN1, NRXN3, NLG1, and GAP43), as well as the growth hormone receptor (GHR). The expression of BDNF, IGF-1, and BMP4 mRNAs was augmented in response to OGD injury, and exposure to OGD/R returned it to normoxic control levels, while the expression of NT-3 increased in both conditions. The addition of GH (10 nM) to hippocampal cultures during OGD reduced apoptosis and induced a significant increase in cell survival, number of cells, and doublecortin immunoreactivity (DCX-IR), above that observed in the OGD/R stage. GH treatment also protected dendrites and neurites during OGD, inducing plastic changes reflected in an increase and complexity of their outgrowths during OGD/R. Furthermore, GH increased the expression of NRXN1, NRXN3, NLG1, and GAP43 after OGD injury. GH also increased the BDNF expression after OGD, but reduced it after OGD/R. Conversely, BMP4 was upregulated by GH after OGD/R. Overall, these results indicate that GH protective actions in the neural tissue may be explained by a synergic combination between its own effect and that of other local neurotrophins regulated by autocrine/paracrine mechanisms, which together accelerate the recovery of tissue damaged by hypoxia-ischemia.
Collapse
|
4
|
Roque C, Pinto N, Vaz Patto M, Baltazar G. Astrocytes contribute to the neuronal recovery promoted by high-frequency repetitive magnetic stimulation in in vitro models of ischemia. J Neurosci Res 2021; 99:1414-1432. [PMID: 33522025 DOI: 10.1002/jnr.24792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 11/07/2022]
Abstract
After decades of effort, there are no effective clinical treatments to induce the recovery of ischemia-injured tissues, and among the several strategies that have been explored, repetitive transcranial magnetic stimulation has proven to be one of the most promising, with beneficial effects in limb motor function, aphasia, hemispatial neglect, or dysphagia. Despite the clinical evidences, little is known about the mechanisms underlying those effects. The present study aimed to explore the cellular and molecular effects of high-frequency repetitive magnetic stimulation (HF-rMS) on an in vitro model of ischemia. Using primary cortical cultures exposed to oxygen and glucose deprivation followed by reperfusion, we observed that HF-rMS treatment prevents the ischemia-induced neuronal death by 21.2%, and the neurite degeneration triggered by ischemia. Our results also demonstrate that with this treatment there is an increase of 89.2% on the number cells expressing ERK1/2, of 20.1% on the number of cells expressing c-Fos, and a synaptogenic effect, through an increase of 62.9% in the number of synaptic puncta as well as of 49.4% in their intensity. Interestingly, our results indicate that astrocytes are crucial to the beneficial effects triggered by HF-rMS after ischemia, thus suggesting a direct effect of HF-rMS on these cells. The modulation of astrocytes with this non-invasive brain stimulation technique is a promising approach to promote the recovery of ischemia-induced injured tissues; however, it is essential to understand how these effects can be modulated in order to optimize the protocols and enhance the beneficial outcomes.
Collapse
Affiliation(s)
- Cláudio Roque
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal.,Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Nuno Pinto
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Maria Vaz Patto
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal.,Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Graça Baltazar
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal.,Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| |
Collapse
|
5
|
Primary cilia safeguard cortical neurons in neonatal mouse forebrain from environmental stress-induced dendritic degeneration. Proc Natl Acad Sci U S A 2020; 118:2012482118. [PMID: 33443207 DOI: 10.1073/pnas.2012482118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The developing brain is under the risk of exposure to a multitude of environmental stressors. While perinatal exposure to excessive levels of environmental stress is responsible for a wide spectrum of neurological and psychiatric conditions, the developing brain is equipped with intrinsic cell protection, the mechanisms of which remain unknown. Here we show, using neonatal mouse as a model system, that primary cilia, hair-like protrusions from the neuronal cell body, play an essential role in protecting immature neurons from the negative impacts of exposure to environmental stress. More specifically, we found that primary cilia prevent the degeneration of dendritic arbors upon exposure to alcohol and ketamine, two major cell stressors, by activating cilia-localized insulin-like growth factor 1 receptor and downstream Akt signaling. We also found that activation of this pathway inhibits Caspase-3 activation and caspase-mediated cleavage/fragmentation of cytoskeletal proteins in stress-exposed neurons. These results indicate that primary cilia play an integral role in mitigating adverse impacts of environmental stressors such as drugs on perinatal brain development.
Collapse
|
6
|
Molinari M, Masciullo M. Stroke and potential benefits of brain-computer interface. HANDBOOK OF CLINICAL NEUROLOGY 2020; 168:25-32. [PMID: 32164857 DOI: 10.1016/b978-0-444-63934-9.00003-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
To treat stroke and, in particular, to alleviate the personal and social burden of stroke survivors is a main challenge for neuroscience research. Advancements in the knowledge of neurobiologic mechanisms subserving stroke-related damage and recovery provide key data to guide clinicians to tailor interventions to specific patient's needs. How does the brain-computer interface (BCI) fit into this scenario? A technique created to allow completely paralyzed individuals to control the environment recently introduced a new line of development: to provide a means to possibly control formation and changes in the brain network organization. In a sort of revolution, similar to the change from geocentric to heliocentric planet organization envisioned by Copernicus, we are facing a critical change in BCI research, moving from a brain to computer direction to a computer to brain one. This direction change will profoundly open up new avenues for BCI research and clinical applications. In this chapter, we address this change and discuss present and future applications of this new line idea of BCI use in stroke.
Collapse
Affiliation(s)
- Marco Molinari
- Department of Neurorehabilitation, Fondazione Santa Lucia IRCCS, Rome, Italy.
| | - Marcella Masciullo
- Department of Neurorehabilitation, Fondazione Santa Lucia IRCCS, Rome, Italy
| |
Collapse
|
7
|
Bisicchia E, Sasso V, Molinari M, Viscomi MT. Plasticity of microglia in remote regions after focal brain injury. Semin Cell Dev Biol 2019; 94:104-111. [PMID: 30703556 DOI: 10.1016/j.semcdb.2019.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
Abstract
The CNS is endowed with an intrinsic ability to recover from and adapt secondary compensatory mechanisms to injury. The basis of recovery stems from brain plasticity, defined as the brain's ability to make adaptive changes on structural and functional levels, ranging from molecular, synaptic, and cellular changes in response to alterations in their environment. In this multitude of responses, microglia have an active role and contribute to brain plasticity through their dynamic responses. This review will provide an overview of microglial responses in the context of acute CNS injury and their function in post-traumatic repair and assess the changes that are induced by damage in remote areas from, but functionally connected to, the primary site of injury. In the second section, we highlight the effects of several therapeutic approaches, with particular interest paid to specialized pro-resolving lipid mediators, in modulating microglial responses in remote regions and enhancing long-term functional recovery via suppression of neurodegenerative cascades that are induced by damage, which may contribute to a translational bridge from bench to bedside.
Collapse
Affiliation(s)
- Elisa Bisicchia
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valeria Sasso
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marco Molinari
- Laboratory of Experimental Neurorehabilitation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Teresa Viscomi
- Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del S. Cuore, Rome, Italy.
| |
Collapse
|
8
|
IL-10 Promotes Neurite Outgrowth and Synapse Formation in Cultured Cortical Neurons after the Oxygen-Glucose Deprivation via JAK1/STAT3 Pathway. Sci Rep 2016; 6:30459. [PMID: 27456198 PMCID: PMC4960594 DOI: 10.1038/srep30459] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/06/2016] [Indexed: 01/02/2023] Open
Abstract
As a classic immunoregulatory and anti-inflammatory cytokine, interleukin-10 (IL-10) provides neuroprotection in cerebral ischemia in vivo or oxygen-glucose deprivation (OGD)-induced injury in vitro. However, it remains blurred whether IL-10 promotes neurite outgrowth and synapse formation in cultured primary cortical neurons after OGD injury. In order to evaluate its effect on neuronal apoptosis, neurite outgrowth and synapse formation, we administered IL-10 or IL-10 neutralizing antibody (IL-10NA) to cultured rat primary cortical neurons after OGD injury. We found that IL-10 treatment activated the Janus kinase 1 (JAK1)/signal transducers and activators of transcription 3 (STAT3) signaling pathway. Moreover, IL-10 attenuated OGD-induced neuronal apoptosis by down-regulating the Bax expression and up-regulating the Bcl-2 expression, facilitated neurite outgrowth by increasing the expression of Netrin-1, and promoted synapse formation in cultured primary cortical neurons after OGD injury. These effects were partly abolished by JAK1 inhibitor GLPG0634. Contrarily, IL-10NA produced opposite effects on the cultured cortical neurons after OGD injury. Taken together, our findings suggest that IL-10 not only attenuates neuronal apoptosis, but also promotes neurite outgrowth and synapse formation via the JAK1/STAT3 signaling pathway in cultured primary cortical neurons after OGD injury.
Collapse
|
9
|
Jin C, Londono I, Mallard C, Lodygensky GA. New means to assess neonatal inflammatory brain injury. J Neuroinflammation 2015; 12:180. [PMID: 26407958 PMCID: PMC4583178 DOI: 10.1186/s12974-015-0397-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/10/2015] [Indexed: 01/23/2023] Open
Abstract
Preterm infants are especially vulnerable to infection-induced white matter injury, associated with cerebral palsy, cognitive and psychomotor impairment, and other adverse neurological outcomes. The etiology of such lesions is complex and multifactorial. Furthermore, timing and length of exposure to infection also influence neurodevelopmental outcomes. Different mechanisms have been posited to mediate the observed brain injury including microglial activation followed by subsequent release of pro-inflammatory species, glutamate-induced excitotoxicity, and vulnerability of developing oligodendrocytes to cerebral insults. The prevalence of such neurological impairments requires an urgent need for early detection and effective neuroprotective strategies. Accordingly, noninvasive methods of monitoring disease progression and therapy effectiveness are essential. While diagnostic tools using biomarkers from bodily fluids may provide useful information regarding potential risks of developing neurological diseases, the use of magnetic resonance imaging/spectroscopy has emerged as a promising candidate for such purpose. Various pharmacological agents have demonstrated protective effects in the immature brain in animal models; however, few studies have progressed to clinical trials with promising results.
Collapse
Affiliation(s)
- Chen Jin
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | - Irene Londono
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | - Carina Mallard
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden.
| | - Gregory A Lodygensky
- Department of Pediatrics, Sainte-Justine Hospital and Research Center, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada. .,Montreal Heart Institute, 5000 Rue Bélanger, Montréal, Québec, Canada. .,Department of Neuroscience and Pharmacology, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
10
|
Lin L, Chen H, Zhang Y, Lin W, Liu Y, Li T, Zeng Y, Chen J, Du H, Chen R, Tan Y, Liu N. IL-10 Protects Neurites in Oxygen-Glucose-Deprived Cortical Neurons through the PI3K/Akt Pathway. PLoS One 2015; 10:e0136959. [PMID: 26366999 PMCID: PMC4569574 DOI: 10.1371/journal.pone.0136959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 08/10/2015] [Indexed: 12/15/2022] Open
Abstract
IL-10, as a cytokine, has an anti-inflammatory cascade following various injuries, but it remains blurred whether IL-10 protects neurites of cortical neurons after oxygen-glucose deprivation injury. Here, we reported that IL-10, in a concentration-dependent manner, reduced neuronal apoptosis and increased neuronal survival in oxygen-glucose-deprived primary cortical neurons, producing an optimal protective effect at 20ng/ml. After staining NF-H and GAP-43, we found that IL-10 significantly protected neurites in terms of axon length and dendrite number by confocal microscopy. Furthermore, it induced the phosphorylation of AKT, suppressed the activation of caspase-3, and up-regulated the protein expression of GAP-43. In contrast, LY294002, a specific inhibitor of PI3K/AKT, reduced the level of AKT phosphorylation and GAP-43 expression, increased active caspase-3 expression and thus significantly weakened IL-10-mediated protective effect in the OGD-induced injury model. IL-10NA, the IL-10 neutralizing antibody, reduced the level of p-PI3K phosphorylation and increased the expression of active caspase-3. These findings suggest that IL-10 provides neuroprotective effects by protecting neurites through PI3K/AKT signaling pathway in oxygen-glucose-deprived primary cortical neurons.
Collapse
Affiliation(s)
- Longzai Lin
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Hongbin Chen
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Yixian Zhang
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
- Department of Rehabilitation, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Wei Lin
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Yong Liu
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Tin Li
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Yongping Zeng
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Jianhao Chen
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Houwei Du
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Ronghua Chen
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Yi Tan
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
| | - Nan Liu
- Department of Neurology, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, Fujian, People’s Republic of China
- Department of Rehabilitation, The Affiliated Union Hospital, Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- * E-mail:
| |
Collapse
|
11
|
Klistorner A, Vootakuru N, Wang C, Yiannikas C, Graham SL, Parratt J, Garrick R, Levin N, Masters L, Lagopoulos J, Barnett MH. Decoding diffusivity in multiple sclerosis: analysis of optic radiation lesional and non-lesional white matter. PLoS One 2015; 10:e0122114. [PMID: 25807541 PMCID: PMC4373765 DOI: 10.1371/journal.pone.0122114] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/17/2015] [Indexed: 12/19/2022] Open
Abstract
Objectives Diffusion tensor imaging (DTI) has been suggested as a new promising tool in MS that may provide greater pathological specificity than conventional MRI, helping, therefore, to elucidate disease pathogenesis and monitor therapeutic efficacy. However, the pathological substrates that underpin alterations in brain tissue diffusivity are not yet fully delineated. Tract-specific DTI analysis has previously been proposed in an attempt to alleviate this problem. Here, we extended this approach by segmenting a single tract into areas bound by seemingly similar pathological processes, which may better delineate the potential association between DTI metrics and underlying tissue damage. Method Several compartments were segmented in optic radiation (OR) of 50 relapsing-remitting MS patients including T2 lesions, proximal and distal parts of fibers transected by lesion and fibers with no discernable pathology throughout the entire length of the OR. Results Asymmetry analysis between lesional and non-lesional fibers demonstrated a marked increase in Radial Diffusivity (RD), which was topographically limited to focal T2 lesions and potentially relates to the lesional myelin loss. A relative elevation of Axial Diffusivity (AD) in the distal part of the lesional fibers was observed in a distribution consistent with Wallerian degeneration, while diffusivity in the proximal portion of transected axons remained normal. A moderate, but significant elevation of RD in OR non-lesional fibers was strongly associated with the global (but not local) T2 lesion burden and is probably related to microscopic demyelination undetected by conventional MRI. Conclusion This study highlights the utility of the compartmentalization approach in elucidating the pathological substrates of diffusivity and demonstrates the presence of tissue-specific patterns of altered diffusivity in MS, providing further evidence that DTI is a sensitive marker of tissue damage in both lesions and NAWM. Our results suggest that, at least within the OR, parallel and perpendicular diffusivities are affected by tissue restructuring related to distinct pathological processes.
Collapse
Affiliation(s)
- Alexander Klistorner
- Department of Ophthalmology, Save Sight Institute, University of Sydney, Sydney, Australia
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
- * E-mail:
| | | | - Chenyu Wang
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | | | - Stuart L. Graham
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | | | | - Netta Levin
- Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Lynette Masters
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Jim Lagopoulos
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Michael H. Barnett
- Brain and Mind Research Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
12
|
Viscomi MT, Molinari M. Remote neurodegeneration: multiple actors for one play. Mol Neurobiol 2014; 50:368-89. [PMID: 24442481 DOI: 10.1007/s12035-013-8629-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 12/24/2013] [Indexed: 12/19/2022]
Abstract
Remote neurodegeneration significantly influences the clinical outcome in many central nervous system (CNS) pathologies, such as stroke, multiple sclerosis, and traumatic brain and spinal cord injuries. Because these processes develop days or months after injury, they are accompanied by a therapeutic window of opportunity. The complexity and clinical significance of remote damage is prompting many groups to examine the factors of remote degeneration. This research is providing insights into key unanswered questions, opening new avenues for innovative neuroprotective therapies. In this review, we evaluate data from various remote degeneration models to describe the complexity of the systems that are involved and the importance of their interactions in reducing damage and promoting recovery after brain lesions. Specifically, we recapitulate the current data on remote neuronal degeneration, focusing on molecular and cellular events, as studied in stroke and brain and spinal cord injury models. Remote damage is a multifactorial phenomenon in which many components become active in specific time frames. Days, weeks, or months after injury onset, the interplay between key effectors differentially affects neuronal survival and functional outcomes. In particular, we discuss apoptosis, inflammation, oxidative damage, and autophagy-all of which mediate remote degeneration at specific times. We also review current findings on the pharmacological manipulation of remote degeneration mechanisms in reducing damage and sustaining outcomes. These novel treatments differ from those that have been proposed to limit primary lesion site damage, representing new perspectives on neuroprotection.
Collapse
Affiliation(s)
- Maria Teresa Viscomi
- Experimental Neurorehabilitation Laboratory, Santa Lucia Foundation I.R.C.C.S., Via del Fosso di Fiorano 65, 00143, Rome, Italy,
| | | |
Collapse
|
13
|
Ling C, Hendrickson ML, Kalil RE. Morphology, classification, and distribution of the projection neurons in the dorsal lateral geniculate nucleus of the rat. PLoS One 2012; 7:e49161. [PMID: 23139837 PMCID: PMC3489731 DOI: 10.1371/journal.pone.0049161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 10/03/2012] [Indexed: 11/18/2022] Open
Abstract
The morphology of confirmed projection neurons in the dorsal lateral geniculate nucleus (dLGN) of the rat was examined by filling these cells retrogradely with biotinylated dextran amine (BDA) injected into the visual cortex. BDA-labeled projection neurons varied widely in the shape and size of their cell somas, with mean cross-sectional areas ranging from 60–340 µm2. Labeled projection neurons supported 7–55 dendrites that spanned up to 300 µm in length and formed dendritic arbors with cross-sectional areas of up to 7.0×104 µm2. Primary dendrites emerged from cell somas in three broad patterns. In some dLGN projection neurons, primary dendrites arise from the cell soma at two poles spaced approximately 180° apart. In other projection neurons, dendrites emerge principally from one side of the cell soma, while in a third group of projection neurons primary dendrites emerge from the entire perimeter of the cell soma. Based on these three distinct patterns in the distribution of primary dendrites from cell somas, we have grouped dLGN projection neurons into three classes: bipolar cells, basket cells and radial cells, respectively. The appendages seen on dendrites also can be grouped into three classes according to differences in their structure. Short “tufted” appendages arise mainly from the distal branches of dendrites; “spine-like” appendages, fine stalks with ovoid heads, typically are seen along the middle segments of dendrites; and “grape-like” appendages, short stalks that terminate in a cluster of ovoid bulbs, appear most often along the proximal segments of secondary dendrites of neurons with medium or large cell somas. While morphologically diverse dLGN projection neurons are intermingled uniformly throughout the nucleus, the caudal pole of the dLGN contains more small projection neurons of all classes than the rostral pole.
Collapse
Affiliation(s)
- Changying Ling
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michael L. Hendrickson
- W.M. Keck Laboratory for Biological Imaging, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ronald E. Kalil
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|