951
|
Hilton BJ, Blanquie O, Tedeschi A, Bradke F. High-resolution 3D imaging and analysis of axon regeneration in unsectioned spinal cord with or without tissue clearing. Nat Protoc 2019; 14:1235-1260. [DOI: 10.1038/s41596-019-0140-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022]
|
952
|
Papa S, Rossi F, Vismara I, Forloni G, Veglianese P. Nanovector-Mediated Drug Delivery in Spinal Cord Injury: A Multitarget Approach. ACS Chem Neurosci 2019; 10:1173-1182. [PMID: 30763071 DOI: 10.1021/acschemneuro.8b00700] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Many preclinical studies seek cures for spinal cord injury (SCI), but when the results are translated to clinical trials they give scant efficacy. One possible reason is that most strategies use treatments directed toward a single pathological mechanism, while a multitherapeutic approach needs to be tested to significantly improve outcomes after SCI. Most of the preclinical reports gave better outcomes when a combination of different compounds was used instead of a single drug. This promising approach, however, must still be improved because it raises some criticism: (i) the blood-spinal cord barrier limits drug distribution, (ii) it is hard to understand the interactions among the pharmacological components after systemic administration, and (iii) the timing of treatments is crucial: the spread of the lesion is a process finely regulated over time, so therapies must be scheduled at precise times during the postinjury course. Nanomedicine could be useful to overcome these limitations. Nanotools allow finely regulated drug administration in terms of cell selectivity and release kinetics. We believe that excellent therapeutic results could be obtained by exploiting this tool in multitherapy. Combining nanoparticles loaded with different compounds that act on the main pathological pathways could overcome the restrictions of traditional drug delivery routes, a major limit for the clinical application of multitherapy. This review digs into these topics, discussing the critical aspects of multitherapies now proposed and suggesting new points of view.
Collapse
Affiliation(s)
- Simonetta Papa
- Dipartimento di Neuroscienze, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via La Masa 19, 20156 Milan, Italy
| | - Filippo Rossi
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “Giulio Natta”, Politecnico di Milano, via Mancinelli 7, 20131 Milan, Italy
| | - Irma Vismara
- Dipartimento di Neuroscienze, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via La Masa 19, 20156 Milan, Italy
| | - Gianluigi Forloni
- Dipartimento di Neuroscienze, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via La Masa 19, 20156 Milan, Italy
| | - Pietro Veglianese
- Dipartimento di Neuroscienze, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via La Masa 19, 20156 Milan, Italy
| |
Collapse
|
953
|
Murphy‐Royal C, Gordon GR, Bains JS. Stress‐induced structural and functional modifications of astrocytes—Further implicating glia in the central response to stress. Glia 2019; 67:1806-1820. [DOI: 10.1002/glia.23610] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Ciaran Murphy‐Royal
- Department of Physiology and Pharmacology, Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| | - Grant R. Gordon
- Department of Physiology and Pharmacology, Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| | - Jaideep S. Bains
- Department of Physiology and Pharmacology, Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| |
Collapse
|
954
|
Matias I, Morgado J, Gomes FCA. Astrocyte Heterogeneity: Impact to Brain Aging and Disease. Front Aging Neurosci 2019; 11:59. [PMID: 30941031 PMCID: PMC6433753 DOI: 10.3389/fnagi.2019.00059] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Astrocytes, one of the largest glial cell population in the central nervous system (CNS), play a key function in several events of brain development and function, such as synapse formation and function, control of neurotransmitters release and uptake, production of trophic factors and control of neuronal survival. Initially described as a homogenous population, several evidences have pointed that astrocytes are highly heterogeneous, both morphologically and functionally, within the same region, and across different brain regions. Recent findings suggest that the heterogeneity in the expression profile of proteins involved in astrocyte function may predict the selective vulnerability of brain regions to specific diseases, as well as to the age-related cognitive decline. However, the molecular mechanisms underlying these changes, either in aging as well as in brain disease are scarce. Neuroinflammation, a hallmark of several neurodegenerative diseases and aging, is reported to have a dubious impact on glial activation, as these cells release pro- and anti-inflammatory cytokines and chemokines, anti-oxidants, free radicals, and neurotrophic factors. Despite the emerging evidences supporting that reactive astrocytes have a duality in their phenotype, neurotoxic or neuroprotective properties, depending on the age and stimuli, the underlying mechanisms of their activation, cellular interplays and the impact of regional astrocyte heterogeneity are still a matter of discussion. In this review article, we will summarize recent findings on astrocyte heterogeneity and phenotypes, as well as their likely impact for the brain function during aging and neural diseases. We will focus on the molecules and mechanisms triggered by astrocyte to control synapse formation in different brain regions. Finally, we will discuss new evidences on how the modulation of astrocyte phenotype and function could impact the synaptic deficits and glial dysfunction present in aging and pathological states.
Collapse
Affiliation(s)
- Isadora Matias
- Laboratory of Cellular Neurobiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Morgado
- Laboratory of Cellular Neurobiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flávia Carvalho Alcantara Gomes
- Laboratory of Cellular Neurobiology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
955
|
Naveed M, Zhou QG, Han F. Cerebrovascular inflammation: A critical trigger for neurovascular injury? Neurochem Int 2019; 126:165-177. [PMID: 30890409 DOI: 10.1016/j.neuint.2019.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/05/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
The cerebrovascular system is not only inert bystandard that support the metabolic demands of the brain but also elicit the barrier functions against risk factors mediated neurovascular injury. The onsets of cerebrovascular inflammation are considered as stimuli that can provoke the host defense system and trigger the development of neurological disorders. Homeostasis of the brain function is regulated by the movement of endothelial, glial, and neuronal cells within the neurovascular unit (NVU), which acts as a "platform" for the coordinated action of anti- and pro-inflammatory mechanisms. The cerebrovascular system plays an integral role in the inflammatory response by either producing or expressing a variety of cytokines, adhesion molecules, metalloproteinases, and serine proteases. Excessive inflammatory cytokine production can further be affecting the blood-brain barrier (BBB) integrity and lead to brain tissue damage. In this review, we summarize the more recent evidence highlighting the importance of cerebrovascular injury in terms of risk prediction, and the mechanisms mediating the upregulation of inflammatory mediators in cerebrovascular dysfunction and neurodegeneration.
Collapse
Affiliation(s)
- Muhammad Naveed
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, PR China
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, PR China; Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, PR China
| | - Feng Han
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, PR China.
| |
Collapse
|
956
|
Jung HS, Kim HH, Shin MH, Kim S, Kim KS, Cho K, Hahn SK. Electroceutical Residue-Free Graphene Device for Dopamine Monitoring and Neural Stimulation. ACS Biomater Sci Eng 2019; 5:2013-2020. [DOI: 10.1021/acsbiomaterials.8b01488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ho Sang Jung
- Advanced Nano-Surface Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam 641-831, Korea
| | | | | | | | - Ki Su Kim
- Department of Organic Materials Science and Engineering, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | | | | |
Collapse
|
957
|
Sun G, Zeng S, Liu X, Shi H, Zhang R, Wang B, Zhou C, Yu T. Synthesis and Characterization of a Silica-Based Drug Delivery System for Spinal Cord Injury Therapy. NANO-MICRO LETTERS 2019; 11:23. [PMID: 34137964 PMCID: PMC7770885 DOI: 10.1007/s40820-019-0252-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/19/2019] [Indexed: 06/11/2023]
Abstract
Acute inflammation is a central component in the progression of spinal cord injury (SCI). Anti-inflammatory drugs used in the clinic are often administered systemically at high doses, which can paradoxically increase inflammation and result in drug toxicity. A cluster-like mesoporous silica/arctigenin/CAQK composite (MSN-FC@ARC-G) drug delivery system was designed to avoid systemic side effects of high-dose therapy by enabling site-specific drug delivery to the spinal cord. In this nanosystem, mesoporous silica was modified with the FITC fluorescent molecule and CAQK peptides that target brain injury and SCI sites. The size of the nanocarrier was kept at approximately 100 nm to enable penetration of the blood-brain barrier. Arctigenin, a Chinese herbal medicine, was loaded into the nanosystem to reduce inflammation. The in vivo results showed that MSN-FC@ARC-G could attenuate inflammation at the injury site. Behavior and morphology experiments suggested that MSN-FC@ARC-G could diminish local microenvironment damage, especially reducing the expression of interleukin-17 (IL-17) and IL-17-related inflammatory factors, inhibiting the activation of astrocytes, thus protecting neurons and accelerating the recovery of SCI. Our study demonstrated that this novel, silica-based drug delivery system has promising potential for clinical application in SCI therapy.
Collapse
Affiliation(s)
- Guodong Sun
- Department of Orthopedics, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Shenghui Zeng
- College of Chemistry and Material Sciences, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Xu Liu
- College of Chemistry and Material Sciences, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Haishan Shi
- College of Chemistry and Material Sciences, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Renwen Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Baocheng Wang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, People's Republic of China
| | - Changren Zhou
- College of Chemistry and Material Sciences, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Tao Yu
- College of Chemistry and Material Sciences, Jinan University, Guangzhou, 510632, People's Republic of China.
| |
Collapse
|
958
|
Fawcett JW. The Paper that Restarted Modern Central Nervous System Axon Regeneration Research. Trends Neurosci 2019; 41:239-242. [PMID: 29703373 DOI: 10.1016/j.tins.2018.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/26/2018] [Indexed: 10/17/2022]
Abstract
Spinal cord repair research appeared to have run out of new ideas in the 1970s. In a 1981 paper, the Aguayo Laboratory revisited an experiment by Tello and Cajal that suggested that central nervous system (CNS) axons could regenerate into peripheral nerve grafts. Using modern axon tracing methods, David and Aguayo showed that axons from neurons in the spinal cord could regenerate for long distances within peripheral nervous system (PNS) grafts, but not back into the CNS. This proved that damaged CNS tissue is inhibitory to axon regeneration while PNS tissue is permissive. The experiment sparked a research revival, leading to the identification of many inhibitory molecules that block axon growth in the mature CNS.
Collapse
Affiliation(s)
- James W Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK; Institute for Experimental Medicine Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
959
|
Sabin KZ, Jiang P, Gearhart MD, Stewart R, Echeverri K. AP-1 cFos/JunB/miR-200a regulate the pro-regenerative glial cell response during axolotl spinal cord regeneration. Commun Biol 2019; 2:91. [PMID: 30854483 PMCID: PMC6403268 DOI: 10.1038/s42003-019-0335-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 02/04/2019] [Indexed: 12/30/2022] Open
Abstract
Salamanders have the remarkable ability to functionally regenerate after spinal cord transection. In response to injury, GFAP+ glial cells in the axolotl spinal cord proliferate and migrate to replace the missing neural tube and create a permissive environment for axon regeneration. Molecular pathways that regulate the pro-regenerative axolotl glial cell response are poorly understood. Here we show axolotl glial cells up-regulate AP-1cFos/JunB after injury, which promotes a pro-regenerative glial cell response. Injury induced upregulation of miR-200a in glial cells supresses c-Jun expression in these cells. Inhibition of miR-200a during regeneration causes defects in axonal regrowth and transcriptomic analysis revealed that miR-200a inhibition leads to differential regulation of genes involved with reactive gliosis, the glial scar, extracellular matrix remodeling and axon guidance. This work identifies a unique role for miR-200a in inhibiting reactive gliosis in axolotl glial cells during spinal cord regeneration. Keith Sabin et al. showed that upregulation of the AP-1 complex, composed of c-Fos and JunB, in the axolotl spinal cord promotes a pro-regenerative glial cell response. This response is impaired by inhibition of miR-200a; suggesting an important role for this microRNA in axolotl spinal cord regeneration.
Collapse
Affiliation(s)
- Keith Z Sabin
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA.,Marine Biological Laboratory, Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, 02543, MA, USA
| | - Peng Jiang
- Morgridge Institute for Research, Madison, 53715, WI, USA
| | - Micah D Gearhart
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, 53715, WI, USA
| | - Karen Echeverri
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA. .,Marine Biological Laboratory, Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, 02543, MA, USA.
| |
Collapse
|
960
|
Lee Y, Lee S, Chang SC, Lee J. Significant roles of neuroinflammation in Parkinson's disease: therapeutic targets for PD prevention. Arch Pharm Res 2019; 42:416-425. [PMID: 30830660 DOI: 10.1007/s12272-019-01133-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023]
Abstract
Glial cells outnumber neurons in the brain and play important roles in the neuroinflammation that accompanies brain damage in neurodegenerative diseases. In Parkinson's disease (PD), dopaminergic neuronal loss is accompanied by inflammatory changes in microglia, astrocytes, innate immune cells, and infiltrating peripheral immune cells. Neuroinflammation is probably a fundamental immune response to protect neurons from harm and compensate for neuronal damage, but at the same time, its neurotoxic effects exacerbate neuron damage. Furthermore, neuroinflammatory response is regulated by immune cells, such as microglia, astrocytes, and peripheral immune cells, and by cytokines and chemokines. Accordingly, it is crucial that we understand how such immune cells in the brain regulate neuroinflammatory responses in PD pathology. This review describes the roles played by glia-mediated neuroinflammation in PD, both good and bad, and the therapeutic strategies used to treat PD.
Collapse
Affiliation(s)
- Yujeong Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seulah Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Seung-Cheol Chang
- Institute of BioPhysio Sensor Technology, Pusan National University, Busan, 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
961
|
Zhang X, Zhao HH, Li D, Li HP. Neuroprotective effects of matrix metalloproteinases in cerebral ischemic rats by promoting activation and migration of astrocytes and microglia. Brain Res Bull 2019; 146:136-142. [DOI: 10.1016/j.brainresbull.2018.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/26/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
|
962
|
Hettiaratchi MH, O'Meara MJ, Teal CJ, Payne SL, Pickering AJ, Shoichet MS. Local delivery of stabilized chondroitinase ABC degrades chondroitin sulfate proteoglycans in stroke-injured rat brains. J Control Release 2019; 297:14-25. [DOI: 10.1016/j.jconrel.2019.01.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 02/07/2023]
|
963
|
Haindl MT, Köck U, Zeitelhofer‐Adzemovic M, Fazekas F, Hochmeister S. The formation of a glial scar does not prohibit remyelination in an animal model of multiple sclerosis. Glia 2019; 67:467-481. [PMID: 30484905 PMCID: PMC6588096 DOI: 10.1002/glia.23556] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 01/12/2023]
Abstract
The role of astrocytes in the pathophysiology of multiple sclerosis (MS) is discussed controversially. Especially the formation of the glial scar is often believed to act as a barrier for remyelination. At the same time, astrocytes are known to produce factors that influence oligodendrocyte precursor cell (OPC) survival. To explore these mechanisms, we investigated the astrocytic reaction in an animal model induced by immunization with myelin oligodendrocyte glycoprotein (MOG) in Dark Agouti (DA) rats, which mimics most of the histological features of MS. We correlated the astroglial reaction by immunohistochemistry (IHC) for glial fibrillary acidic protein (GFAP) to the remyelination capacity by in situ hybridization for mRNA of proteolipid protein (PLP), indicative of OPCs, over the full course of the disease. PLP mRNA peaked in early remyelinating lesions while the amount of GFAP positive astrocytes was highest in remyelinated lesions. In shadow plaques, we found at the same time all features of a glial scar and numbers of OPCs and mature oligodendrocytes, which were nearly equal to that in unaffected white matter areas. To assess the plaque environment, we furthermore quantitatively analyzed factors expressed by astrocytes previously suggested to influence remyelination. From our data, we conclude that remyelination occurs despite an abundant glial reaction in this animal model. The different patterns of astrocytic factors and the occurrence of different astrocytic phenotypes during lesion evolution furthermore indicate a finely regulated, balanced astrocytic involvement leading to successful repair.
Collapse
Affiliation(s)
| | - Ulrike Köck
- Center for Brain ResearchMedical University of ViennaViennaAustria
| | | | - Franz Fazekas
- Department of NeurologyMedical University of GrazGrazAustria
| | | |
Collapse
|
964
|
Mu X, Wang J, Li Y, Xu F, Long W, Ouyang L, Liu H, Jing Y, Wang J, Dai H, Liu Q, Sun Y, Liu C, Zhang XD. Redox Trimetallic Nanozyme with Neutral Environment Preference for Brain Injury. ACS NANO 2019; 13:1870-1884. [PMID: 30753061 DOI: 10.1021/acsnano.8b08045] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Metal nanozyme has attracted wide interest for biomedicine, and a highly catalytic material in the physiological environment is highly desired. However, catalytic selectivity of nanozyme is still highly challenging, limiting its wide application. Here, we show a trimetallic (triM) nanozyme with highly catalytic activity and environmental selectivity. Enzyme-mimicked investigations find that the triM system possesses multi-enzyme-mimetic activity for removing reactive oxygen species (ROS) and reactive nitrogen species (RNS), such as 1O2, H2O2, •OH, and •NO. Importantly, triM nanozyme exhibits the significant neutral environment preference for removing the •OH, 1O2, and •NO free radical, indicating its highly catalytic selectivity. The density functional theory (DFT) calculations reveal that triM nanozyme can capture electrons very easily and provides more attraction to reactive oxygen and nitrogen species (RONS) radicals in the neutral environment. In vitro experiments show that triM nanozyme can improve the viability of injured neural cell. In the LPS-induced brain injury model, the superoxide dismutase (SOD) activity and lipid peroxidation can be greatly recovered after triM nanozyme treatment. Moreover, the triM nanozyme treatment can significantly improve the survival rate, neuroinflammation, and reference memory of injured mice. Present work provides a feasible route for improving selectivity of nanozyme in the physiological environment as well as exploring potential applications in brain science.
Collapse
Affiliation(s)
- Xiaoyu Mu
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Junying Wang
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Yonghui Li
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Fujuan Xu
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Wei Long
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Lufei Ouyang
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Haile Liu
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Yaqi Jing
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Jingya Wang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Haitao Dai
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Qiang Liu
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Yuanming Sun
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Changlong Liu
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, Institute of Advanced Materials Physics, School of Sciences , Tianjin University , Tianjin 300350 , China
| |
Collapse
|
965
|
Aberrant Calcium Signals in Reactive Astrocytes: A Key Process in Neurological Disorders. Int J Mol Sci 2019; 20:ijms20040996. [PMID: 30823575 PMCID: PMC6413203 DOI: 10.3390/ijms20040996] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are abundant cells in the brain that regulate multiple aspects of neural tissue homeostasis by providing structural and metabolic support to neurons, maintaining synaptic environments and regulating blood flow. Recent evidence indicates that astrocytes also actively participate in brain functions and play a key role in brain disease by responding to neuronal activities and brain insults. Astrocytes become reactive in response to injury and inflammation, which is typically described as hypertrophy with increased expression of glial fibrillary acidic protein (GFAP). Reactive astrocytes are frequently found in many neurological disorders and are a hallmark of brain disease. Furthermore, reactive astrocytes may drive the initiation and progression of disease processes. Recent improvements in the methods to visualize the activity of reactive astrocytes in situ and in vivo have helped elucidate their functions. Ca2+ signals in reactive astrocytes are closely related to multiple aspects of disease and can be a good indicator of disease severity/state. In this review, we summarize recent findings concerning reactive astrocyte Ca2+ signals. We discuss the molecular mechanisms underlying aberrant Ca2+ signals in reactive astrocytes and the functional significance of aberrant Ca2+ signals in neurological disorders.
Collapse
|
966
|
Chen W, Huang Q, Ma S, Li M. Progress in Dopaminergic Cell Replacement and Regenerative Strategies for Parkinson's Disease. ACS Chem Neurosci 2019; 10:839-851. [PMID: 30346716 DOI: 10.1021/acschemneuro.8b00389] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder symptomatically characterized by resting tremor, rigidity, bradykinesia, and gait impairment. These motor deficits suffered by PD patients primarily result from selective dysfunction or loss of dopaminergic neurons of the substantia nigra pars compacta (SNpc). Most of the existing therapies for PD are based on the replacement of dopamine, which is symptomatically effective in the early stage but becomes increasingly less effective and is accompanied by serious side effects in the advanced stages of the disease. Currently, there are no strategies to slow neuronal degeneration or prevent the progression of PD. Thus, the prospect of regenerating functional dopaminergic neurons is very attractive. Over the last few decades, significant progress has been made in the development of dopaminergic regenerative strategies for curing PD. The most promising approach seems to be cell-replacement therapy (CRT) using human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs), which are unlimitedly available and have gained much success in preclinical trials. Despite the challenges, stem cell-based CRT will make significant steps toward the clinic in the coming decade. Alternatively, direct lineage reprogramming, especially in situ direct conversion of glia cells to induced neurons, which exhibits some advantages including no ethical concerns, no risk of tumor formation, and even no need for transplantation, has gained much attention recently. Evoking the endogenous regeneration ability of neural stem cells (NSCs) is an idyllic method of dopaminergic neuroregeneration which remains highly controversial. Here, we review many of these advances, highlighting areas and strategies that might be particularly suited to the development of regenerative approaches that restore dopaminergic function in PD.
Collapse
Affiliation(s)
- Weizhao Chen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Qiaoying Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| |
Collapse
|
967
|
Sadick JS, Liddelow SA. Don't forget astrocytes when targeting Alzheimer's disease. Br J Pharmacol 2019; 176:3585-3598. [PMID: 30636042 DOI: 10.1111/bph.14568] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are essential for CNS health, regulating homeostasis, metabolism, and synaptic transmission. In addition to these and many other physiological roles, the pathological impact of astrocytes ("reactive astrocytes") in acute trauma and chronic disease like Alzheimer's disease (AD) is well established. Growing evidence supports a fundamental and active role of astrocytes in multiple neurodegenerative diseases. With a growing interest in normal astrocyte biology, and countless studies on changes in astrocyte function in the context of disease, it may be a surprise that no therapies exist incorporating astrocytes as key targets. Here, we examine unintentional effects of current AD therapies on astrocyte function and theorize how astrocytes may be intentionally targeted for more efficacious therapeutic outcomes. Given their integral role in normal neuronal functioning, incorporating astrocytes as key criteria for AD drug development can only lead to more effective therapies for the millions of AD sufferers worldwide. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Jessica S Sadick
- Neuroscience Institute, NYU Langone Medical Center, New York, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Langone Medical Center, New York, USA.,Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, USA.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
968
|
Hassannejad Z, Yousefifard M, Azizi Y, Zadegan SA, Sajadi K, Sharif-Alhoseini M, Shakouri-Motlagh A, Mokhatab M, Rezvan M, Shokraneh F, Hosseini M, Vaccaro AR, Harrop JS, Rahimi-Movaghar V. Axonal degeneration and demyelination following traumatic spinal cord injury: A systematic review and meta-analysis. J Chem Neuroanat 2019; 97:9-22. [PMID: 30726717 DOI: 10.1016/j.jchemneu.2019.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/22/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022]
Abstract
The pathophysiology of spinal cord injury (SCI) related processes of axonal degeneration and demyelination are poorly understood. The present systematic review and meta-analysis were performed such to establish quantitative results of animal studies regarding the role of injury severity, SCI models and level of injury on the pathophysiology of axon and myelin sheath degeneration. 39 related articles were included in the analysis. The compiled data showed that the total number of axons, number of myelinated axons, myelin sheath thickness, axonal conduction velocity, and internode length steadily decreased as time elapsed from the injury (Pfor trend<0.0001). The rate of axonal retrograde degeneration was affected by SCI model and severity of the injury. Axonal degeneration was higher in injuries of the thoracic region. The SCI model and the site of the injury also affected axonal retrograde degeneration. The number of myelinated axons in the caudal region of the injury was significantly higher than the lesion site and the rostral region. The findings of the present meta-analysis show that the pathophysiology of axons and myelin sheath differ in various phases of SCI and are affected by multiple factors related to the injury.
Collapse
Affiliation(s)
- Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yaser Azizi
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Abdollah Zadegan
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiavash Sajadi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Sharif-Alhoseini
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Shakouri-Motlagh
- Department of Chemical and Biomolecular Engineering, University of Melbourne, Victoria 3010, Australia
| | - Mona Mokhatab
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Motahareh Rezvan
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Shokraneh
- Cochrane Schizophrenia Group, Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexander R Vaccaro
- Department of Orthopedics and Neurosurgery, Rothman Institute, Thomas Jefferson University Philadelphia, USA
| | - James S Harrop
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Brain and Spinal Injuries Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
969
|
Hassannejad Z, Zadegan SA, Vaccaro AR, Rahimi-Movaghar V, Sabzevari O. Biofunctionalized peptide-based hydrogel as an injectable scaffold for BDNF delivery can improve regeneration after spinal cord injury. Injury 2019; 50:278-285. [PMID: 30595411 DOI: 10.1016/j.injury.2018.12.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/18/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND The complex pathophysiological events occurring after traumatic spinal cord injuries (TSCI) make this devastating trauma still incurable. Peptide amphiphile (PA) hydrogels are nanobiomaterials displaying desirable properties for application in regenerative medicine because they are absorbable, injectable, allowing biofunctionalization, controlling release of trophic factors and mimic extracellular matrix (ECM). In this study, we explored the potentiality of the IKVAV-functionalized PA hydrogel to provide a permissive environment for cell migration and growth as well as sustained release of BDNF at the lesion after severe compression injury model. METHODS The IKVAV-functionalized PA was synthesized by automated solid-phase approach and its secondary structure was evaluated by Circular dichroism (CD) spectroscopy. The potential of IKVAV-functionalized PA to self-assemble into nanofibers and hydrogel formation were assessed using transmission electron microscopy (TEM). Release profiles of BDNF from hydrogel and the bioactivity of the released BDNF from hydrogel were determined using ELISA and DRG bioassay, respectively. Severe spinal cord injury was induced using clip compression at T7-T8 vertebral segment. Twenty four hours post-injury the animals were treated by either IKVAV PA hydrogel, BDNF-loaded IKVAV PA hydrogel, BDNF solution or saline. Two and six weeks later, animals were sacrificed and the lesion site was evaluated based on GFAP, CD68 and ß III tubulin immunoreactivity. Also, locomotor recovery was assessed during 6 weeks using Basso, Beattie, Bresnahan (BBB) scoring test. RESULTS The IKVAV PA arranged into nanofibrous structure and provided a sustained release of BDNF over 21 days while preserved the bioactivity of BDNF. Also, BDNF loading influenced the hydrogel nanostructure resulting in aligned orientation of nanofibers. Injection of BDNF-loaded IKVAV PA hydrogel resulted in a considerable axon preservation and astrogliosis reduction at 6 weeks post-injury without showing any inflammatory reaction. However, the BBB score was not statistically different between different treatment groups. CONCLUSION Although the locomotor functional recovery was not observed in this study, the axon preservation and minimal inflammation in animals treated with BDNF-incorporated hydrogel indicate the potentiality of the designed intervention for further evaluations in the path of developing efficient therapies for severe spinal cord injury.
Collapse
Affiliation(s)
- Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayan Abdollah Zadegan
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexander R Vaccaro
- Department of Orthopaedic Surgery, Rothman Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Sabzevari
- Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
970
|
Huang Z, Yarong G, Shimoda Y, Watanabe K, Liu Y. Induced NB-3 Limits Regenerative Potential of Serotonergic Axons after Complete Spinal Transection. J Neurotrauma 2019; 36:436-447. [PMID: 30156464 DOI: 10.1089/neu.2018.5652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
NB-3 (contactin-6) is a member of the contactin family and has a wide range of roles during central nervous system development and disease. Here, we found that NB-3 was simultaneously induced in the serotonergic raphespinal tract (sRST) axons and in the scar-forming cells after spinal cord injury (SCI). Regrowth of sRST axons was promoted in vivo by blocking NB-3 expression in either sRST axons or scar-forming cells when post-traumatic axons of the sRST tried to penetrate the glial scar. NB-3 deficiency promoted synapse reformation between sRST regenerative axons and motor neurons and enhanced the potential for electrical activity of muscle contraction and motor coordination. In vivo evidence also suggested that NB-3 induction in both sRST axons and scar-forming cells was required to mediate NB-3 signaling inhibition of sRST axon regeneration after SCI. Our findings suggest that NB-3 protein is a potential molecular target for future SCI treatments.
Collapse
Affiliation(s)
- Zhenhui Huang
- 1 Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Gao Yarong
- 1 Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yasushi Shimoda
- 2 Department of Bioengineering, Nagaoka University of Technology, Niigata, Japan
| | | | - Yaobo Liu
- 1 Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
971
|
Conditional BDNF Delivery from Astrocytes Rescues Memory Deficits, Spine Density, and Synaptic Properties in the 5xFAD Mouse Model of Alzheimer Disease. J Neurosci 2019; 39:2441-2458. [PMID: 30700530 DOI: 10.1523/jneurosci.2121-18.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 12/25/2022] Open
Abstract
It has been well documented that neurotrophins, including brain-derived neurotrophic factor (BDNF), are severely affected in Alzheimer's disease (AD), but their administration faces a myriad of technical challenges. Here we took advantage of the early astrogliosis observed in an amyloid mouse model of AD (5xFAD) and used it as an internal sensor to administer BDNF conditionally and locally. We first demonstrate the relevance of BDNF release from astrocytes by evaluating the effects of coculturing WT neurons and BDNF-deficient astrocytes. Next, we crossed 5xFAD mice with pGFAP:BDNF mice (only males were used) to create 5xFAD mice that overexpress BDNF when and where astrogliosis is initiated (5xF:pGB mice). We evaluated the behavioral phenotype of these mice. We first found that BDNF from astrocytes is crucial for dendrite outgrowth and spine number in cultured WT neurons. Double-mutant 5xF:pGB mice displayed improvements in cognitive tasks compared with 5xFAD littermates. In these mice, there was a rescue of BDNF/TrkB downstream signaling activity associated with an improvement of dendritic spine density and morphology. Clusters of synaptic markers, PSD-95 and synaptophysin, were also recovered in 5xF:pGB compared with 5xFAD mice as well as the number of presynaptic vesicles at excitatory synapses. Additionally, experimentally evoked LTP in vivo was increased in 5xF:pGB mice. The beneficial effects of conditional BDNF production and local delivery at the location of active neuropathology highlight the potential to use endogenous biomarkers with early onset, such as astrogliosis, as regulators of neurotrophic therapy in AD.SIGNIFICANCE STATEMENT Recent evidence places astrocytes as pivotal players during synaptic plasticity and memory processes. In the present work, we first provide evidence that astrocytes are essential for neuronal morphology via BDNF release. We then crossed transgenic mice (5xFAD mice) with the transgenic pGFAP-BDNF mice, which express BDNF under the GFAP promoter. The resultant double-mutant mice 5xF:pGB mice displayed a full rescue of hippocampal BDNF loss and related signaling compared with 5xFAD mice and a significant and specific improvement in all the evaluated cognitive tasks. These improvements did not correlate with amelioration of β amyloid load or hippocampal adult neurogenesis rate but were accompanied by a dramatic recovery of structural and functional synaptic plasticity.
Collapse
|
972
|
Zhu W, Chen X, Ning L, Jin K. Network Analysis Reveals TNF as a Major Hub of Reactive Inflammation Following Spinal Cord Injury. Sci Rep 2019; 9:928. [PMID: 30700814 PMCID: PMC6354014 DOI: 10.1038/s41598-018-37357-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023] Open
Abstract
Spinal cord injury (SCI) leads to reactive inflammation and other harmful events that limit spinal cord regeneration. We propose an approach for studying the mechanisms at the levels of network topology, gene ontology, signaling pathways, and disease inference. We treated inflammatory mediators as toxic chemicals and retrieved the genes and interacting proteins associated with them via a set of biological medical databases and software. We identified >10,000 genes associated with SCI. Tumor necrosis factor (TNF) had the highest scores, and the top 30 were adopted as core data. In the core interacting protein network, TNF and other top 10 nodes were the major hubs. The core members were involved in cellular responses and metabolic processes, as components of the extracellular space and regions, in protein-binding and receptor-binding functions, as well as in the TNF signaling pathway. In addition, both seizures and SCI were highly associated with TNF levels; therefore, for achieving a better curative effect on SCI, TNF and other major hubs should be targeted together according to the theory of network intervention, rather than a single target such as TNF alone. Furthermore, certain drugs used to treat epilepsy could be used to treat SCI as adjuvants.
Collapse
Affiliation(s)
- Weiping Zhu
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, 200072, P. R. China.
| | - Xuning Chen
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, 200072, P. R. China
| | - Le Ning
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, 200072, P. R. China
| | - Kan Jin
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, 200072, P. R. China
| |
Collapse
|
973
|
State of Astrocytes in the Mice Brain under Conditions of Herpes Viral Infection and Modeled Stroke. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09757-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
974
|
Wheeler MA, Jaronen M, Covacu R, Zandee SEJ, Scalisi G, Rothhammer V, Tjon EC, Chao CC, Kenison JE, Blain M, Rao VTS, Hewson P, Barroso A, Gutiérrez-Vázquez C, Prat A, Antel JP, Hauser R, Quintana FJ. Environmental Control of Astrocyte Pathogenic Activities in CNS Inflammation. Cell 2019; 176:581-596.e18. [PMID: 30661753 PMCID: PMC6440749 DOI: 10.1016/j.cell.2018.12.012] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/01/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
Genome-wide studies have identified genetic variants linked to neurologic diseases. Environmental factors also play important roles, but no methods are available for their comprehensive investigation. We developed an approach that combines genomic data, screens in a novel zebrafish model, computational modeling, perturbation studies, and multiple sclerosis (MS) patient samples to evaluate the effects of environmental exposure on CNS inflammation. We found that the herbicide linuron amplifies astrocyte pro-inflammatory activities by activating signaling via sigma receptor 1, inositol-requiring enzyme-1α (IRE1α), and X-box binding protein 1 (XBP1). Indeed, astrocyte-specific shRNA- and CRISPR/Cas9-driven gene inactivation combined with RNA-seq, ATAC-seq, ChIP-seq, and study of patient samples suggest that IRE1α-XBP1 signaling promotes CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, MS. In summary, these studies define environmental mechanisms that control astrocyte pathogenic activities and establish a multidisciplinary approach for the systematic investigation of the effects of environmental exposure in neurologic disorders.
Collapse
Affiliation(s)
- Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Merja Jaronen
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ruxandra Covacu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie E J Zandee
- Neuroimmunology Research Lab, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Giulia Scalisi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica E Kenison
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Manon Blain
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Vijayaraghava T S Rao
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Patrick Hewson
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andreia Barroso
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Cristina Gutiérrez-Vázquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandre Prat
- Neuroimmunology Research Lab, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Russ Hauser
- Department of Epidemiology and Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
975
|
Koyama Y, Sumie S, Nakano Y, Nagao T, Tokumaru S, Michinaga S. Endothelin-1 stimulates expression of cyclin D1 and S-phase kinase-associated protein 2 by activating the transcription factor STAT3 in cultured rat astrocytes. J Biol Chem 2019; 294:3920-3933. [PMID: 30670587 DOI: 10.1074/jbc.ra118.005614] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/17/2019] [Indexed: 01/04/2023] Open
Abstract
Brain injury-mediated induction of reactive astrocytes often leads to glial scar formation in damaged brain regions. Activation of signal transducer and activator of transcription 3 (STAT3), a member of the STAT family of transcription factors, plays a pivotal role in inducing reactive astrocytes and glial scar formation. Endothelin-1 (ET-1) is a vasoconstrictor peptide, and its levels increase in brain disorders and promote astrocytic proliferation through ETB receptors. To clarify the mechanisms underlying ET-1-mediated astrocytic proliferation, here we examined its effects on STAT3 in cultured rat astrocytes. ET-1 treatment stimulated Ser-727 phosphorylation of STAT3 in the astrocytes, but Tyr-705 phosphorylation was unaffected, and ET-induced STAT3 Ser-727 phosphorylation was reduced by the ETB antagonist BQ788. ET-1 stimulated STAT3 binding to its consensus DNA-binding motifs. Monitoring G1/S phase cell cycle transition through bromodeoxyuridine (BrdU) incorporation, we found that ET-1 increases BrdU incorporation into the astrocytic nucleus, indicating cell cycle progression. Of note, STAT3 chemical inhibition (with stattic or 5,15-diphenyl-porphine (5,15-DPP)) or siRNA-mediated STAT3 silencing reduced ET-induced BrdU incorporation. Moreover, ET-1 increased astrocytic expression levels of cyclin D1 and S-phase kinase-associated protein 2 (SKP2), which were reduced by stattic, 5,15-DPP, and STAT3 siRNA. ChIP-based PCR analysis revealed that ET-1 promotes the binding of SAT3 to the 5'-flanking regions of rat cyclin D1 and SKP2 genes. Our results suggest that STAT3-mediated regulation of cyclin D1 and SKP2 expression underlies ET-induced astrocytic proliferation.
Collapse
Affiliation(s)
- Yutaka Koyama
- From the Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan and
| | - Satoshi Sumie
- the Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka 584-8540, Japan
| | - Yasutaka Nakano
- the Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka 584-8540, Japan
| | - Tomoya Nagao
- the Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka 584-8540, Japan
| | - Shiho Tokumaru
- the Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka 584-8540, Japan
| | - Shotaro Michinaga
- the Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka 584-8540, Japan
| |
Collapse
|
976
|
Repetitive Diffuse Mild Traumatic Brain Injury Causes an Atypical Astrocyte Response and Spontaneous Recurrent Seizures. J Neurosci 2019; 39:1944-1963. [PMID: 30665946 DOI: 10.1523/jneurosci.1067-18.2018] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 11/21/2022] Open
Abstract
Focal traumatic brain injury (TBI) induces astrogliosis, a process essential to protecting uninjured brain areas from secondary damage. However, astrogliosis can cause loss of astrocyte homeostatic functions and possibly contributes to comorbidities such as posttraumatic epilepsy (PTE). Scar-forming astrocytes seal focal injuries off from healthy brain tissue. It is these glial scars that are associated with epilepsy originating in the cerebral cortex and hippocampus. However, the vast majority of human TBIs also present with diffuse brain injury caused by acceleration-deceleration forces leading to tissue shearing. The resulting diffuse tissue damage may be intrinsically different from focal lesions that would trigger glial scar formation. Here, we used mice of both sexes in a model of repetitive mild/concussive closed-head TBI, which only induced diffuse injury, to test the hypothesis that astrocytes respond uniquely to diffuse TBI and that diffuse TBI is sufficient to cause PTE. Astrocytes did not form scars and classic astrogliosis characterized by upregulation of glial fibrillary acidic protein was limited. Surprisingly, an unrelated population of atypical reactive astrocytes was characterized by the lack of glial fibrillary acidic protein expression, rapid and sustained downregulation of homeostatic proteins and impaired astrocyte coupling. After a latency period, a subset of mice developed spontaneous recurrent seizures reminiscent of PTE in human TBI patients. Seizing mice had larger areas of atypical astrocytes compared with nonseizing mice, suggesting that these atypical astrocytes might contribute to epileptogenesis after diffuse TBI.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is a leading cause of acquired epilepsies. Reactive astrocytes have long been associated with seizures and epilepsy in patients, particularly after focal/lesional brain injury. However, most TBIs also include nonfocal, diffuse injuries. Here, we showed that repetitive diffuse TBI is sufficient for the development of spontaneous recurrent seizures in a subset of mice. We identified an atypical response of astrocytes induced by diffuse TBI characterized by the rapid loss of homeostatic proteins and lack of astrocyte coupling while reactive astrocyte markers or glial scar formation was absent. Areas with atypical astrocytes were larger in animals that later developed seizures suggesting that this response may be one root cause of epileptogenesis after diffuse TBI.
Collapse
|
977
|
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
|
978
|
Vidal PM, Pacheco R. Targeting the Dopaminergic System in Autoimmunity. J Neuroimmune Pharmacol 2019; 15:57-73. [PMID: 30661214 DOI: 10.1007/s11481-019-09834-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
Dopamine has emerged as a fundamental regulator of inflammation. In this regard, it has been shown that dopaminergic signalling pathways are key players promoting homeostasis between the central nervous system and the immune system. Dysregulation in the dopaminergic system affects both innate and adaptive immunity, contributing to the development of numerous autoimmune and inflammatory pathologies. This makes dopamine receptors interesting therapeutic targets for either the development of new treatments or repurposing of already available pharmacological drugs. Dopamine receptors are broadly expressed on different immune cells with multifunctional effects depending on the dopamine concentration available and the pattern of expression of five dopamine receptors displaying different affinities for dopamine. Thus, impaired dopaminergic signalling through different dopamine receptors may result in altered behaviour of immunity, contributing to the development and progression of autoimmune pathologies. In this review we discuss the current evidence involving the dopaminergic system in inflammatory bowel disease, multiple sclerosis and Parkinson's disease. In addition, we summarise and analyse the therapeutic approaches designed to attenuate disease development and progression by targeting the dopaminergic system. Graphical Abstract Targetting the dopaminergic system in autoimmunity. Effector T-cells (Teff) orchestrate inflamamtion involved in autoimmunity, whilst regulatory T-cells (Tregs) suppress Teff activity promoting tolerance to self-constituents. Dopamine has emerged as a key regulator of Teff and Tregs function, thereby dopamine receptors have becoming important therapeutic targets in autoimmune disorders, especially in those affecting the brain and the gut, where dopamine levels strongly change with inflammation.
Collapse
Affiliation(s)
- Pia M Vidal
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, Ñuñoa, 7780272, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, Ñuñoa, 7780272, Santiago, Chile. .,Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, 8370146, Santiago, Chile.
| |
Collapse
|
979
|
He Z, Zang H, Zhu L, Huang K, Yi T, Zhang S, Cheng S. An anti-inflammatory peptide and brain-derived neurotrophic factor-modified hyaluronan-methylcellulose hydrogel promotes nerve regeneration in rats with spinal cord injury. Int J Nanomedicine 2019; 14:721-732. [PMID: 30705588 PMCID: PMC6342221 DOI: 10.2147/ijn.s187854] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Traumatic spinal cord injury (SCI) causes neuronal death, demyelination, axonal degeneration, inflammation, glial scar formation, and cystic cavitation resulting in interruption of neural signaling and loss of nerve function. Multifactorial targeted therapy is a promising strategy for SCI. Methods The anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK) and brain-derived neurotrophic factor (BDNF)-modified hyaluronan-methylcellulose (HAMC) hydrogel was designed for minimally invasive, localized, and sustained intrathecal protein delivery. The physical and biological characteristics of HAMC-KAFAK/BDNF hydrogel were measured in vitro. SCI model was performed in rats and HAMC-KAFAK/BDNF hydrogel was injected into the injured site of spinal cord. The neuronal regeneration effect was evaluated by inflammatory cytokine levels, behavioral test and histological analysis at 8 weeks post operation. Results HAMC-KAFAK/BDNF hydrogel showed minimally swelling property and sustained release of the KAFAK and BDNF. HAMC-KAFAK/BDNF hydrogel significantly improved the proliferation of PC12 cells in vitro without cytotoxicity. Significant recovery in both neurological function and nerve tissue morphology in SCI rats were observed in HAMC-KAFAK/BDNF group. HAMC-KAFAK/BDNF group showed significant reduction in proinflammatory cytokines expression and cystic cavitation, decreased glial scar formation, and improved neuronal survival in the rat SCI model compared to HAMC group and SCI group. Conclusion The HAMC-KAFAK/BDNF hydrogel promotes functional recovery of rats with spinal cord injury by regulating inflammatory cytokine levels and improving axonal regeneration.
Collapse
Affiliation(s)
- Zhijiang He
- Logistics University of Chinese People's Armed Police Force (PAP), Tianjin 300309, China
| | - Hongxin Zang
- Department of Nursing, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, China
| | - Lei Zhu
- Department of Orthopaedics Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, China
| | - Kui Huang
- Logistics University of Chinese People's Armed Police Force (PAP), Tianjin 300309, China
| | - Tailong Yi
- Institute of TBI and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin Key Laboratory of Neurotrauma Repair, Tianjin 300162, China, ;
| | - Sai Zhang
- Institute of TBI and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin Key Laboratory of Neurotrauma Repair, Tianjin 300162, China, ;
| | - Shixiang Cheng
- Institute of TBI and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin Key Laboratory of Neurotrauma Repair, Tianjin 300162, China, ;
| |
Collapse
|
980
|
Karova K, Wainwright JV, Machova-Urdzikova L, Pisal RV, Schmidt M, Jendelova P, Jhanwar-Uniyal M. Transplantation of neural precursors generated from spinal progenitor cells reduces inflammation in spinal cord injury via NF-κB pathway inhibition. J Neuroinflammation 2019; 16:12. [PMID: 30654804 PMCID: PMC6335809 DOI: 10.1186/s12974-019-1394-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/02/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) triggers a chain of events that is accompanied by an inflammatory reaction leading to necrotic cell death at the core of the injury site, which is restricted by astrogliosis and apoptotic cell death in the surrounding areas. Activation of nuclear factor-κB (NF-κB) signaling pathway has been shown to be associated with inflammatory response induced by SCI. Here, we elucidate the pattern of activation of NF-κB in the pathology of SCI in rats and investigate the effect of transplantation of spinal neural precursors (SPC-01) on its activity and related astrogliosis. METHODS Using a rat compression model of SCI, we transplanted SPC-01 cells or injected saline into the lesion 7 days after SCI induction. Paraffin-embedded sections were used to assess p65 NF-κB nuclear translocation at days 1, 3, 7, 10, 14, and 28 and to determine levels of glial scaring, white and gray matter preservation, and cavity size at day 28 after SCI. Additionally, levels of p65 phosphorylated at Serine536 were determined 10, 14, and 28 days after SCI as well as levels of locally secreted TNF-α. RESULTS We determined a bimodal activation pattern of canonical p65 NF-κB signaling pathway in the pathology of SCI with peaks at 3 and 28 days after injury induction. Transplantation of SCI-01 cells resulted in significant downregulation of TNF-α production at 10 and 14 days after SCI and in strong inhibition of p65 NF-κB activity at 28 days after SCI, mainly in the gray matter. Moreover, reduced formation of glial scar was found in SPC-01-transplanted rats along with enhanced gray matter preservation and reduced cavity size. CONCLUSIONS The results of this study demonstrate strong immunomodulatory properties of SPC-01 cells based on inhibition of a major signaling pathway. Canonical NF-κB pathway activation underlines much of the immune response after SCI including cytokine, chemokine, and apoptosis-related factor production as well as immune cell activation and infiltration. Reduced inflammation may have led to observed tissue sparing. Additionally, such immune response modulation could have impacted astrocyte activation resulting in a reduced glial scar.
Collapse
Affiliation(s)
- Kristyna Karova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | | | - Lucia Machova-Urdzikova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Rishikaysh V Pisal
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Meic Schmidt
- New York Medical College, Valhalla, NY, 10595, USA
| | - Pavla Jendelova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic. .,2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06, Prague, Czech Republic.
| | | |
Collapse
|
981
|
Koffler J, Zhu W, Qu X, Platoshyn O, Dulin JN, Brock J, Graham L, Lu P, Sakamoto J, Marsala M, Chen S, Tuszynski MH. Biomimetic 3D-printed scaffolds for spinal cord injury repair. Nat Med 2019; 25:263-269. [PMID: 30643285 DOI: 10.1038/s41591-018-0296-z] [Citation(s) in RCA: 390] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 11/08/2018] [Indexed: 12/22/2022]
Abstract
Current methods for bioprinting functional tissue lack appropriate biofabrication techniques to build complex 3D microarchitectures essential for guiding cell growth and promoting tissue maturation1. 3D printing of central nervous system (CNS) structures has not been accomplished, possibly owing to the complexity of CNS architecture. Here, we report the use of a microscale continuous projection printing method (μCPP) to create a complex CNS structure for regenerative medicine applications in the spinal cord. μCPP can print 3D biomimetic hydrogel scaffolds tailored to the dimensions of the rodent spinal cord in 1.6 s and is scalable to human spinal cord sizes and lesion geometries. We tested the ability of µCPP 3D-printed scaffolds loaded with neural progenitor cells (NPCs) to support axon regeneration and form new 'neural relays' across sites of complete spinal cord injury in vivo in rodents1,2. We find that injured host axons regenerate into 3D biomimetic scaffolds and synapse onto NPCs implanted into the device and that implanted NPCs in turn extend axons out of the scaffold and into the host spinal cord below the injury to restore synaptic transmission and significantly improve functional outcomes. Thus, 3D biomimetic scaffolds offer a means of enhancing CNS regeneration through precision medicine.
Collapse
Affiliation(s)
- Jacob Koffler
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA.
| | - Wei Zhu
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
| | - Xin Qu
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
| | - Oleksandr Platoshyn
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - Jennifer N Dulin
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
| | - John Brock
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
| | - Lori Graham
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
| | - Paul Lu
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
| | - Jeff Sakamoto
- Mechanical Engineering Department, University of Michigan, Ann Arbor, MI, USA
| | - Martin Marsala
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA.
| | - Mark H Tuszynski
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA. .,Veterans Affairs Medical Center, San Diego, CA, USA.
| |
Collapse
|
982
|
Astrocyte Signaling in the Neurovascular Unit After Central Nervous System Injury. Int J Mol Sci 2019; 20:ijms20020282. [PMID: 30642007 PMCID: PMC6358919 DOI: 10.3390/ijms20020282] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 12/20/2022] Open
Abstract
Astrocytes comprise the major non-neuronal cell population in the mammalian neurovascular unit. Traditionally, astrocytes are known to play broad roles in central nervous system (CNS) homeostasis, including the management of extracellular ion balance and pH, regulation of neurotransmission, and control of cerebral blood flow and metabolism. After CNS injury, cell–cell signaling between neuronal, glial, and vascular cells contribute to repair and recovery in the neurovascular unit. In this mini-review, we propose the idea that astrocytes play a central role in organizing these signals. During CNS recovery, reactive astrocytes communicate with almost all CNS cells and peripheral progenitors, resulting in the promotion of neurogenesis and angiogenesis, regulation of inflammatory response, and modulation of stem/progenitor response. Reciprocally, changes in neurons and vascular components of the remodeling brain should also influence astrocyte signaling. Therefore, understanding the complex and interdependent signaling pathways of reactive astrocytes after CNS injury may reveal fundamental mechanisms and targets for re-integrating the neurovascular unit and augmenting brain recovery.
Collapse
|
983
|
Complement protein levels in plasma astrocyte-derived exosomes are abnormal in conversion from mild cognitive impairment to Alzheimer's disease dementia. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:61-66. [PMID: 31032394 PMCID: PMC6477776 DOI: 10.1016/j.dadm.2018.11.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction Levels of complement proteins (CPs) in plasma astrocyte-derived exosomes (ADEs) that are abnormal in Alzheimer's disease (AD) have not been assessed in mild cognitive impairment (MCI). Methods Participants (n = 20 per group) had either MCI converting to dementia within 3 years (MCIC), MCI remaining stable over 3 years (MCIS), Alzheimer's disease, or were controls. CPs of ADEs isolated from plasmas by anti-human glutamine aspartate transporter antibody absorption were quantified by ELISAs. Results ADE levels of C1q and C4b of the classical pathway, factor D and fragment Bb of the alternative pathway, and C5b, C3b, and C5b-C9 of both pathways were significantly higher in patients with MCIC than those with MCIS. ADE levels of inhibitory CPs decay-accelerating factor, CD46, CD59, and type 1 complement receptor were significantly lower in patients with MCIC than those with MCIS. Discussion ADE CPs are components of neurotoxic neuroinflammation that may be predictive biomarkers of MCI conversion to Alzheimer's disease.
Collapse
|
984
|
Comparison between Polybutylcyanoacrylate Nanoparticles with Either Surface-Adsorbed or Encapsulated Brain-Derived Neurotrophic Factor on the Neural Differentiation of iPSCs. Int J Mol Sci 2019; 20:ijms20010182. [PMID: 30621332 PMCID: PMC6337453 DOI: 10.3390/ijms20010182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/22/2018] [Accepted: 12/31/2018] [Indexed: 01/18/2023] Open
Abstract
The brain-derived neurotrophic factor (BDNF) is vital in the neural differentiation of neural stem/progenitor cells, and together may have therapeutic potential for neural regeneration. In this study, a multiplexed polybutylcyanoacrylate nanoparticle (PBCA NP) delivery platform was constructed, incorporating either surface-adsorbed or encapsulated BDNF for the induction of neural differentiation in induced pleuripotent stem cells (iPSCs), where tween 80 (T80) and superparamagnetic iron oxide (SPIO) were added for central nervous system (CNS) targeting and magnetic resonance (MR) image tracking, respectively. Both methods by which the BDNF was carried resulted in loading efficiencies greater than 95%. The nanoparticle-mediated delivery of BDNF resulted in neural differentiation of iPSCs detected on immunofluorescence staining as early as 7 days, with enhanced differentiation efficiency by 1.3-fold compared to the control on flow cytometry; the delivery system of surface-adsorbed BDNF gave rise to cells that had the best neural development than the encapsulated formulation. T80-coating disrupted the in vitro blood–brain barrier model with a corresponding 1.5- to two-fold increase in permeability. SPIO-loaded PBCA NPs exhibited a concentration-dependent, rapid decay in signal intensity on the phantom MR experiment. This study demonstrates the versatility of the PBCA NP, and the surface-adsorption of BDNF is the preferred method of delivery for the differentiation of iPSCs.
Collapse
|
985
|
Abstract
Astrocytes play complex roles in health and disease. Here, we review recent findings on molecular pathways that control astrocyte function in multiple sclerosis (MS) as well as new tools for their investigation. In particular, we describe positive and negative regulators of astrocyte-mediated pathogenesis in MS, such as sphingolipid metabolism and aryl hydrocarbon receptor signaling, respectively. In addition, we also discuss the issue of astrocyte heterogeneity and its relevance for the contribution of astrocytes to MS pathogenesis. Finally, we discuss how new genomic tools could transform the study of astrocyte biology in MS.
Collapse
|
986
|
Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 2019; 565:246-250. [PMID: 30602786 DOI: 10.1038/s41586-018-0824-5] [Citation(s) in RCA: 493] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 11/05/2018] [Indexed: 11/08/2022]
Abstract
In addition to maintaining immune tolerance, FOXP3+ regulatory T (Treg) cells perform specialized functions in tissue homeostasis and remodelling1,2. However, the characteristics and functions of brain Treg cells are not well understood because there is a low number of Treg cells in the brain under normal conditions. Here we show that there is massive accumulation of Treg cells in the mouse brain after ischaemic stroke, and this potentiates neurological recovery during the chronic phase of ischaemic brain injury. Although brain Treg cells are similar to Treg cells in other tissues such as visceral adipose tissue and muscle3-5, they are apparently distinct and express unique genes related to the nervous system including Htr7, which encodes the serotonin receptor 5-HT7. The amplification of brain Treg cells is dependent on interleukin (IL)-2, IL-33, serotonin and T cell receptor recognition, and infiltration into the brain is driven by the chemokines CCL1 and CCL20. Brain Treg cells suppress neurotoxic astrogliosis by producing amphiregulin, a low-affinity epidermal growth factor receptor (EGFR) ligand. Stroke is a leading cause of neurological disability, and there are currently few effective recovery methods other than rehabilitation during the chronic phase. Our findings suggest that Treg cells and their products may provide therapeutic opportunities for neuronal protection against stroke and neuroinflammatory diseases.
Collapse
|
987
|
Ashammakhi N, Ahadian S, Darabi MA, El Tahchi M, Lee J, Suthiwanich K, Sheikhi A, Dokmeci MR, Oklu R, Khademhosseini A. Minimally Invasive and Regenerative Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804041. [PMID: 30565732 PMCID: PMC6709364 DOI: 10.1002/adma.201804041] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/20/2018] [Indexed: 05/03/2023]
Abstract
Advances in biomaterial synthesis and fabrication, stem cell biology, bioimaging, microsurgery procedures, and microscale technologies have made minimally invasive therapeutics a viable tool in regenerative medicine. Therapeutics, herein defined as cells, biomaterials, biomolecules, and their combinations, can be delivered in a minimally invasive way to regenerate different tissues in the body, such as bone, cartilage, pancreas, cardiac, skeletal muscle, liver, skin, and neural tissues. Sophisticated methods of tracking, sensing, and stimulation of therapeutics in vivo using nano-biomaterials and soft bioelectronic devices provide great opportunities to further develop minimally invasive and regenerative therapeutics (MIRET). In general, minimally invasive delivery methods offer high yield with low risk of complications and reduced costs compared to conventional delivery methods. Here, minimally invasive approaches for delivering regenerative therapeutics into the body are reviewed. The use of MIRET to treat different tissues and organs is described. Although some clinical trials have been performed using MIRET, it is hoped that such therapeutics find wider applications to treat patients. Finally, some future perspective and challenges for this emerging field are highlighted.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mario El Tahchi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- LBMI, Department of Physics, Lebanese University - Faculty of Sciences 2, PO Box 90656, Jdeidet, Lebanon
| | - Junmin Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Kasinan Suthiwanich
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Amir Sheikhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, California, USA
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
988
|
Ma C, Zhang P, Shen Y. Progress in research into spinal cord injury repair: Tissue engineering scaffolds and cell transdifferentiation. JOURNAL OF NEURORESTORATOLOGY 2019. [DOI: 10.26599/jnr.2019.9040024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
As with all tissues of the central nervous system, the low regeneration ability of spinal cord tissue after injury decreases the potential for repair and recovery. Initially, in spinal cord injuries (SCI), often the surgeon can only limit further damage by early surgical decompression. However, with the development of basic science, especially the development of genetic engineering, molecular biology, tissue engineering, and materials science, some promising progress has been made in promoting the repair of central nervous system injuries. For example, transplantation of neural stem cells (NSCs), olfactory ensheathing cells (OECs), and gene- mediated transdifferentiation to repair central nervous system injury. This paper summarizes the progress and prospects of SCI repair with tissue engineering scaffold and cell transdifferentiation from an extensive literatures.
Collapse
|
989
|
Gomes ED, Silva NA, Salgado AJ. Combinatorial therapies for spinal cord injury: strategies to induce regeneration. Neural Regen Res 2019; 14:69-71. [PMID: 30531075 PMCID: PMC6262995 DOI: 10.4103/1673-5374.243705] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
990
|
Tsui C, Koss K, Churchward MA, Todd KG. Biomaterials and glia: Progress on designs to modulate neuroinflammation. Acta Biomater 2019; 83:13-28. [PMID: 30414483 DOI: 10.1016/j.actbio.2018.11.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/05/2018] [Accepted: 11/06/2018] [Indexed: 02/06/2023]
Abstract
Microglia are multi-functional cells that play a vital role in establishing and maintaining the function of the nervous system and determining the fate of neurons following injury or neuropathology. The roles of microglia are diverse and essential to the capacity of the nervous system to recover from injury, however sustained inflammation can limit recovery and drive chronic disease processes such as neurodegenerative disorders. When assessing implantable therapeutic devices in the central nervous system, an improved lifetime of the implant is considered achievable through the attenuation of microglial inflammation. Consequently, there is a tremendous underexplored potential in biomaterial and engineered design to modulate neuroinflammation for therapeutic benefit. Several strategies for improving device compatibility reviewed here include: biocompatible coatings, improved designs in finer and flexible shapes to reduce tissue shear-related scarring, and loading of anti-inflammatory drugs. Studies about microglial cell cultures in 3D hydrogels and nanoscaffolds to assess various injuries and disorders are also discussed. A variety of other microglia-targeting treatments are also reviewed, including nanoparticulate systems, cellular backpacks, and gold plinths, with the intention of delivering anti-inflammatory drugs by targeting the phagocytic nature of microglia. Overall, this review highlights recent advances in biomaterials targeting microglia and inflammatory function with the potential for improving implant rejection and biocompatibility studies. STATEMENT OF SIGNIFICANCE: Microglia are the resident immune cells of the central nervous system, and thus play a central role in the neuroinflammatory response against conditions than span acute injuries, neuropsychiatric disorders, and neurodegenerative disorders. This review article presents a summary of biomaterials research that target microglia and other glial cells in order to attenuate neuroinflammation, including but not limited to: design of mechanically compliant and biocompatible stimulation electrodes, hydrogels for high-throughput 3D modelling of nervous tissue, and uptake of nanoparticle drug delivery systems. The goal of this paper is to identify strengths and gaps in the relevant literature, and to promote further consideration of microglia behaviour and neuroinflammation in biomaterial design.
Collapse
Affiliation(s)
- C Tsui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - K Koss
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2R3, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - M A Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2R3, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - K G Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2R3, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
991
|
Yang T, Terrando N. The Evolving Role of Specialized Pro-resolving Mediators in Modulating Neuroinflammation in Perioperative Neurocognitive Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:27-35. [PMID: 31562619 DOI: 10.1007/978-3-030-21735-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Surgery can be a life-saving procedure; however, significant complications may occur after routine procedures especially in older and more frail patients. Perioperative neurocognitive disorders (PNDs), including delirium and postoperative cognitive dysfunction, are the most common complications in older adults following common procedures such as orthopedic or cardiac surgery. The consequences of PNDs can be devastating, with longer in-hospital stay, poorer prognosis, and higher mortality rates. Inflammation is gaining considerable interest as a critical driver of cognitive deficits. In this regard, resolution of inflammation, once thought to be a passive process, may provide novel approaches to treat neuroinflammation and PNDs. Herein we review the role for impaired resolution after surgery and the growing role of specialized pro-resolving mediators (SPMs) in regulating postoperative neuroinflammation and neurological complications after surgery.
Collapse
Affiliation(s)
- Ting Yang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
992
|
Valles SL, Iradi A, Aldasoro M, Vila JM, Aldasoro C, de la Torre J, Campos-Campos J, Jorda A. Function of Glia in Aging and the Brain Diseases. Int J Med Sci 2019; 16:1473-1479. [PMID: 31673239 PMCID: PMC6818212 DOI: 10.7150/ijms.37769] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/18/2019] [Indexed: 12/13/2022] Open
Abstract
Microglia cells during aging, neurodegeneration and neuroinflammation show different morphological and transcriptional profiles (related to axonal direction and cell adhesion). Furthermore, expressions of the receptors on the surface and actin formation compared to young are also different. This review delves into the role of glia during aging and the development of the diseases. The susceptibility of different regions of the brain to disease are linked to the overstimulation of signals related to the immune system during aging, as well as the damaging impact of these cascades on the functionality of different populations of microglia present in each region of the brain. Furthermore, a decrease in microglial phagocytosis has been related to many diseases and also has been detected during aging. In this paper we also describe the role of glia in different illness, such as AD, ALS, pain related disorders, cancer, developmental disorders and the problems produced by opening of the blood brain barrier. Future studies will clarify many points planted by this review.
Collapse
Affiliation(s)
- Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Antonio Iradi
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Martin Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Jose M Vila
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Constanza Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | | | - Juan Campos-Campos
- Department of Nursing, Faculty of Nursing and Podiatry, University of Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Spain.,Department of Nursing, Faculty of Nursing and Podiatry, University of Valencia, Spain
| |
Collapse
|
993
|
Lie ME, Gowing EK, Johansen NB, Dalby NO, Thiesen L, Wellendorph P, Clarkson AN. GAT3 selective substrate l-isoserine upregulates GAT3 expression and increases functional recovery after a focal ischemic stroke in mice. J Cereb Blood Flow Metab 2019; 39:74-88. [PMID: 29160736 PMCID: PMC6311676 DOI: 10.1177/0271678x17744123] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ischemic stroke triggers an elevation in tonic GABA inhibition that impairs the ability of the brain to form new structural and functional cortical circuits required for recovery. This stroke-induced increase in tonic inhibition is caused by impaired GABA uptake via the glial GABA transporter GAT3, highlighting GAT3 as a novel target in stroke recovery. Using a photothrombotic stroke mouse model, we show that GAT3 protein levels are decreased in peri-infarct tissue from 6 h to 42 days post-stroke. Prior studies have shown that GAT substrates can increase GAT surface expression. Therefore, we aimed to assess whether the GAT3 substrate, L-isoserine, could increase post-stroke functional recovery. L-Isoserine (38 µM or 380 µM) administered directly into the infarct from day 5 to 32 post-stroke, significantly increased motor performance in the grid-walking and cylinder tasks in a concentration-dependent manner, without affecting infarct volumes. Additionally, L-isoserine induced a lasting increase in GAT3 expression in peri-infarct regions accompanied by a small decrease in GFAP expression. This study is the first to show that a GAT3 substrate can increase GAT3 expression and functional recovery after focal ischemic stroke following a delayed long-term treatment. We propose that enhancing GAT3-mediated uptake dampens tonic inhibition and promotes functional recovery after stroke.
Collapse
Affiliation(s)
- Maria Ek Lie
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,2 Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Emma K Gowing
- 2 Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Nina B Johansen
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Nils Ole Dalby
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Louise Thiesen
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Andrew N Clarkson
- 2 Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand.,3 Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
994
|
Li H, Zhang X, Qi X, Zhu X, Cheng L. Icariin Inhibits Endoplasmic Reticulum Stress-induced Neuronal Apoptosis after Spinal Cord Injury through Modulating the PI3K/AKT Signaling Pathway. Int J Biol Sci 2019; 15:277-286. [PMID: 30745820 PMCID: PMC6367543 DOI: 10.7150/ijbs.30348] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum (ER) stress-induced neuronal apoptosis is a crucial pathological process of spinal cord injury (SCI). In our previous study, icariin (ICA) showed neuroprotective effects in SCI. However, the relationships between ER stress and ICA in SCI are unclear yet. Therefore, whether ICA could ameliorate SCI via attenuating ER stress was investigated in vitro and in vivo. Adult mice were established SCI model and received vehicle solution or ICA by gavage once per day in vivo. The primary cultured cells were treated with or without thapsigargin (TG), ICA or LY294002 to induce ER stress in vitro. Motor dysfunction, neuronal apoptosis, tissue damage and inhibition of PI3K/AKT pathway were induced by ER stress after SCI. But ICA administration significantly enhanced motor recovery and protected spinal cord tissues against infraction and hemorrhage, etc. post injury. Meanwhile, the expression of ER stress markers ATF6, IRE1α, GRP78, XBP1 and eIF2α was decreased, while the level of p-AKT/AKT was increased by ICA. Furthermore, ICA significantly inhibited the expression of ER stress apoptotic proteins caspase-12, CHOP, Bax/Bcl-2, caspase-9 and caspase-3. Moreover, immunofluorescence double staining indicated that ICA reduced GRP78, CHOP and TUNEL positive neurons following SCI. However, this beneficial effect of ICA was abolished by PI3K/AKT inhibitor LY294002 in vitro. Finally, ICA preserved the ultra-structure of ER by transmission electron microscope histologically. This study suggested that the neuroprotective effect of ICA on motor recovery and neuronal survival was related to attenuating ER stress via PI3K/AKT signaling pathway after SCI.
Collapse
Affiliation(s)
- Haotian Li
- Department of Spine Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of spine and spinal cord injury repair and regeneration (Tongji University), Ministry of Education, Shanghai, China
| | - Xinran Zhang
- School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Xi Qi
- Department of Spine Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of spine and spinal cord injury repair and regeneration (Tongji University), Ministry of Education, Shanghai, China
| | - Xu Zhu
- Department of Spine Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of spine and spinal cord injury repair and regeneration (Tongji University), Ministry of Education, Shanghai, China
| | - Liming Cheng
- Department of Spine Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of spine and spinal cord injury repair and regeneration (Tongji University), Ministry of Education, Shanghai, China
| |
Collapse
|
995
|
Kleene R, Loers G, Jakovcevski I, Mishra B, Schachner M. Histone H1 improves regeneration after mouse spinal cord injury and changes shape and gene expression of cultured astrocytes. Restor Neurol Neurosci 2019; 37:291-313. [PMID: 31227672 DOI: 10.3233/rnn-190903] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We have shown that histone H1 is a binding partner for polysialic acid (PSA) and that it improves functional recovery, axon regrowth/sprouting, and target reinnervation after mouse femoral nerve injury. OBJECTIVE Here, we analyzed whether histone H1 affects functional recovery, axon regrowth/sprouting, and target reinnervation after spinal cord injury of adult mice. Furthermore, we tested in vitro histone H1's effect on astrocytic gene expression, cell shape and migration as well as on cell survival of cultured motoneurons. METHODS We applied histone H1 to compressed spinal cord and determined functional recovery and number of fibrillary acidic protein (GFAP)- and neuron-glial antigen 2 (NG2)- positive glial cells, which contribute to glial scarring. Histone H1's effect on migration of astrocytes, astrocytic gene expression and motoneuronal survival was determined using scratch-wounded astroglial monolayer cultures, astrocyte cultures for microarray analysis, and motoneuron cell culture under oxidative stress conditions, respectively. RESULTS Histone H1 application improves locomotor functions and enhances monoaminergic and cholinergic reinnervation of the spinal cord. Expression levels of GFAP and NG2 around the lesion site were decreased in histone H1-treated mice relative to vehicle-treated mice six weeks after injury. Histone H1 reduced astrocytic migration, changed the shape of GFAP- and NG2-positive glial cells and altered gene expression. Gene ontology enrichment analysis indicated that in particular genes coding for proteins involved in proliferation, differentiation, migration and apoptosis are dysregulated. The up- and down-regulation of distinct genes was confirmed by qPCR and Western blot analysis. Moreover, histone H1 reduced hydrogen peroxide-induced cell death of cultured motoneurons. CONCLUSIONS The combined observations indicate that histone H1 locally applied to the lesion site, improves regeneration after spinal cord injury. Some of these beneficial functions of histone H1 in vivo and in vitro can be attributed to its interaction with PSA-carrying neural cell adhesion molecule.
Collapse
Affiliation(s)
- Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Igor Jakovcevski
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
996
|
Verkhratsky A, Ho MS, Vardjan N, Zorec R, Parpura V. General Pathophysiology of Astroglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:149-179. [PMID: 31583588 PMCID: PMC7188602 DOI: 10.1007/978-981-13-9913-8_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astroglial cells are involved in most if not in all pathologies of the brain. These cells can change the morpho-functional properties in response to pathology or innate changes of these cells can lead to pathologies. Overall pathological changes in astroglia are complex and diverse and often vary with different disease stages. We classify astrogliopathologies into reactive astrogliosis, astrodegeneration with astroglial atrophy and loss of function, and pathological remodelling of astrocytes. Such changes can occur in neurological, neurodevelopmental, metabolic and psychiatric disorders as well as in infection and toxic insults. Mutation in astrocyte-specific genes leads to specific pathologies, such as Alexander disease, which is a leukodystrophy. We discuss changes in astroglia in the pathological context and identify some molecular entities underlying pathology. These entities within astroglia may repent targets for novel therapeutic intervention in the management of brain pathologies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Margaret S Ho
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
997
|
Schackel T, Kumar P, Günther M, Liu S, Brunner M, Sandner B, Puttagunta R, Müller R, Weidner N, Blesch A. Peptides and Astroglia Improve the Regenerative Capacity of Alginate Gels in the Injured Spinal Cord. Tissue Eng Part A 2018; 25:522-537. [PMID: 30351234 DOI: 10.1089/ten.tea.2018.0082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Axonal bridging across a lesion in the injured spinal cord requires a growth substrate and guidance cues. Using alginate hydrogels with capillary channels we show that poly-l-ornithine and laminin can be stably bound and improve cell adhesion and neurite growth in vitro, and axon growth in vivo by enhancing host cell infiltration in the injured spinal cord. Filling of coated hydrogels with postnatal astrocytes further increases short-distance axon growth and results in a continuous astroglial substrate across the host/graft interface. Thus, positively charged bioactive molecules can be stably bound to anisotropic capillary alginate hydrogels and early astrocytes further promote tissue integration.
Collapse
Affiliation(s)
- Thomas Schackel
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Prateek Kumar
- 2 Department of Neurological Surgery and Goodman Campbell Brain and Spine, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Manuel Günther
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Shengwen Liu
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany.,3 Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Manuel Brunner
- 4 Department of Physical and Theoretical Chemistry, University of Regensburg, Regensburg, Germany
| | - Beatrice Sandner
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Rainer Müller
- 4 Department of Physical and Theoretical Chemistry, University of Regensburg, Regensburg, Germany
| | - Norbert Weidner
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Armin Blesch
- 1 Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany.,2 Department of Neurological Surgery and Goodman Campbell Brain and Spine, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
998
|
Dzyubenko E, Manrique-Castano D, Kleinschnitz C, Faissner A, Hermann DM. Role of immune responses for extracellular matrix remodeling in the ischemic brain. Ther Adv Neurol Disord 2018; 11:1756286418818092. [PMID: 30619510 PMCID: PMC6299337 DOI: 10.1177/1756286418818092] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is one of the key components contributing to the devastating outcome of ischemic stroke. Starting with stroke onset, inflammatory processes contribute both to cell damage and tissue remodeling. The early release of alarmins triggers the upregulation of multiple proinflammatory cytokines, resulting in the compromised integrity of the blood–brain barrier. From this moment on, the infiltration of peripheral immune cells, reactive gliosis and extracellular matrix (ECM) alterations become intricately intertwined and act as one unit during the tissue remodeling. While the mechanisms of leukocyte and glia activation are amply reviewed, the field of ECM modification remains as yet under explored. In this review, we focus on the interplay between neuroinflammatory cascades and ECM in the ischemic brain. By summarizing the currently available evidence obtained by in vitro research, animal experimentation and human studies, we aim to propose a new direction for the future investigation of stroke recovery.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Neurology, University Hospital Essen, Essen, Germany
| | | | | | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Hufelandstraße 55, D-45122 Essen, Germany
| |
Collapse
|
999
|
Kaushik R, Morkovin E, Schneeberg J, Confettura AD, Kreutz MR, Senkov O, Dityatev A. Traditional Japanese Herbal Medicine Yokukansan Targets Distinct but Overlapping Mechanisms in Aged Mice and in the 5xFAD Mouse Model of Alzheimer's Disease. Front Aging Neurosci 2018; 10:411. [PMID: 30631278 PMCID: PMC6315162 DOI: 10.3389/fnagi.2018.00411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 11/28/2018] [Indexed: 12/18/2022] Open
Abstract
Yokukansan (YKS) is a traditional Japanese herbal medicine that has been used in humans for the treatment of several neurological conditions, such as age-related anxiety and behavioral and psychological symptoms (BPSD) related to multiple forms of dementia, including Alzheimer’s disease (AD). However, the cellular and molecular mechanisms targeted by YKS in the brain are not completely understood. Here, we compared the efficacy of YKS in ameliorating the age- and early-onset familial AD-related behavioral and cellular defects in two groups of animals: 18- to 22-month-old C57BL6/J wild-type mice and 6- to 9-month-old 5xFAD mice, as a transgenic mouse model of this form of AD. Animals were fed food pellets that contained YKS or vehicle. After 1–2 months of YKS treatment, we evaluated the cognitive improvements in both the aged and 5xFAD transgenic mice, and their brain tissues were further investigated to assess the molecular and cellular changes that occurred following YKS intake. Our results show that both the aged and 5xFAD mice exhibited impaired behavioral performance in novel object recognition and contextual fear conditioning (CFC) tasks, which was significantly improved by YKS. Further analyses of the brain tissue from these animals indicated that in aged mice, this improvement was associated with a reduction in astrogliosis, microglia activation and downregulation of the extracellular matrix (ECM), whereas in 5xFAD mice, none of these mechanisms were evident. These results show the differential action of YKS in healthy aged and 5xFAD mice. However, both aged and 5xFAD YKS-treated mice showed increased neuroprotective signaling through protein kinase B/Akt as the common mode of action. Our data suggest that YKS may impart its beneficial effects through Akt signaling in both 5xFAD mice and aged mice, with multiple additional mechanisms potentially contributing to its beneficial effects in aged animals.
Collapse
Affiliation(s)
- Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Evgeny Morkovin
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Department of Fundamental Medicine and Biology, Volgograd State Medical University (VSMU), Volgograd, Russia.,Laboratory of Genomic and Proteomic Research, Volgograd Medical Research Center (VMRC), Volgograd, Russia
| | - Jenny Schneeberg
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | | | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology (LG), Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Hamburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Oleg Senkov
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
1000
|
Guttenplan KA, Liddelow SA. Astrocytes and microglia: Models and tools. J Exp Med 2018; 216:71-83. [PMID: 30541903 PMCID: PMC6314517 DOI: 10.1084/jem.20180200] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/16/2018] [Accepted: 11/26/2018] [Indexed: 01/05/2023] Open
Abstract
An amazing array of tools both old and new are available to investigate the function of astrocytes and microglia. Guttenplan and Liddelow discuss tools available to study the physiology and pathophysiology of these cells both in vivo and in culture systems. Glial cells serve as fundamental regulators of the central nervous system in development, homeostasis, and disease. Discoveries into the function of these cells have fueled excitement in glial research, with enthusiastic researchers addressing fundamental questions about glial biology and producing new scientific tools for the community. Here, we outline the pros and cons of in vivo and in vitro techniques to study astrocytes and microglia with the goal of helping researchers quickly identify the best approach for a given research question in the context of glial biology. It is truly a great time to be a glial biologist.
Collapse
Affiliation(s)
| | - Shane A Liddelow
- Neuroscience Institute, NYU Langone Medical Center, New York, NY.,Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|