301
|
Mine Y, Momiyama T, Hayashi T, Kawase T. Grafted Miniature-Swine Neural Stem Cells of Early Embryonic Mesencephalic Neuroepithelial Origin can Repair the Damaged Neural Circuitry of Parkinson's Disease Model Rats. Neuroscience 2018; 386:51-67. [PMID: 29932984 DOI: 10.1016/j.neuroscience.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 06/02/2018] [Accepted: 06/04/2018] [Indexed: 12/21/2022]
Abstract
Although recent progress in the use of human iPS cell-derived midbrain dopaminergic progenitors is remarkable, alternatives are essential in the strategies of treatment of basal-ganglia-related diseases. Attention has been focused on neural stem cells (NSCs) as one of the possible candidates of donor material for neural transplantation, because of their multipotency and self-renewal characteristics. In the present study, miniature-swine (mini-swine) mesencephalic neuroepithelial stem cells (M-NESCs) of embryonic 17 and 18 days grafted in the parkinsonian rat striatum were assessed immunohistochemically, behaviorally and electrophysiologically to confirm their feasibility for the neural xenografting as a donor material. Grafted mini-swine M-NESCs survived in parkinsonian rat striatum at 8 weeks after transplantation and many of them differentiated into tyrosine hydroxylase (TH)-positive cells. The parkinsonian model rats grafted with mini-swine M-NESCs exhibited a functional recovery from their parkinsonian behavioral defects. The majority of donor-derived TH-positive cells exhibited a matured morphology at 8 weeks. Whole-cell recordings from donor-derived neurons in the host rat brain slices incorporating the graft revealed the presence of multiple types of neurons including dopaminergic. Glutamatergic and GABAergic post-synaptic currents were evoked in the donor-derived cells by stimulation of the host site, suggesting they receive both excitatory and inhibitory synaptic inputs from host area. The present study shows that non-rodent mammalian M-NESCs can differentiate into functionally active neurons in the diseased xenogeneic environment and could improve the parkinsonian behavioral defects over the species. Neuroepithelial stem cells could be an attractive candidate as a source of donor material for neural transplantation.
Collapse
Affiliation(s)
- Yutaka Mine
- Department of Neurosurgery and Endovascular Surgery, Brain Nerve Center, Saiseikai Yokohamashi Tobu Hospital, Yokohama 230-8765, Japan; Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Clinical Research, Tochigi Medical Center, National Hospital Organization, Utsunomiya 320-8580, Japan
| | - Toshihiko Momiyama
- Division of Cerebral Structure, National Institute for Physiological Sciences, Okazaki 444-8787, Japan; Department of Pharmacology, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Takuro Hayashi
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurosurgery, Tokyo Medical Center, National Hospital Organization, Tokyo 152-8902, Japan
| | - Takeshi Kawase
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
302
|
Haeren R, Rijkers K, Schijns O, Dings J, Hoogland G, van Zandvoort M, Vink H, van Overbeeke J. In vivo assessment of the human cerebral microcirculation and its glycocalyx: A technical report. J Neurosci Methods 2018; 303:114-125. [DOI: 10.1016/j.jneumeth.2018.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 03/12/2018] [Accepted: 03/21/2018] [Indexed: 10/17/2022]
|
303
|
Salehi A, Jullienne A, Wendel KM, Hamer M, Tang J, Zhang JH, Pearce WJ, DeFazio RA, Vexler ZS, Obenaus A. A Novel Technique for Visualizing and Analyzing the Cerebral Vasculature in Rodents. Transl Stroke Res 2018; 10:10.1007/s12975-018-0632-0. [PMID: 29766452 DOI: 10.1007/s12975-018-0632-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/27/2018] [Accepted: 04/25/2018] [Indexed: 12/13/2022]
Abstract
We introduce a novel protocol to stain, visualize, and analyze blood vessels from the rat and mouse cerebrum. This technique utilizes the fluorescent dye, DiI, to label the lumen of the vasculature followed by perfusion fixation. Following brain extraction, the labeled vasculature is then imaged using wide-field fluorescence microscopy for axial and coronal images and can be followed by regional confocal microscopy. Axial and coronal images can be analyzed using classical angiographic methods for vessel density, length, and other features. We also have developed a novel fractal analysis to assess vascular complexity. Our protocol has been optimized for adult rat, adult mouse, and neonatal mouse studies. The protocol is efficient, can be rapidly completed, stains cerebral vessels with a bright fluorescence, and provides valuable quantitative data. This method has a broad range of applications, and we demonstrate its use to study the vasculature in assorted models of acquired brain injury.
Collapse
Affiliation(s)
- Arjang Salehi
- Cell, Molecular and Developmental Biology Program, University of California, Riverside, 1140 Bachelor Hall, Riverside, CA, 92521, USA
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Amandine Jullienne
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Kara M Wendel
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, 92697-4475, USA
| | - Mary Hamer
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, 92697-4475, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - William J Pearce
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
- Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Richard A DeFazio
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48101, USA
| | - Zinaida S Vexler
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Andre Obenaus
- Cell, Molecular and Developmental Biology Program, University of California, Riverside, 1140 Bachelor Hall, Riverside, CA, 92521, USA.
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA, 92354, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, 92697-4475, USA.
- Department of Pediatrics, University of California, Irvine, Irvine, CA, 92697-4475, USA.
| |
Collapse
|
304
|
Zhang X, Guo X, Zhang N, Cai H, Sun J, Wang Q, Qi Y, Zhang L, Yang L, Shi FD, Yu C. Cerebral Blood Flow Changes in Multiple Sclerosis and Neuromyelitis Optica and Their Correlations With Clinical Disability. Front Neurol 2018; 9:305. [PMID: 29780351 PMCID: PMC5946009 DOI: 10.3389/fneur.2018.00305] [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: 11/29/2017] [Accepted: 04/18/2018] [Indexed: 11/13/2022] Open
Abstract
Distinguishing relapsing-remitting multiple sclerosis (RRMS) and neuromyelitis optica (NMO) is clinically important because they differ in prognosis and treatment. This study aimed to identify perfusion abnormalities in RRMS and NMO and their correlations with gray matter volume (GMV) atrophy and clinical parameters. Structural and arterial spin labeling MRI scans were performed in 39 RRMS patients, 62 NMO patients, and 73 healthy controls. The gray matter cerebral blood flow (CBF) values were voxel-wisely compared among the three groups with and without GMV correction. The regional CBF changes were correlated with the Expanded Disability Status Scale scores in the corresponding patient groups. Although multiple brain regions showed CBF differences among the three groups without GMV correction, only three of these regions remained significant after GMV correction. Specifically, both the RRMS and NMO groups showed reduced CBF in the occipital cortex and increased CBF in the right putamen compared to the control group. The RRMS group had increased CBF only in the medial prefrontal cortex compared to the other two groups. The occipital CBF was negatively correlated with clinical disability in the NMO group; however, the CBF in the right putamen was positively correlated with clinical disability in both patient groups. These findings suggest that there are perfusion alterations independent of GMV atrophy in RRMS and NMO patients. The regional CBF in the occipital cortex and putamen could be used as imaging features to objectively assess clinical disability in these patients.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| | - Xi Guo
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| | - Ningnannan Zhang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| | - Huanhuan Cai
- Laboratory of Digital Medical Imaging, Medical Imaging Center, The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, China
| | - Jie Sun
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| | - Qiuhui Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| | - Yuan Qi
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Linjie Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Yang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin, China
| |
Collapse
|
305
|
Troxerutin and Cerebroprotein Hydrolysate Injection Protects Neurovascular Units from Oxygen-Glucose Deprivation and Reoxygenation-Induced Injury In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:9859672. [PMID: 29853981 PMCID: PMC5954925 DOI: 10.1155/2018/9859672] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/02/2018] [Indexed: 12/19/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury involves complex events of cellular and molecular processes. Previous studies suggest that a neurovascular unit (NVU) acts as an intricate network to maintain the neuronal homeostatic microenvironment. The present study established an NVU model for oxygen-glucose deprivation and reoxygenation (OGD/R) damage, trying to target the major components of the NVU using a coculture of rat neurons, astrocytes, and rat brain microvascular endothelial cells (rBMECs) to investigate the therapeutic effects of troxerutin and cerebroprotein hydrolysate injections (TCHis). The study observed that OGD/R downregulated the expressions of GAP-43, Claudin-5, and AQP-4 obviously detected by Western blotting and immunocytochemical analysis, respectively, while TCHi ameliorated the effect of OGD/R significantly. Meanwhile, TCHi alleviated the abnormalities of ultrastructure of neurons and rBMECs induced by OGD/R. Furthermore, both levels of inflammatory cytokines (IL-1β, IL-6, and TNF-α) and cell adhesion molecules (VCAM-1 and ICAM-1) detected by ELISA in NVU supernatant were found elevated significantly through OGD/R, but TCHi ameliorated the trend. In addition, TCHi also mitigated the increase of proapoptotic factors (Bax, p53, and caspase-3) induced by OGD/R in NVU model statistically. All these findings demonstrated that TCHis played a protective role, which was reflected in anti-inflammation, antiapoptosis, and blood–brain barrier maintenance. The results of the study concluded that the NVU is an ideal target and TCHi acts as a neuroprotective agent against cerebral I/R injuries.
Collapse
|
306
|
Yang S, Jin H, Zhu Y, Wan Y, Opoku EN, Zhu L, Hu B. Diverse Functions and Mechanisms of Pericytes in Ischemic Stroke. Curr Neuropharmacol 2018; 15:892-905. [PMID: 28088914 PMCID: PMC5652032 DOI: 10.2174/1570159x15666170112170226] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/30/2016] [Accepted: 12/28/2016] [Indexed: 12/26/2022] Open
Abstract
Background: Every year, strokes take millions of lives and leave millions of individuals living with permanent disabilities. Recently more researchers embrace the concept of the neurovascular unit (NVU), which encompasses neurons, endothelial cells (ECs), pericytes, astrocyte, microglia, and the extracellular matrix. It has been well-documented that NVU emerged as a new paradigm for the exploration of mechanisms and therapies in ischemic stroke. To better understand the complex NVU and broaden therapeutic targets, we must probe the roles of multiple cell types in ischemic stroke. The aims of this paper are to introduce the biological characteristics of brain pericytes and the available evidence on the diverse functions and mechanisms involving the pericytes in the context of ischemic stroke. Methods: Research and online content related to the biological characteristics and pathophysiological roles of pericytes is review. The new research direction on the Pericytes in ischemic stroke, and the potential therapeutic targets are provided. Results: During the different stages of ischemic stroke, pericytes play different roles: 1) On the hyperacute phase of stroke, pericytes constriction and death may be a cause of the no-reflow phenomenon in brain capillaries; 2) During the acute phase, pericytes detach from microvessels and participate in inflammatory-immunological response, resulting in the BBB damage and brain edema. Pericytes also provide benefit for neuroprotection by protecting endothelium, stabilizing BBB and releasing neurotrophins; 3) Similarly, during the later recovery phase of stroke, pericytes also contribute to angiogenesis, neurogenesis, and thereby promote neurological recovery. Conclusion: This emphasis on the NVU concept has shifted the focus of ischemic stroke research from neuro-centric views to the complex interactions within NVU. With this new perspective, pericytes that are centrally positioned in the NVU have been widely studied in ischemic stroke. More work is needed to elucidate the beneficial and detrimental roles of brain pericytes in ischemic stroke that may serve as a basis for potential therapeutic targets.
Collapse
Affiliation(s)
- Shuai Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiyi Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Elvis Nana Opoku
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lingqiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
307
|
Makris K, Haliassos A, Chondrogianni M, Tsivgoulis G. Blood biomarkers in ischemic stroke: potential role and challenges in clinical practice and research. Crit Rev Clin Lab Sci 2018; 55:294-328. [DOI: 10.1080/10408363.2018.1461190] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Konstantinos Makris
- Clinical Biochemistry Department, KAT General Hospital, Kifissia, Athens, Greece
| | | | - Maria Chondrogianni
- Second Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
308
|
Permeability of the Blood-Brain Barrier and Transport of Nanobodies Across the Blood-Brain Barrier. FOLIA VETERINARIA 2018. [DOI: 10.2478/fv-2018-0009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The presence of a blood-brain barrier (BBB) and a blood-cerebrospinal fluid barrier presents animmense challenge for effective delivery of therapeutics to the central nervous system. Many potential drugs, which are effective at their site of action, have failed due to the lack of distribution in sufficient quantity to the central nervous system (CNS). In consequence, many diseases of the central nervous system remain undertreated. Antibodies, IgG for example, are difficult to deliver to the CNS due to their size (~155 kDa), physico-chemical properties and the presence of Fc receptor on the blood-brain barrier. Smaller antibodies, like the recently developed nanobodies, may overcome the obstacle of the BBB and enter into the CNS. The nanobodies are the smallest available antigen-binding fragments harbouring the full antigenbinding capacity of conventional antibodies. They represent a new generation of therapeutics with exceptional properties, such as: recognition of unique epitopes, target specificity, high affinity, high solubility, high stability and high expression yields in cost-effective recombinant production. Their ability to permeate across the BBBmakes thema promising alternative for central nervous system disease therapeutics. In this review, we have systematically presented different aspects of the BBB, drug delivery mechanisms employed to cross the BBB, and finally nanobodies — a potential therapeutic molecule against neuroinfections.
Collapse
|
309
|
Goto S, Morikawa T, Kubo A, Takubo K, Fukuda K, Kajimura M, Suematsu M. Quantitative imaging mass spectroscopy reveals roles of heme oxygenase-2 for protecting against transhemispheric diaschisis in the brain ischemia. J Clin Biochem Nutr 2018; 63:70-79. [PMID: 30087547 PMCID: PMC6064818 DOI: 10.3164/jcbn.17-136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/09/2018] [Indexed: 11/22/2022] Open
Abstract
Carbon monoxide-generating heme oxygenase-2 is expressed in neurons and plays a crucial role for regulating hypoxic vasodilation through mechanisms unlocking carbon monoxide-dependent inhibition of H2S-generating cystathionine β-synthase expressed in astrocytes. This study aims to examine whether heme oxygenase-2 plays a protective role in mice against stroke. Focal ischemia was induced by middle cerebral artery occlusion. Regional differences in metabolites among ipsilateral and contralateral hemispheres were analysed by quantitative imaging mass spectrometry equipped with an image-processing platform to optimize comparison of local metabolite contents among different animals. Under normoxia, blood flow velocity in precapillary arterioles were significantly elevated in heme oxygenase-2-null mice vs controls, while metabolic intermediates of central carbon metabolism and glutamate synthesis were elevated in the brain of heme oxygenase-2-null mice, suggesting greater metabolic demands to induce hyperemia in these mice. In response to focal ischemia, heme oxygenase-2-null mice exhibited greater regions of ischemic core that coincide with notable decreases in energy metabolism in the contralateral hemisphere as well as in penumbra. In conclusion, these findings suggest that heme oxygenase-2 is involved in mechanisms by which not only protects against compromised energy metabolism of the ipsilateral hemisphere but also ameliorates transhemispheric diaschisis of the contralateral hemisphere in ischemic brain.
Collapse
Affiliation(s)
- Shinichi Goto
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan.,Department of Cardiology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takayuki Morikawa
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akiko Kubo
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Keiyo Takubo
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mayumi Kajimura
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
310
|
Padel T, Roth M, Gaceb A, Li JY, Björkqvist M, Paul G. Brain pericyte activation occurs early in Huntington's disease. Exp Neurol 2018; 305:139-150. [PMID: 29630897 DOI: 10.1016/j.expneurol.2018.03.015] [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: 12/15/2017] [Revised: 03/03/2018] [Accepted: 03/29/2018] [Indexed: 02/03/2023]
Abstract
Microvascular changes have recently been described for several neurodegenerative disorders, including Huntington's disease (HD). HD is characterized by a progressive neuronal cell loss due to a mutation in the Huntingtin gene. However, the temporal and spatial microvascular alterations in HD remain unclear. Also, knowledge on the implication of pericytes in HD pathology is still sparse and existing findings are contradictory. Here we examine alterations in brain pericytes in the R6/2 mouse model of HD and in human post mortem HD brain sections. To specifically track activated pericytes, we crossbred R6/2 mice with transgenic mice expressing the Green fluorescent protein gene under the Regulator of G-protein signaling 5 (Rgs5) promoter. We demonstrate an increase in activated pericytes in the R6/2 brain and in post mortem HD brain tissue. Importantly, pericyte changes are already detected before striatal neuronal cell loss, weight loss or behavioural deficits occur in R6/2 mice. This is associated with vascular alterations, whereby striatal changes precede cortical changes. Our findings suggest that pericyte activation may be one of the initial steps contributing to the observed vascular modifications in HD. Thus, pericytes may constitute an important target to address early microvascular changes contributing to disease progression in HD.
Collapse
Affiliation(s)
- Thomas Padel
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Michaela Roth
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden; Department of Neurology, Scania University Hospital, 22185 Lund, Sweden.
| |
Collapse
|
311
|
Pozhilenkova EA, Lopatina OL, Komleva YK, Salmin VV, Salmina AB. Blood-brain barrier-supported neurogenesis in healthy and diseased brain. Rev Neurosci 2018; 28:397-415. [PMID: 28195555 DOI: 10.1515/revneuro-2016-0071] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/23/2016] [Indexed: 12/23/2022]
Abstract
Adult neurogenesis is one of the most important mechanisms contributing to brain development, learning, and memory. Alterations in neurogenesis underlie a wide spectrum of brain diseases. Neurogenesis takes place in highly specialized neurogenic niches. The concept of neurogenic niches is becoming widely accepted due to growing evidence of the important role of the microenvironment established in the close vicinity to stem cells in order to provide adequate control of cell proliferation, differentiation, and apoptosis. Neurogenic niches represent the platform for tight integration of neurogenesis and angiogenesis supported by specific properties of cerebral microvessel endothelial cells contributing to establishment of partially compromised blood-brain barrier (BBB) for the adjustment of local conditions to the current metabolic needs of stem and progenitor cells. Here, we review up-to-date data on microvascular dynamics in activity-dependent neurogenesis, specific properties of BBB in neurogenic niches, endothelial-driven mechanisms of clonogenic activity, and future perspectives for reconstructing the neurogenic niches in vitro.
Collapse
|
312
|
Xu J, Long H, Chen W, Cheng X, Yu H, Huang Y, Wang X, Li F. Ultrastructural Features of Neurovascular Units in a Rat Model of Chronic Compressive Spinal Cord Injury. Front Neuroanat 2018; 11:136. [PMID: 29375327 PMCID: PMC5767600 DOI: 10.3389/fnana.2017.00136] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 12/26/2017] [Indexed: 12/16/2022] Open
Abstract
Chronic spinal cord compression is the most common cause of spinal cord impairment worldwide. Objective of this study is to assess the ultrastructural features of the neurovascular unit (NVU) in a rat model of chronic compressive spinal cord injury, 24 SD rats were divided into two groups: the control group (n = 12), and the compression group (n = 12). A C6 semi-laminectomy was performed in the control group, whereas a water-absorbent polyurethane polymer was implanted into the C6 epidural space in the compression group. The Basso Beattie Bresnahan (BBB) scores and the somatosensory evoked potentials (SEP) were used to evaluate neurological functions. Transmission Electron Microscopy (TEM) was performed to investigate the change of NVU at the 28th day after modeling. Compared with the control group, the compression group shows a significant reduction (P < 0.05) of BBB score and a significant severity (P < 0.05) of abnormal SEP. TEM results of the compression group showed a striking increase in endothelial caveolae and vacuoles; a number of small spaces in tight junctions; a significant increase in pericyte processing area and vessel coverage; an expansion of the basement membrane region; swollen astrocyte endfeet and mitochondria; and the degeneration of neurons and axons. Our study revealed that damage to NVU components occurred followed by chronic compressive spinal cord injury. Several compensatory changes characterized by thicker endothelium, expansive BM, increased pericyte processing area and vessel coverage were also observed.
Collapse
Affiliation(s)
- Jinghui Xu
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Houqing Long
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenli Chen
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xing Cheng
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Haoyang Yu
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yangliang Huang
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaobo Wang
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fobao Li
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
313
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
314
|
Osaki T, Shin Y, Sivathanu V, Campisi M, Kamm RD. In Vitro Microfluidic Models for Neurodegenerative Disorders. Adv Healthc Mater 2018; 7. [PMID: 28881425 DOI: 10.1002/adhm.201700489] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/18/2017] [Indexed: 01/09/2023]
Abstract
Microfluidic devices enable novel means of emulating neurodegenerative disease pathophysiology in vitro. These organ-on-a-chip systems can potentially reduce animal testing and substitute (or augment) simple 2D culture systems. Reconstituting critical features of neurodegenerative diseases in a biomimetic system using microfluidics can thereby accelerate drug discovery and improve our understanding of the mechanisms of several currently incurable diseases. This review describes latest advances in modeling neurodegenerative diseases in the central nervous system and the peripheral nervous system. First, this study summarizes fundamental advantages of microfluidic devices in the creation of compartmentalized cell culture microenvironments for the co-culture of neurons, glial cells, endothelial cells, and skeletal muscle cells and in their recapitulation of spatiotemporal chemical gradients and mechanical microenvironments. Then, this reviews neurodegenerative-disease-on-a-chip models focusing on Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Finally, this study discusses about current drawbacks of these models and strategies that may overcome them. These organ-on-chip technologies can be useful to be the first line of testing line in drug development and toxicology studies, which can contribute significantly to minimize the phase of animal testing steps.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Vivek Sivathanu
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Marco Campisi
- Department of Mechanical and Aerospace EngineeringPolitecnico di Torino Corso Duca degli Abruzzi 24 10129 Torino Italy
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
- Department of Biological EngineeringMassachusetts Institutes of Technology 500 Technology Square, MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| |
Collapse
|
315
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 964] [Impact Index Per Article: 160.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
316
|
Lytton WW, Arle J, Bobashev G, Ji S, Klassen TL, Marmarelis VZ, Schwaber J, Sherif MA, Sanger TD. Multiscale modeling in the clinic: diseases of the brain and nervous system. Brain Inform 2017; 4:219-230. [PMID: 28488252 PMCID: PMC5709279 DOI: 10.1007/s40708-017-0067-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 04/27/2017] [Indexed: 12/26/2022] Open
Abstract
Computational neuroscience is a field that traces its origins to the efforts of Hodgkin and Huxley, who pioneered quantitative analysis of electrical activity in the nervous system. While also continuing as an independent field, computational neuroscience has combined with computational systems biology, and neural multiscale modeling arose as one offshoot. This consolidation has added electrical, graphical, dynamical system, learning theory, artificial intelligence and neural network viewpoints with the microscale of cellular biology (neuronal and glial), mesoscales of vascular, immunological and neuronal networks, on up to macroscales of cognition and behavior. The complexity of linkages that produces pathophysiology in neurological, neurosurgical and psychiatric disease will require multiscale modeling to provide understanding that exceeds what is possible with statistical analysis or highly simplified models: how to bring together pharmacotherapeutics with neurostimulation, how to personalize therapies, how to combine novel therapies with neurorehabilitation, how to interlace periodic diagnostic updates with frequent reevaluation of therapy, how to understand a physical disease that manifests as a disease of the mind. Multiscale modeling will also help to extend the usefulness of animal models of human diseases in neuroscience, where the disconnects between clinical and animal phenomenology are particularly pronounced. Here we cover areas of particular interest for clinical application of these new modeling neurotechnologies, including epilepsy, traumatic brain injury, ischemic disease, neurorehabilitation, drug addiction, schizophrenia and neurostimulation.
Collapse
Affiliation(s)
- William W. Lytton
- Department of Physiology and Pharmacology and Neurology, SUNY Downstate, Kings County Hospital, Brooklyn, NY 11203 USA
| | | | | | - Songbai Ji
- Thayer School of Engineering, Department of Surgery and of Orthopaedic Surgery, Geisel School of Medicine, Dartmouth College, Hanover, NH 3755 USA
| | | | | | | | - Mohamed A. Sherif
- Yale U, New Haven, CT USA
- VA Connecticut Healthcare System, West Haven, CT USA
- Ain Shams U Institute of Psychiatry, Cairo, Egypt
| | | |
Collapse
|
317
|
García-Berrocoso T, Llombart V, Colàs-Campàs L, Hainard A, Licker V, Penalba A, Ramiro L, Simats A, Bustamante A, Martínez-Saez E, Canals F, Sanchez JC, Montaner J. Single Cell Immuno-Laser Microdissection Coupled to Label-Free Proteomics to Reveal the Proteotypes of Human Brain Cells After Ischemia. Mol Cell Proteomics 2017; 17:175-189. [PMID: 29133510 DOI: 10.1074/mcp.ra117.000419] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Indexed: 12/13/2022] Open
Abstract
Cerebral ischemia entails rapid tissue damage in the affected brain area causing devastating neurological dysfunction. How each component of the neurovascular unit contributes or responds to the ischemic insult in the context of the human brain has not been solved yet. Thus, the analysis of the proteome is a straightforward approach to unraveling these cell proteotypes. In this study, post-mortem brain slices from ischemic stroke patients were obtained corresponding to infarcted (IC) and contralateral (CL) areas. By means of laser microdissection, neurons and blood brain barrier structures (BBB) were isolated and analyzed using label-free quantification. MS data are available via ProteomeXchange with identifier PXD003519. Ninety proteins were identified only in neurons, 260 proteins only in the BBB and 261 proteins in both cell types. Bioinformatics analyses revealed that repair processes, mainly related to synaptic plasticity, are outlined in microdissected neurons, with nonexclusive important functions found in the BBB. A total of 30 proteins showing p < 0.05 and fold-change> 2 between IC and CL areas were considered meaningful in this study: 13 in neurons, 14 in the BBB and 3 in both cell types. Twelve of these proteins were selected as candidates and analyzed by immunohistofluorescence in independent brains. The MS findings were completely verified for neuronal SAHH2 and SRSF1 whereas the presence in both cell types of GABT and EAA2 was only validated in neurons. In addition, SAHH2 showed its potential as a prognostic biomarker of neurological improvement when analyzed early in the plasma of ischemic stroke patients. Therefore, the quantitative proteomes of neurons and the BBB (or proteotypes) after human brain ischemia presented here contribute to increasing the knowledge regarding the molecular mechanisms of ischemic stroke pathology and highlight new proteins that might represent putative biomarkers of brain ischemia or therapeutic targets.
Collapse
Affiliation(s)
- Teresa García-Berrocoso
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Víctor Llombart
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Colàs-Campàs
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alexandre Hainard
- §Proteomics Core Facility, Faculty of medicine, University Medical Center, University of Geneva, Geneva, Switzerland
| | - Virginie Licker
- ¶Neuroproteomics Group, Human protein sciences department, University Medical Center, University of Geneva, Geneva, Switzerland
| | - Anna Penalba
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Ramiro
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Simats
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandro Bustamante
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Martínez-Saez
- ‖Neuropathology, Pathology department, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Canals
- **Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jean-Charles Sanchez
- ‡‡Translational biomarker group, Human protein sciences department, University Medical Center, University of Geneva, Geneva, Switzerland
| | - Joan Montaner
- From the ‡Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain;
| |
Collapse
|
318
|
Reproducibility of task activation using the Addenbrooke’s cognitive examination in healthy controls: A functional Transcranial Doppler ultrasonography study. J Neurosci Methods 2017; 291:131-140. [DOI: 10.1016/j.jneumeth.2017.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/04/2017] [Accepted: 08/14/2017] [Indexed: 11/22/2022]
|
319
|
Heroin Contaminated with Fentanyl Dramatically Enhances Brain Hypoxia and Induces Brain Hypothermia. eNeuro 2017; 4:eN-NWR-0323-17. [PMID: 29085909 PMCID: PMC5661359 DOI: 10.1523/eneuro.0323-17.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 09/27/2017] [Indexed: 01/24/2023] Open
Abstract
While opioid abuse is an established medical and public health issue, the increased availability of highly potent synthetic opioids, such as fentanyl, has given rise to acute health complications, including a comatose state and death during drug overdose. Since respiratory depression that leads to acute hypoxia is the most dangerous complication of opioid drug use, we examined the effects of intravenous heroin and heroin contaminated with 10% fentanyl on oxygen levels in the nucleus accumbens (NAc) monitored using high-speed amperometry in freely moving rats. Additionally, we examined the effects of heroin, fentanyl, and their mixture on locomotion and temperatures in the NAc, temporal muscle, and skin. Both fentanyl and heroin at human-relevant doses (400 and 40 μg/kg, respectively) induced rapid, strong and transient decreases in NAc oxygen, indicative of brain hypoxia. When the heroin-fentanyl mixture was injected, the NAc hypoxic response was greatly potentiated in its duration, suggesting sustained hypoxia. In contrast to modest, monophasic brain temperature increases caused by heroin alone, the heroin-fentanyl mixture induced a biphasic temperature response, with a prominent postinjection decrease resulting from peripheral vasodilation. This hypothermic effect, albeit much smaller and more transient, was typical of fentanyl injected alone. Our findings indicate that accidental use of fentanyl instead of heroin, or even a relatively minor contamination of “street heroin” with fentanyl, poses great danger for acute health complications, including a comatose state and death.
Collapse
|
320
|
Anraku Y, Kuwahara H, Fukusato Y, Mizoguchi A, Ishii T, Nitta K, Matsumoto Y, Toh K, Miyata K, Uchida S, Nishina K, Osada K, Itaka K, Nishiyama N, Mizusawa H, Yamasoba T, Yokota T, Kataoka K. Glycaemic control boosts glucosylated nanocarrier crossing the BBB into the brain. Nat Commun 2017; 8:1001. [PMID: 29042554 PMCID: PMC5645389 DOI: 10.1038/s41467-017-00952-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023] Open
Abstract
Recently, nanocarriers that transport bioactive substances to a target site in the body have attracted considerable attention and undergone rapid progression in terms of the state of the art. However, few nanocarriers can enter the brain via a systemic route through the blood-brain barrier (BBB) to efficiently reach neurons. Here we prepare a self-assembled supramolecular nanocarrier with a surface featuring properly configured glucose. The BBB crossing and brain accumulation of this nanocarrier are boosted by the rapid glycaemic increase after fasting and by the putative phenomenon of the highly expressed glucose transporter-1 (GLUT1) in brain capillary endothelial cells migrating from the luminal to the abluminal plasma membrane. The precisely controlled glucose density on the surface of the nanocarrier enables the regulation of its distribution within the brain, and thus is successfully optimized to increase the number of nanocarriers accumulating in neurons.There are only a few examples of nanocarriers that can transport bioactive substances across the blood-brain barrier. Here the authors show that by rapid glycaemic increase the accumulation of a glucosylated nanocarrier in the brain can be controlled.
Collapse
Affiliation(s)
- Y Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - H Kuwahara
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Y Fukusato
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - A Mizoguchi
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - T Ishii
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - K Nitta
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Y Matsumoto
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - K Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - K Miyata
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - S Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - K Nishina
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - K Osada
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - K Itaka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - N Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, R1-11, 4259 Nagatsuta, Midori-ku, Yokohama, 226-8503, Japan
| | - H Mizusawa
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - T Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - T Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan. .,Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| | - K Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan. .,Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
321
|
Toutounchian JJ, McCarty JH. Selective expression of eGFP in mouse perivascular astrocytes by modification of the Mlc1 gene using T2A-based ribosome skipping. Genesis 2017; 55. [PMID: 28929580 DOI: 10.1002/dvg.23071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/12/2017] [Accepted: 09/17/2017] [Indexed: 11/12/2022]
Abstract
Perivascular astrocyte end feet closely juxtapose cerebral blood vessels to regulate important developmental and physiological processes including endothelial cell proliferation and sprouting as well as the formation of the blood-brain barrier (BBB). The mechanisms underlying these events remain largely unknown due to a lack of experimental models for identifying perivascular astrocytes and distinguishing these cell types from other astroglial populations. Megalencephalic leukoencephalopathy with subcortical cysts 1 (Mlc1) is a transmembrane protein that is expressed in perivascular astrocyte end feet where it controls BBB development and homeostasis. On the basis of this knowledge, we used T2A peptide-skipping strategies to engineer a knock-in mouse model in which the endogenous Mlc1 gene drives expression of enhanced green fluorescent protein (eGFP), without impacting expression of Mlc1 protein. Analysis of fetal, neonatal and adult Mlc1-eGFP knock-in mice revealed a dynamic spatiotemporal expression pattern of eGFP in glial cells, including nestin-expressing neuroepithelial cells during development and glial fibrillary acidic protein (GFAP)-expressing perivascular astrocytes in the postnatal brain. EGFP was not expressed in neurons, microglia, oligodendroglia, or cerebral vascular cells. Analysis of angiogenesis in the neonatal retina also revealed enriched Mlc1-driven eGFP expression in perivascular astrocytes that contact sprouting blood vessels and regulate blood-retinal barrier permeability. A cortical injury model revealed that Mlc1-eGFP expression is progressively induced in reactive astrocytes that form a glial scar. Hence, Mlc1-eGFP knock-in mice are a new and powerful tool to identify perivascular astrocytes in the brain and retina and characterize how these cell types regulate cerebral blood vessel functions in health and disease.
Collapse
Affiliation(s)
- Jordan J Toutounchian
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| | - Joseph H McCarty
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, 77030
| |
Collapse
|
322
|
Halford J, Shen S, Itamura K, Levine J, Chong AC, Czerwieniec G, Glenn TC, Hovda DA, Vespa P, Bullock R, Dietrich WD, Mondello S, Loo JA, Wanner IB. New astroglial injury-defined biomarkers for neurotrauma assessment. J Cereb Blood Flow Metab 2017; 37:3278-3299. [PMID: 28816095 PMCID: PMC5624401 DOI: 10.1177/0271678x17724681] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 05/01/2017] [Accepted: 05/25/2017] [Indexed: 01/08/2023]
Abstract
Traumatic brain injury (TBI) is an expanding public health epidemic with pathophysiology that is difficult to diagnose and thus treat. TBI biomarkers should assess patients across severities and reveal pathophysiology, but currently, their kinetics and specificity are unclear. No single ideal TBI biomarker exists. We identified new candidates from a TBI CSF proteome by selecting trauma-released, astrocyte-enriched proteins including aldolase C (ALDOC), its 38kD breakdown product (BDP), brain lipid binding protein (BLBP), astrocytic phosphoprotein (PEA15), glutamine synthetase (GS) and new 18-25kD-GFAP-BDPs. Their levels increased over four orders of magnitude in severe TBI CSF. First post-injury week, ALDOC levels were markedly high and stable. Short-lived BLBP and PEA15 related to injury progression. ALDOC, BLBP and PEA15 appeared hyper-acutely and were similarly robust in severe and mild TBI blood; 25kD-GFAP-BDP appeared overnight after TBI and was rarely present after mild TBI. Using a human culture trauma model, we investigated biomarker kinetics. Wounded (mechanoporated) astrocytes released ALDOC, BLBP and PEA15 acutely. Delayed cell death corresponded with GFAP release and proteolysis into small GFAP-BDPs. Associating biomarkers with cellular injury stages produced astroglial injury-defined (AID) biomarkers that facilitate TBI assessment, as neurological deficits are rooted not only in death of CNS cells, but also in their functional compromise.
Collapse
Affiliation(s)
- Julia Halford
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Sean Shen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Kyohei Itamura
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Jaclynn Levine
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Albert C Chong
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Gregg Czerwieniec
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Thomas C Glenn
- Department of Neurosurgery, Brain Injury Research Center, Department of Molecular and Medical Pharmacology
| | - David A Hovda
- Department of Neurosurgery, Brain Injury Research Center, Department of Molecular and Medical Pharmacology
| | - Paul Vespa
- Department of Neurology, UCLA-David Geffen School of Medicine, Los Angeles, CA, USA
| | - Ross Bullock
- Department of Neurological Surgery, Jackson Memorial Hospital, Miami, FL, USA
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami-Miller School of Medicine, Miami, FL, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
- Department of Biological Chemistry, UCLA Molecular Biology Institute, and UCLA/DOE Institute for Genomics and Proteomics, University of California, Los Angeles, CA, USA
| | - Ina-Beate Wanner
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| |
Collapse
|
323
|
Bang S, Lee SR, Ko J, Son K, Tahk D, Ahn J, Im C, Jeon NL. A Low Permeability Microfluidic Blood-Brain Barrier Platform with Direct Contact between Perfusable Vascular Network and Astrocytes. Sci Rep 2017; 7:8083. [PMID: 28808270 PMCID: PMC5556097 DOI: 10.1038/s41598-017-07416-0] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/26/2017] [Indexed: 12/14/2022] Open
Abstract
A novel three dimensional blood brain barrier (BBB) platform was developed by independently supplying different types of media to separate cell types within a single device. One channel (vascular channel, VC) is connected to the inner lumen of the vascular network while the other supplies media to the neural cells (neural channel, NC). Compared to co-cultures supplied with only one type of medium (or 1:1 mixture), best barrier properties and viability were obtained with culturing HUVECs with endothelial growth medium (EGM) and neural cells with neurobasal medium supplemented with fetal bovine serum (NBMFBS) independently. The measured vascular network permeability were comparable to reported in vivo values (20 kDa FITC-dextran, 0.45 ± 0.11 × 10−6 cm/s; 70 kDa FITC-dextran, 0.36 ± 0.05 × 10−6 cm/s) and a higher degree of neurovascular interfacing (astrocytic contact with the vascular network, GFAP-CD31 stain overlap) and presence of synapses (stained with synaptophysin). The BBB platform can dependably imitate the perivascular network morphology and synaptic structures characteristic of the NVU. This microfluidic BBB model can find applications in screening pharmaceuticals that target the brain for in neurodegenerative diseases.
Collapse
Affiliation(s)
- Seokyoung Bang
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Seung-Ryeol Lee
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Jihoon Ko
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Kyungmin Son
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Dongha Tahk
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Jungho Ahn
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Changkyun Im
- BK21 Plus Transformative Training Program for Creative Mechanical Engineers, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- Division of WCU (World Class University) Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea. .,Institute of Advanced Machinery and Design, Seoul National University, Seoul, South Korea.
| |
Collapse
|
324
|
Proximate Mediators of Microvascular Dysfunction at the Blood-Brain Barrier: Neuroinflammatory Pathways to Neurodegeneration. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1549194. [PMID: 28890893 PMCID: PMC5584365 DOI: 10.1155/2017/1549194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/09/2017] [Indexed: 12/14/2022]
Abstract
Current projections are that by 2050 the numbers of people aged 65 and older with Alzheimer's disease (AD) in the US may increase threefold while dementia is projected to double every 20 years reaching ~115 million by 2050. AD is clinically characterized by progressive dementia and neuropathologically by neuronal and synapse loss, accumulation of amyloid plaques, and neurofibrillary tangles (NFTs) in specific brain regions. The preclinical or presymptomatic stage of AD-related brain changes may begin over 20 years before symptoms occur, making development of noninvasive biomarkers essential. Distinct from neuroimaging and cerebrospinal fluid biomarkers, plasma or serum biomarkers can be analyzed to assess (i) the presence/absence of AD, (ii) the risk of developing AD, (iii) the progression of AD, or (iv) AD response to treatment. No unifying theory fully explains the neurodegenerative brain lesions but neuroinflammation (a lethal stressor for healthy neurons) is universally present. Current consensus is that the earlier the diagnosis, the better the chance to develop treatments that influence disease progression. In this article we provide a detailed review and analysis of the role of the blood-brain barrier (BBB) and damage-associated molecular patterns (DAMPs) as well as coagulation molecules in the onset and progression of these neurodegenerative disorders.
Collapse
|
325
|
Zou J, Chen Z, Wei X, Chen Z, Fu Y, Yang X, Chen D, Wang R, Jenner P, Lu JH, Li M, Zhang Z, Tang B, Jin K, Wang Q. Cystatin C as a potential therapeutic mediator against Parkinson's disease via VEGF-induced angiogenesis and enhanced neuronal autophagy in neurovascular units. Cell Death Dis 2017; 8:e2854. [PMID: 28569795 PMCID: PMC5520899 DOI: 10.1038/cddis.2017.240] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/18/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023]
Abstract
Cystatin C (CYS C, Cst3) is an endogenous cysteine protease inhibitor that plays neuroprotective roles in neurodegenerative diseases. We aimed to explore the association of CYS C with Parkinson’s disease (PD) models and investigate its involvement in the role of neurovascular units (NVUs) in PD neuro-pathogenesis. We used A53T α-synuclein (SNCA) transgenic mice and 6-hydroxydopamine-lesioned DAergic PC12 cells as experimental PD models to investigate the mechanisms behind this association. The injections of CYS C were administered to the right substantia nigra (SN) of A53T SNCA transgenic mice to measure the effects of CYS C in transgenic A53T SNCA mice. To explore the angiogenesis in vivo and in vitro, we used the chick embryo chorioallantoic membrane (CAM) assay and tube formation (TF) assay. We found that CYS C has a neuroprotective effect in this in vivo PD model. We observed increased VEGF, NURR1 and autophagy markers LC3B and decreased SNCA and apoptosis marker cleaved CASP3 in different brain regions of CYS C-treated A53T SNCA transgenic mice. In vitro, we observed that CYS C-induced VEGF, a secreted protein, attenuated 6-OHDA-lesioned DAergic PC12 cell degeneration by regulating p-PKC-α/p-ERK1/2-Nurr1 signaling and inducing autophagy. VEGF-mediated angiogenesis was markedly enhanced in the conditioned media of 6-OHDA-lesioned PC12 cells with CYS C-overexpression, whereas blockage of autophagy in CYS C-overexpressing PC12 cells significantly downregulated VEGF expression and the associated angiogenesis. Our data indicate that CYS C displays dual neuronal–vascular functions, promoting PC12 cell survival and angiogenesis via regulating the level of secreted VEGF in NVUs. Our study provides evidence that may aid in the development of an alternative approach for the treatment of PD through modulation of CYS C-mediated neuronal-vascular pathways.
Collapse
Affiliation(s)
- Jing Zou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Zhaoyu Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiaobo Wei
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Zhigang Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Yongmei Fu
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoyan Yang
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Dan Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Rui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Faculty of Health Sciences and Medicine, King's College, London SE1 1UL, UK
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Zhuohua Zhang
- Department of Neurology, Xiangya School of Medicine and The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | - Beisha Tang
- Department of Neurology, Xiangya School of Medicine and The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Qing Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
326
|
Reactivity in the human retinal microvasculature measured during acute gas breathing provocations. Sci Rep 2017; 7:2113. [PMID: 28522835 PMCID: PMC5437020 DOI: 10.1038/s41598-017-02344-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/10/2017] [Indexed: 11/11/2022] Open
Abstract
Although changes in vessel diameter following gas perturbation have been documented in retinal arterioles and venules, these responses have yet to be quantified in the smallest vessels of the human retina. Here, using in vivo adaptive optics, we imaged 3–25 µm diameter vessels of the human inner retinal circulation and monitored the effects of altered gas-breathing conditions. During isocapnic hyperoxia, definite constrictions were seen in 51% of vessel segments (mean ± SD for pre-capillary arterioles −9.5 ± 3.0%; capillaries −11.8 ± 3.3%; post-capillary venules −6.3 ± 2.8%); these are comparable with responses previously reported in larger vessels. During isoxic hypercapnia, definite dilations were seen in 47% of vessel segments (mean ± SD for pre-capillary arterioles +9.8 ± 1.5%; capillaries +13.7 ± 3.8%; post-capillary venules +7.5 ± 4.2%); these are proportionally greater than responses previously reported in larger vessels. The magnitude of these proportional changes implies that the capillary beds themselves play an important role in the retinal response to changes in carbon dioxide levels. Interestingly, the distribution of microvascular responses shown here differs from our previously reported responses to flicker stimulation, suggesting differences in the way blood supply is coordinated following gas perturbation and altered neural activity.
Collapse
|
327
|
Wang DP, Liu KJ, Kasper G, Lin Q, Hai J. Inhibition of SENP3 by URB597 ameliorates neurovascular unit dysfunction in rats with chronic cerebral hypoperfusion. Biomed Pharmacother 2017; 91:872-879. [PMID: 28501776 DOI: 10.1016/j.biopha.2017.05.021] [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] [Received: 01/30/2017] [Revised: 04/20/2017] [Accepted: 05/04/2017] [Indexed: 12/14/2022] Open
Abstract
Disruption of the neurovascular unit (NVU), induced by chronic cerebral hypoperfusion (CCH), has been broadly found in various neurological disorders. SUMO-specific protease 3 (SENP3) is expressed in neurons, astrocytes, and microglia, and regulates a variety of cell events. However, whether SENP3 is involved in neurovascular injury under the condition of CCH is still elusive. To address this issue, we investigated the effect of the fatty acid amide hydrolase (FAAH) inhibitor URB597 on NVU and the role of SENP3 in this process, as well as the underling mechanisms. The expression of SENP3 was detected by immunochemistry. The function and structure of the NVU was assessed by Western blot analysis and transmission electron microscopy. CCH caused the upregulation of SENP3, the disruption of cell and non-cell components at the protein level within the NVU, and ultrastructural deterioration. The NVU impairment as well as overexpression of SENP3 were reversed by treatment with URB597. These results reveal a novel neuroprotective role in URB597, which implicates URB597 in the amelioration of CCH-induced NVU impairment by inhibiting SENP3.
Collapse
Affiliation(s)
- Da-Peng Wang
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai 200065, China; Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Ke-Jia Liu
- Department of Cell Biology, Key Laboratory of Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Graham Kasper
- McGill Neuroscience, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Qi Lin
- Department of Pharmacy, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Hai
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai 200065, China.
| |
Collapse
|
328
|
Acosta C, Anderson HD, Anderson CM. Astrocyte dysfunction in Alzheimer disease. J Neurosci Res 2017; 95:2430-2447. [PMID: 28467650 DOI: 10.1002/jnr.24075] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022]
Abstract
Astrocytes are glial cells that are distributed throughout the central nervous system in an arrangement optimal for chemical and physical interaction with neuronal synapses and brain blood supply vessels. Neurotransmission modulates astrocytic excitability by activating an array of cell surface receptors and transporter proteins, resulting in dynamic changes in intracellular Ca2+ or Na+ . Ionic and electrogenic astrocytic changes, in turn, drive vital cell nonautonomous effects supporting brain function, including regulation of synaptic activity, neuronal metabolism, and regional blood supply. Alzheimer disease (AD) is associated with aberrant oligomeric amyloid β generation, which leads to extensive proliferation of astrocytes with a reactive phenotype and abnormal regulation of these processes. Astrocytic morphology, Ca2+ responses, extracellular K+ removal, glutamate transport, amyloid clearance, and energy metabolism are all affected in AD, resulting in a deleterious set of effects that includes glutamate excitotoxicity, impaired synaptic plasticity, reduced carbon delivery to neurons for oxidative phosphorylation, and dysregulated linkages between neuronal energy demand and regional blood supply. This review summarizes how astrocytes are affected in AD and describes how these changes are likely to influence brain function. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Crystal Acosta
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Canadian Centre for Agri-food Research in Health and Medicine, St. Boniface Hospital Research, Winnipeg, Manitoba, Canada
| | - Hope D Anderson
- Canadian Centre for Agri-food Research in Health and Medicine, St. Boniface Hospital Research, Winnipeg, Manitoba, Canada.,College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Christopher M Anderson
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
329
|
McInerney MP, Short JL, Nicolazzo JA. Neurovascular Alterations in Alzheimer's Disease: Transporter Expression Profiles and CNS Drug Access. AAPS JOURNAL 2017; 19:940-956. [PMID: 28462473 DOI: 10.1208/s12248-017-0077-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/15/2017] [Indexed: 01/05/2023]
Abstract
Despite a century of steady and incremental progress toward understanding the underlying biochemical mechanisms, Alzheimer's disease (AD) remains a complicated and enigmatic disease, and greater insight will be necessary before substantive clinical success is realised. Over the last decade in particular, a large body of work has highlighted the cerebral microvasculature as an anatomical region with an increasingly apparent role in the pathogenesis of AD. The causative interplay and temporal cascade that manifest between the brain vasculature and the wider disease progression of AD are not yet fully understood, and further inquiry is required to properly characterise these relationships. The purpose of this review is to highlight the recent advancements in research implicating neurovascular factors in AD, at both the molecular and anatomical levels. We begin with a brief introduction of the biochemical and genetic aspects of AD, before reviewing the essential concepts of the blood-brain barrier (BBB) and the neurovascular unit (NVU). In detail, we then examine the evidence demonstrating involvement of BBB dysfunction in AD pathogenesis, highlighting the importance of neurovascular components in AD. Lastly, we include within this review research that focuses on how altered properties of the BBB in AD impact upon CNS exposure of therapeutic agents and the potential clinical impact that this may have on people with this disease.
Collapse
Affiliation(s)
- Mitchell P McInerney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Jennifer L Short
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, VIC, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
330
|
Solis E, Cameron-Burr KT, Kiyatkin EA. Rapid Physiological Fluctuations in Nucleus Accumbens Oxygen Levels Induced by Arousing Stimuli: Relationships with Changes in Brain Glucose and Metabolic Neural Activation. Front Integr Neurosci 2017; 11:9. [PMID: 28484378 PMCID: PMC5401908 DOI: 10.3389/fnint.2017.00009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/06/2017] [Indexed: 01/01/2023] Open
Abstract
Proper entry of oxygen from arterial blood into the brain is essential for maintaining brain metabolism under normal conditions and during functional neural activation. However, little is known about physiological fluctuations in brain oxygen and their underlying mechanisms. To address this issue, we employed high-speed amperometry with platinum oxygen sensors in freely moving male rats. Recordings were conducted in the nucleus accumbens (NAc), a critical structure for sensorimotor integration. Rats were exposed to arousing stimuli of different nature (brief auditory tone, a 1-min novel object presentation, a 3-min social interaction with a conspecific, and a 3-min tail-pinch). We found that all arousing stimuli increased NAc oxygen levels. Increases were rapid (4–10-s onset latencies), modest in magnitude (1–3 μM or 5%–15% over baseline) and duration (5–20 min), and generally correlated with the arousing potential of each stimulus. Two strategies were used to determine the mechanisms underlying the observed increases in NAc oxygen levels. First, we showed that NAc oxygen levels phasically increase following intra-NAc microinjections of glutamate (GLU) that excite accumbal neurons. Therefore, local neural activation with subsequent local vasodilation is involved in mediating physiological increases in NAc oxygen induced by arousing stimuli. Second, by employing oxygen monitoring in the subcutaneous space, a highly-vascularized area with no metabolic activity, we determined that physiological increases in NAc oxygen also depend on the rise in blood oxygen levels caused by respiratory activation. Due to the co-existence of different mechanisms governing oxygen entry into brain tissue, NAc oxygen responses differ from fluctuations in NAc glucose, which, within a normal behavioral continuum, are regulated exclusively by neuro-vascular coupling due to glucose’s highly stable levels in the blood. Finally, we discuss the relationships between physiological fluctuations in NAc oxygen, glucose and metabolic brain activation assessed by intra-brain heat production.
Collapse
Affiliation(s)
- Ernesto Solis
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse-Intramural Research Program, Department of Health and Human Services, National Institutes of HealthBaltimore, MD, USA
| | - Keaton T Cameron-Burr
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse-Intramural Research Program, Department of Health and Human Services, National Institutes of HealthBaltimore, MD, USA
| | - Eugene A Kiyatkin
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse-Intramural Research Program, Department of Health and Human Services, National Institutes of HealthBaltimore, MD, USA
| |
Collapse
|
331
|
Radu BM, Epureanu FB, Radu M, Fabene PF, Bertini G. Nonsteroidal anti-inflammatory drugs in clinical and experimental epilepsy. Epilepsy Res 2017; 131:15-27. [DOI: 10.1016/j.eplepsyres.2017.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/04/2017] [Accepted: 02/07/2017] [Indexed: 01/01/2023]
|
332
|
Adriani G, Ma D, Pavesi A, Kamm RD, Goh ELK. A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood-brain barrier. LAB ON A CHIP 2017; 17:448-459. [PMID: 28001148 DOI: 10.1039/c6lc00638h] [Citation(s) in RCA: 288] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The neurovascular unit is a complex, interdependent system composed of neurons and neural supporting cells, such as astrocytes, as well as cells that comprise the vascular system including endothelial cells, pericytes, and smooth muscle cells. Each cell type in the neurovascular unit plays an essential role, either in transmitting and processing neural signals or in maintaining the appropriate microenvironmental conditions for healthy neural function. In vitro neurovascular models can be useful for understanding the different roles and functions of the cells composing the neurovascular unit, as well as for assessing the effects on neural function of therapeutic compounds after crossing the endothelial barrier. Here, we report a novel three-dimensional neurovascular microfluidic model consisting of primary rat astrocytes and neurons together with human cerebral microvascular endothelial cells. These three cell types in our neurovascular chip (NVC) show distinct cell type-specific morphological characteristics and functional properties. In particular, morphological and functional analysis of neurons enables quantitative assessment of neuronal responses, while human cerebral endothelial cells form monolayers with size-selective permeability similar to existing in vitro blood-brain barrier (BBB) models.
Collapse
Affiliation(s)
- Giulia Adriani
- Singapore-MIT Alliance for Research and Technology, 138602 Singapore
| | - Dongliang Ma
- Department of Research, National Neuroscience Institute, 20 College Road, 169856 Singapore and Neuroscience Academic Clinical Programme, Duke-NUS Medical School, 169857 Singapore.
| | - Andrea Pavesi
- Singapore-MIT Alliance for Research and Technology, 138602 Singapore
| | - Roger D Kamm
- Singapore-MIT Alliance for Research and Technology, 138602 Singapore and Massachusetts Institute of Technology, Cambridge, MA, 02139 USA.
| | - Eyleen L K Goh
- Department of Research, National Neuroscience Institute, 20 College Road, 169856 Singapore and Neuroscience Academic Clinical Programme, Duke-NUS Medical School, 169857 Singapore. and Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore and KK Research Center, KK Women's and Children's Hospital, Singapore 229899, Singapore
| |
Collapse
|
333
|
Lee JM, Park JM, Song MK, Oh YJ, Kim CJ, Kim YJ. The ameliorative effects of exercise on cognitive impairment and white matter injury from blood-brain barrier disruption induced by chronic cerebral hypoperfusion in adolescent rats. Neurosci Lett 2016; 638:83-89. [PMID: 27956237 DOI: 10.1016/j.neulet.2016.12.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/13/2016] [Accepted: 12/08/2016] [Indexed: 01/13/2023]
Abstract
Vascular dementia is the progressive change in blood vessels that leads to neuronal injuries in vulnerable areas induced by chronic cerebral hypoperfusion (CCH). CCH induces disruption of blood-brain barrier (BBB), and this BBB disruption can initiate the cognitive impairment and white matter injury. In the present study, we evaluated the effect of treadmill exercise on the cognitive impairment, white matter injury, and BBB disruption induced by CCH. Vascular dementia was induced by permanent bilateral common carotid arteries occlusion (BCCAO) in rats. The rats in the exercise group were made to run on a treadmill for 30min once a day for 14 weeks, starting 4 weeks after birth. Our results revealed that treadmill exercise group was alleviated the cognitive impairment and myelin degradation induced by CCH. The disruption of BBB after CCH indicates degradation of occludin, zonula occluden-1 (ZO-1), and up-regulation of matrix metalloproteinases (MMPs). Treadmill exercise may provide protective effects on BBB disruption from degradation of occludin, ZO-1, and overexpression of MMP-9 after CCH. These findings suggest that treadmill exercise ameliorates cognitive impairment and white matter injury from BBB disruption induced by CCH in rats. The present study will be valuable for means of prophylactic and therapeutic intervention for patients with CCH.
Collapse
Affiliation(s)
- Jae-Min Lee
- Department of Physiology, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, South Korea
| | - Jong-Min Park
- Department of Physiology, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, South Korea
| | - Min Kyung Song
- Department of Basic Nursing Science, College of Nursing Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701 South Korea
| | - Yoo Joung Oh
- Department of Basic Nursing Science, College of Nursing Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701 South Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, South Korea
| | - Youn-Jung Kim
- Department of Basic Nursing Science, College of Nursing Science, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701 South Korea.
| |
Collapse
|
334
|
Festoff BW, Sajja RK, van Dreden P, Cucullo L. HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J Neuroinflammation 2016; 13:194. [PMID: 27553758 PMCID: PMC4995775 DOI: 10.1186/s12974-016-0670-z] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/17/2016] [Indexed: 01/11/2023] Open
Abstract
Background The blood-brain barrier (BBB) dysfunction represents an early feature of Alzheimer’s disease (AD) that precedes the hallmarks of amyloid beta (amyloid β) plaque deposition and neuronal neurofibrillary tangle (NFT) formation. A damaged BBB correlates directly with neuroinflammation involving microglial activation and reactive astrogliosis, which is associated with increased expression and/or release of high-mobility group box protein 1 (HMGB1) and thrombin. However, the link between the presence of these molecules, BBB damage, and progression to neurodegeneration in AD is still elusive. Therefore, we aimed to profile and validate non-invasive clinical biomarkers of BBB dysfunction and neuroinflammation to assess the progression to neurodegeneration in mild cognitive impairment (MCI) and AD patients. Methods We determined the serum levels of various proinflammatory damage-associated molecules in aged control subjects and patients with MCI or AD using validated ELISA kits. We then assessed the specific and direct effects of such molecules on BBB integrity in vitro using human primary brain microvascular endothelial cells or a cell line. Results We observed a significant increase in serum HMGB1 and soluble receptor for advanced glycation end products (sRAGE) that correlated well with amyloid beta levels in AD patients (vs. control subjects). Interestingly, serum HMGB1 levels were significantly elevated in MCI patients compared to controls or AD patients. In addition, as a marker of BBB damage, soluble thrombomodulin (sTM) antigen, and activity were significantly (and distinctly) increased in MCI and AD patients. Direct in vitro BBB integrity assessment further revealed a significant and concentration-dependent increase in paracellular permeability to dextrans by HMGB1 or α-thrombin, possibly through disruption of zona occludins-1 bands. Pre-treatment with anti-HMGB1 monoclonal antibody blocked HMGB1 effects and leaving BBB integrity intact. Conclusions Our current studies indicate that thrombin and HMGB1 are causal proximate proinflammatory mediators of BBB dysfunction, while sTM levels may indicate BBB endothelial damage; HMGB1 and sRAGE might serve as clinical biomarkers for progression and/or therapeutic efficacy along the AD spectrum.
Collapse
Affiliation(s)
- Barry W Festoff
- pHLOGISTIX LLC, 4220 Shawnee Mission Parkway, Fairway, KS, 66205, USA.,Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Ravi K Sajja
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Patrick van Dreden
- Clinical Research Department, R&D, Diagnostica Stago, Gennevilliers, France
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA.
| |
Collapse
|
335
|
Bhat NR. Vasculoprotection as a Convergent, Multi-Targeted Mechanism of Anti-AD Therapeutics and Interventions. J Alzheimers Dis 2016; 46:581-91. [PMID: 26402511 DOI: 10.3233/jad-150098] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Using a variety of animal models of Alzheimer's disease (AD), there have been a number of recent studies reporting varying degrees of success with anti-AD therapeutics. The efficacies are often discussed in terms of the modulatory effects of the compounds tested on identified or assumed targets among the known (or proposed) pathogenic and neuroprotective mechanisms, largely within the context of the dominant amyloid cascade hypothesis. However, it is clear that several of the relatively more efficacious treatments tend to be multifunctional and target multiple pathological processes associated with AD including most commonly, oxidative and metabolic stress and neuroinflammation. Increasing evidence suggests that vascular and neurodegenerative pathologies often co-exist and that neurovascular dysfunction plays a critical role in the development or progression of AD. In this review, we will discuss the significance of vasculoprotection or neurovascular unit integrity as a common, multi-targeted mechanism underlying the reported efficacy of a majority of anti-AD therapeutics--amyloid-targeted or otherwise--while providing a strong support for future neurovascular-based treatment strategies and interventions.
Collapse
|
336
|
Obtulowicz P, Lech W, Strojek L, Sarnowska A, Domanska-Janik K. Induction of Endothelial Phenotype from Wharton's Jelly-Derived MSCs and Comparison of Their Vasoprotective and Neuroprotective Potential with Primary WJ-MSCs in CA1 Hippocampal Region Ex Vivo. Cell Transplant 2016; 25:715-27. [DOI: 10.3727/096368915x690369] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Ischemic stroke results in violent impairment of tissue homeostasis leading to severe perturbation within the neurovascular unit (NVU) during the recovery period. The aim of this study was to assess the potential of mesenchymal stem cells (MSCs) originating from Wharton's jelly (WJ) to differentiate into functionally competent cells of endothelial lineage (WJ-EPCs). The protective effect(s) of either primary WJ-MSCs or induced WJ-EPCs was investigated and compared after oxygen–glucose deprivation (OGD) of hippocampal organotypic slices (OHC) in the indirect coculture model. WJ-MSCs, primed in EGM-2 (Lonza commercial medium) under 5% O2, acquired cobblestone endothelial-like morphology, formed capillary-like structures and actively took up DiI-Ac-LDL. Both cell types (WJ-MSCs and WJ-EPCs) were positive for CD73, CD90, CD105, VEGFR-2, and VEGF, but only endothelial-like culture expressed vWF and PECAM-1 markers at significant levels. In the presence of either WJ-MSCs or WJ-EPCs in the compartment below OGD-injured slices, cell death and vascular atrophy in the hypoxia-sensitive CA1 region were substantially decreased. This suggests that a paracrine mechanism may mediate WJ-MSC- and WJ-EPC-dependent protection. Thus, finally, we estimated secretion of the neuro/angio/immunomodulatory molecules IL-6, TGF-β1, and VEGF by these cell cultures. We have found that release of TGF-β1 and IL-6 was TLR ligand [LPS and Poly(I:C)] concentration dependent and stronger in WJ-EPC than WJ-MSC cultures. Simultaneously, the uneven pattern of TLR receptors and modulatory cytokine gene expression was confirmed also on qRT-PCR level, but no significant differences were noticed between WJ-EPC and primary WJ-MSC cultures.
Collapse
Affiliation(s)
- Patrycja Obtulowicz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Wioletta Lech
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz Strojek
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Sarnowska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
337
|
Karamanos Y, Pottiez G. Proteomics and the blood-brain barrier: how recent findings help drug development. Expert Rev Proteomics 2016; 13:251-8. [PMID: 26778576 DOI: 10.1586/14789450.2016.1143780] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The drug discovery and development processes are divided into different stages separated by milestones to indicate that significant progress has been made and that certain criteria for target validation, hits, leads and candidate drugs have been met. Proteomics is a promising approach for the identification of protein targets and biochemical pathways involved in disease process and thus plays an important role in several stages of the drug development. The blood-brain barrier is considered as a major bottleneck when trying to target new compounds to treat neurodegenerative diseases. Based on the survey of recent findings and with a projection on expected improvements, this report attempt to address how proteomics participates in drug development.
Collapse
Affiliation(s)
- Yannis Karamanos
- a Laboratoire de la Barrière Hématoencéphalique (LBHE) , Univesrité d'Artois EA2465 , Lens , France
| | - Gwënaël Pottiez
- a Laboratoire de la Barrière Hématoencéphalique (LBHE) , Univesrité d'Artois EA2465 , Lens , France
| |
Collapse
|
338
|
Chen H, Liu N, Li Y, Chen F, Zhu G. Permeability imaging in cerebrovascular diseases: applications and progress in research. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s40809-016-0015-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
339
|
Xue Q, Liu Y, He R, Yang S, Tong J, Li X, Chen Y, Xu X. Lyophilized Powder of Catalpol and Puerarin Protects Neurovascular Unit from Stroke. Int J Biol Sci 2016; 12:367-80. [PMID: 27019622 PMCID: PMC4807157 DOI: 10.7150/ijbs.14059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/08/2015] [Indexed: 01/06/2023] Open
Abstract
Hunting for an effective medicine for brain stroke has been a medical task in neuroscience for decades. The present research showed that the lyophilized Powder of Catalpol and Puerarin (C-P) in all the tested doses (65.4 mg/kg, 32.7 mg/kg, 16.4 mg/kg) significantly reduced the neurological deficiency, infarct volume and apoptotic cells in ischemic/reperfusion (I/R) rats. It also promoted astrocyte processes and prolonged neuron axons in infarct area. Further, it decreased MDA, NO, NF-κB/p65, TNF-α, IL-1β and IL-6 and enhanced the EPOR and GAF-43. 65.4 mg/kg and 32.7 mg/kg C-P could up-regulated EPO and VEGF significantly. In vitro, 49 μg/mL and 24.5 μg/mL C-P decreased the leakage of sodium fluorescein and increased the activity of γ-GTP. Additionally, it increased SOD and decreased MDA, NO, and LDH and decreased NF-κB/p65, TNF-α, IL-1β and IL-6 and unregulated EPO, EPOR, VEGF, and GAP-43. Only the dose of 49 μg/mL increased TEER and Claudin-5 and turned the typically damaged morphologies of neurons, astrocytes and endothelium into a favorable trend. These data imply that C-P improved the recovery of neurological deficiency in motor, sense, balance and reflex, and protected the whole NVU by anti-oxidative stress, anti-inflammation and up-regulating some protective factors. This research provides a candidate medicine for brain stroke and, at the same time, a pattern for drug study targeting NVU in vitro.
Collapse
Affiliation(s)
- Qiang Xue
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yang Liu
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Ran He
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Sheng Yang
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Jie Tong
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Xu Li
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yi Chen
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Xiaoyu Xu
- 1. College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing 400715, China;; 2. Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China;; 3. Institute of Chinese Medicine, Southwest University, Chongqing 400715, China
| |
Collapse
|
340
|
The neuropathology and cerebrovascular mechanisms of dementia. J Cereb Blood Flow Metab 2016; 36:172-86. [PMID: 26174330 PMCID: PMC4758551 DOI: 10.1038/jcbfm.2015.164] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/12/2015] [Accepted: 06/15/2015] [Indexed: 12/23/2022]
Abstract
The prevalence of dementia is increasing in our aging population at an alarming rate. Because of the heterogeneity of clinical presentation and complexity of disease neuropathology, dementia classifications remain controversial. Recently, the National Plan to address Alzheimer’s Disease prioritized Alzheimer’s disease-related dementias to include: Alzheimer’s disease, dementia with Lewy bodies, frontotemporal dementia, vascular dementia, and mixed dementias. While each of these dementing conditions has their unique pathologic signature, one common etiology shared among all these conditions is cerebrovascular dysfunction at some point during the disease process. The goal of this comprehensive review is to summarize the current findings in the field and address the important contributions of cerebrovascular, physiologic, and cellular alterations to cognitive impairment in these human dementias. Specifically, evidence will be presented in support of small-vessel disease as an underlying neuropathologic hallmark of various dementias, while controversial findings will also be highlighted. Finally, the molecular mechanisms shared among all dementia types including hypoxia, oxidative stress, mitochondrial bioenergetics, neuroinflammation, neurodegeneration, and blood–brain barrier permeability responsible for disease etiology and progression will be discussed.
Collapse
|
341
|
Goodin DS. The epidemiology of multiple sclerosis: insights to a causal cascade. HANDBOOK OF CLINICAL NEUROLOGY 2016; 138:173-206. [PMID: 27637959 DOI: 10.1016/b978-0-12-802973-2.00011-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
MS-pathogenesis involves both genetic-susceptibility and environmental determinants. Three (or more) sequential environmental-factors are implicated. The first acts near birth, the second acts during childhood/adolescence, and the third acts subsequently. Two candidate factors (vitamin D deficiency and Epstein-Barr viral infection) seem particularly well-suited to the first two environmental-events but other factors (e.g., obesity and smoking behavior) seem also to be involved in the causal scheme. MS-pathogenesis can be modeled by incorporating both the environmental and genetic-factors into a causal scheme, which can then help to explain some of the changes in MS-epidemiology (e.g., increasing disease-prevalence, changing sex-ratio, and regional-variations in monozygotic-twin-concordance-rates), which have been taking place recently. This model suggests that genetic-susceptibility is overwhelmingly the most important determinant of MS and that, at least, 92.5% of individuals (and likely much more) are, essentially, incapable of developing MS, regardless of their specific environmental-exposures. Nevertheless, the genetics is complex and the contribution of any specific gene to MS-susceptibility seems to be quite modest. Thus, even for the DRB1*1501 allele (the strongest known MS-susceptibility marker), most carriers are not in the genetically-susceptible group. Moreover, 45-50% of individuals with MS lack this allele entirely and some of the haplotypes that carry this allele don't also confer any disease-risk. Finally, because the prevalence of genetic-susceptibility seems to be so similar throughout North America and Europe, and despite the crucial importance of a person's genetic make-up to disease pathogenesis, it is the environmental-factors, which largely responsible for the observed regional variations in disease-characteristics. Thus, despite MS being more common in women, men are more likely to be genetically-susceptible. This apparent paradox seems to relate to the fact that women are much more responsive than men to the recent changes in environmental-exposure (whatever these have been). These gender-differences may help to explain changes in the sex-ratio and the increasing disease-prevalence, which have both been observed recently. The potential importance of these conclusions regarding the role of environment in MS-pathogenesis is that they open the door to the possibility of pursuing strategies for primary primary disease prevention in the future.
Collapse
Affiliation(s)
- D S Goodin
- Multiple Sclerosis Center at the University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
342
|
Llombart V, García-Berrocoso T, Bech-Serra JJ, Simats A, Bustamante A, Giralt D, Reverter-Branchat G, Canals F, Hernández-Guillamon M, Montaner J. Characterization of secretomes from a human blood brain barrier endothelial cells in-vitro model after ischemia by stable isotope labeling with aminoacids in cell culture (SILAC). J Proteomics 2015; 133:100-112. [PMID: 26718731 DOI: 10.1016/j.jprot.2015.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/04/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
Abstract
UNLABELLED The human immortalized brain endothelial cell line hCMEC/D3 is considered a simple in-vitro model of the blood-brain-barrier. Our aim was to characterize changes in the secretome of hCMEC/D3 subjected to oxygen and glucose deprivation (OGD) to identify new proteins altered after ischemia and that might trigger blood-brain-barrier disruption and test their potential as blood biomarkers for ischemic stroke. Using a quantitative proteomic approach based on SILAC, 19 proteins were found differentially secreted between OGD and normoxia/normoglycemia conditions. Among the OGD-secreted proteins, protein folding was the main molecular function identified and for the main canonical pathways there was an enrichment in epithelial adherens junctions and aldosterone signaling. Western blot was used to verify the MS results in a set of 9 differentially secreted proteins and 5 of these were analyzed in serum samples of 38 ischemic stroke patients, 18 stroke-mimicking conditions and 18 healthy controls. SIGNIFICANCE "We characterized changes in the secretome of hCMEC/D3 cells after an ischemic insult by SILAC and identified proteins associated with ischemia that might be involved in the disruption of the blood-brain barrier. Besides we analyzed the putative potential of the candidate proteins to become biomarkers for the diagnosis of ischemic stroke.
Collapse
Affiliation(s)
- Victor Llombart
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Joan Josep Bech-Serra
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Alba Simats
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Dolors Giralt
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Gemma Reverter-Branchat
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Francesc Canals
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
343
|
Lucke-Wold BP, Logsdon AF, Smith KE, Turner RC, Alkon DL, Tan Z, Naser ZJ, Knotts CM, Huber JD, Rosen CL. Bryostatin-1 Restores Blood Brain Barrier Integrity following Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2015; 52:1119-1134. [PMID: 25301233 PMCID: PMC5000781 DOI: 10.1007/s12035-014-8902-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023]
Abstract
Recent wars in Iraq and Afghanistan have accounted for an estimated 270,000 blast exposures among military personnel. Blast traumatic brain injury (TBI) is the 'signature injury' of modern warfare. Blood brain barrier (BBB) disruption following blast TBI can lead to long-term and diffuse neuroinflammation. In this study, we investigate for the first time the role of bryostatin-1, a specific protein kinase C (PKC) modulator, in ameliorating BBB breakdown. Thirty seven Sprague-Dawley rats were used for this study. We utilized a clinically relevant and validated blast model to expose animals to moderate blast exposure. Groups included: control, single blast exposure, and single blast exposure + bryostatin-1. Bryostatin-1 was administered i.p. 2.5 mg/kg after blast exposure. Evan's blue, immunohistochemistry, and western blot analysis were performed to assess injury. Evan's blue binds to albumin and is a marker for BBB disruption. The single blast exposure caused an increase in permeability compared to control (t = 4.808, p < 0.05), and a reduction back toward control levels when bryostatin-1 was administered (t = 5.113, p < 0.01). Three important PKC isozymes, PKCα, PKCδ, and PKCε, were co-localized primarily with endothelial cells but not astrocytes. Bryostatin-1 administration reduced toxic PKCα levels back toward control levels (t = 4.559, p < 0.01) and increased the neuroprotective isozyme PKCε (t = 6.102, p < 0.01). Bryostatin-1 caused a significant increase in the tight junction proteins VE-cadherin, ZO-1, and occludin through modulation of PKC activity. Bryostatin-1 ultimately decreased BBB breakdown potentially due to modulation of PKC isozymes. Future work will examine the role of bryostatin-1 in preventing chronic neurodegeneration following repetitive neurotrauma.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Aric F Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Kelly E Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, 26506, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Office of Professional Studies in Health Sciences, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Chelsea M Knotts
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jason D Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Charles L Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV, 26506-9183, USA.
| |
Collapse
|
344
|
Hung VKL, Yeung PKK, Lai AKW, Ho MCY, Lo ACY, Chan KC, Wu EXK, Chung SSM, Cheung CW, Chung SK. Selective astrocytic endothelin-1 overexpression contributes to dementia associated with ischemic stroke by exaggerating astrocyte-derived amyloid secretion. J Cereb Blood Flow Metab 2015; 35:1687-96. [PMID: 26104290 PMCID: PMC4640314 DOI: 10.1038/jcbfm.2015.109] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 04/26/2015] [Accepted: 04/28/2015] [Indexed: 12/27/2022]
Abstract
Endothelin-1 (ET-1) is synthesized by endothelial cells and astrocytes in stroke and in brains of Alzheimer's disease patients. Our transgenic mice with ET-1 overexpression in the endothelial cells (TET-1) showed more severe blood-brain barrier (BBB) breakdown, neuronal apoptosis, and glial reactivity after 2-hour transient middle cerebral artery occlusion (tMCAO) with 22-hour reperfusion and more severe cognitive deficits after 30 minutes tMCAO with 5 months reperfusion. However, the role of astrocytic ET-1 in contributing to poststroke cognitive deficits after tMCAO is largely unknown. Therefore, GET-1 mice were challenged with tMCAO to determine its effect on neurologic and cognitive deficit. The GET-1 mice transiently displayed a sensorimotor deficit after reperfusion that recovered shortly, then more severe deficit in spatial learning and memory was observed at 3 months after ischemia compared with that of the controls. Upregulation of TNF-α, cleaved caspase-3, and Thioflavin-S-positive aggregates was observed in the ipsilateral hemispheres of the GET-1 brains as early as 3 days after ischemia. In an in vitro study, ET-1 overexpressing astrocytic cells showed amyloid secretion after hypoxia/ischemia insult, which activated endothelin A (ETA) and endothelin B (ETB) receptors in a PI3K/AKT-dependent manner, suggesting role of astrocytic ET-1 in dementia associated with stroke by astrocyte-derived amyloid production.
Collapse
Affiliation(s)
- Victor K L Hung
- Department of Anatomy, The University of Hong Kong, HKSAR, China
| | - Patrick K K Yeung
- Department of Anatomy, The University of Hong Kong, HKSAR, China.,School of Biomedical Sciences, The University of Hong Kong, HKSAR, China
| | - Angela K W Lai
- Department of Anatomy, The University of Hong Kong, HKSAR, China
| | - Maggie C Y Ho
- Department of Anatomy, The University of Hong Kong, HKSAR, China
| | - Amy C Y Lo
- Department of Anatomy, The University of Hong Kong, HKSAR, China
| | - Kevin C Chan
- University of Biomedical Imaging and Signal Processing, The University of Hong Kong, HKSAR, China
| | - Ed X K Wu
- University of Biomedical Imaging and Signal Processing, The University of Hong Kong, HKSAR, China
| | | | - Chi W Cheung
- Department of Anaesthesiology, The University of Hong Kong, HKSAR, China.,Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, HKSAR, China
| | - Sookja K Chung
- Department of Anatomy, The University of Hong Kong, HKSAR, China.,School of Biomedical Sciences, The University of Hong Kong, HKSAR, China.,Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, HKSAR, China.,The State Key Laboratory of Pharmaceutical Biotechnology, Zhuhai, Guandong, China
| |
Collapse
|
345
|
Goodin DS, Reder AT, Bermel RA, Cutter GR, Fox RJ, John GR, Lublin FD, Lucchinetti CF, Miller AE, Pelletier D, Racke MK, Trapp BD, Vartanian T, Waubant E. Relapses in multiple sclerosis: Relationship to disability. Mult Scler Relat Disord 2015; 6:10-20. [PMID: 27063617 DOI: 10.1016/j.msard.2015.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 08/21/2015] [Accepted: 09/02/2015] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a recurrent inflammatory disease of the central nervous system, which ultimately causes substantial disability in many patients. A key clinical feature of this disease is the occurrence of relapses, consisting of episodes of neurological dysfunction followed by periods of remission. This review considers in detail the importance of the occurrence of relapses to the ultimate course of MS and the impact of relap setreatment (both acutely and prophylactically) on the long-term outcome for individuals. The ultimate goal of therapy in MS is the reduction of long-term disability. Clinical trials in MS, however, typically only extend for a very short time period compared to the time it takes for disability to evolve. Consequently, short-term outcome measures that are associated with, and predict, future disability need to be identified. In this regard, not only are relapses a characteristic feature of MS, they have also been proven to be associated with the occurrence of long-term disability. Moreover, treatments that reduce the number and severity of these attacks improve the long-term prognosis.
Collapse
Affiliation(s)
- Douglas S Goodin
- Multiple Sclerosis Center, University of California, San Francisco Medical Center, San Francisco, CA, United States; Department of Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, United States.
| | - Anthony T Reder
- Department of Neurology, The University of Chicago, Chicago, IL, United States
| | - Robert A Bermel
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, United States
| | - Gary R Cutter
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Gareth R John
- Multiple Sclerosis Research Laboratory, Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Friedman Brain Institute, New York, NY, United States; Department of Neurology, Mount Sinai School of Medicine, New York, NY, United States
| | - Fred D Lublin
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Aaron E Miller
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Daniel Pelletier
- Neuro-Immunology Division and Yale Multiple Sclerosis Center, Advanced Imaging in Multiple Sclerosis (AIMS) Laboratory, Yale University School of Medicine, New Haven, CT, United States
| | - Michael K Racke
- Department of Neurology, Wexner Medical Center at The Ohio State University, Columbus, OH, United States
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Timothy Vartanian
- Judith Jaffe Multiple Sclerosis Center, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medical College, United States
| | - Emmanuelle Waubant
- UCSF Regional Pediatric MS Center, Race to Erase MS, San Francisco, CA, United States
| |
Collapse
|
346
|
Ji J, Yan H, Chen ZZ, Zhao Z, Yang DD, Sun XL, Shi YP. Iptakalim protects against ischemic injury by improving neurovascular unit function in the mouse brain. Clin Exp Pharmacol Physiol 2015; 42:766-71. [DOI: 10.1111/1440-1681.12426] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Juan Ji
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Hui Yan
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Zheng-Zhen Chen
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Zhan Zhao
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Dan-Dan Yang
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | - Xiu-Lan Sun
- Department of Pharmacology; Nanjing Medical University; Nanjing China
| | | |
Collapse
|
347
|
Disruption in the Blood-Brain Barrier: The Missing Link between Brain and Body Inflammation in Bipolar Disorder? Neural Plast 2015; 2015:708306. [PMID: 26075104 PMCID: PMC4444594 DOI: 10.1155/2015/708306] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 02/02/2015] [Accepted: 02/05/2015] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) regulates the transport of micro- and macromolecules between the peripheral blood and the central nervous system (CNS) in order to maintain optimal levels of essential nutrients and neurotransmitters in the brain. In addition, the BBB plays a critical role protecting the CNS against neurotoxins. There has been growing evidence that BBB disruption is associated with brain inflammatory conditions such as Alzheimer's disease and multiple sclerosis. Considering the increasing role of inflammation and oxidative stress in the pathophysiology of bipolar disorder (BD), here we propose a novel model wherein transient or persistent disruption of BBB integrity is associated with decreased CNS protection and increased permeability of proinflammatory (e.g., cytokines, reactive oxygen species) substances from the peripheral blood into the brain. These events would trigger the activation of microglial cells and promote localized damage to oligodendrocytes and the myelin sheath, ultimately compromising myelination and the integrity of neural circuits. The potential implications for research in this area and directions for future studies are discussed.
Collapse
|
348
|
Muñoz MF, Puebla M, Figueroa XF. Control of the neurovascular coupling by nitric oxide-dependent regulation of astrocytic Ca(2+) signaling. Front Cell Neurosci 2015; 9:59. [PMID: 25805969 PMCID: PMC4354411 DOI: 10.3389/fncel.2015.00059] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/07/2015] [Indexed: 12/28/2022] Open
Abstract
Neuronal activity must be tightly coordinated with blood flow to keep proper brain function, which is achieved by a mechanism known as neurovascular coupling. Then, an increase in synaptic activity leads to a dilation of local parenchymal arterioles that matches the enhanced metabolic demand. Neurovascular coupling is orchestrated by astrocytes. These glial cells are located between neurons and the microvasculature, with the astrocytic endfeet ensheathing the vessels, which allows fine intercellular communication. The neurotransmitters released during neuronal activity reach astrocytic receptors and trigger a Ca2+ signaling that propagates to the endfeet, activating the release of vasoactive factors and arteriolar dilation. The astrocyte Ca2+ signaling is coordinated by gap junction channels and hemichannels formed by connexins (Cx43 and Cx30) and channels formed by pannexins (Panx-1). The neuronal activity-initiated Ca2+ waves are propagated among neighboring astrocytes directly via gap junctions or through ATP release via connexin hemichannels or pannexin channels. In addition, Ca2+ entry via connexin hemichannels or pannexin channels may participate in the regulation of the astrocyte signaling-mediated neurovascular coupling. Interestingly, nitric oxide (NO) can activate connexin hemichannel by S-nitrosylation and the Ca2+-dependent NO-synthesizing enzymes endothelial NO synthase (eNOS) and neuronal NOS (nNOS) are expressed in astrocytes. Therefore, the astrocytic Ca2+ signaling triggered in neurovascular coupling may activate NO production, which, in turn, may lead to Ca2+ influx through hemichannel activation. Furthermore, NO release from the hemichannels located at astrocytic endfeet may contribute to the vasodilation of parenchymal arterioles. In this review, we discuss the mechanisms involved in the regulation of the astrocytic Ca2+ signaling that mediates neurovascular coupling, with a special emphasis in the possible participation of NO in this process.
Collapse
Affiliation(s)
- Manuel F Muñoz
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Mariela Puebla
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Xavier F Figueroa
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
349
|
Wang P, Guan YF, Li WL, Lu GC, Liu JM, Miao CY. Nicotinamide phosphoribosyltransferase facilitates post-stroke angiogenesis. CNS Neurosci Ther 2015; 21:475-7. [PMID: 25753018 DOI: 10.1111/cns.12388] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 12/27/2022] Open
Affiliation(s)
- Pei Wang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
350
|
Palmiotti CA, Prasad S, Naik P, Abul KMD, Sajja RK, Achyuta AH, Cucullo L. In vitro cerebrovascular modeling in the 21st century: current and prospective technologies. Pharm Res 2014; 31:3229-50. [PMID: 25098812 PMCID: PMC4225221 DOI: 10.1007/s11095-014-1464-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/24/2014] [Indexed: 12/26/2022]
Abstract
The blood-brain barrier (BBB) maintains the brain homeostasis and dynamically responds to events associated with systemic and/or rheological impairments (e.g., inflammation, ischemia) including the exposure to harmful xenobiotics. Thus, understanding the BBB physiology is crucial for the resolution of major central nervous system CNS) disorders challenging both health care providers and the pharmaceutical industry. These challenges include drug delivery to the brain, neurological disorders, toxicological studies, and biodefense. Studies aimed at advancing our understanding of CNS diseases and promoting the development of more effective therapeutics are primarily performed in laboratory animals. However, there are major hindering factors inherent to in vivo studies such as cost, limited throughput and translational significance to humans. These factors promoted the development of alternative in vitro strategies for studying the physiology and pathophysiology of the BBB in relation to brain disorders as well as screening tools to aid in the development of novel CNS drugs. Herein, we provide a detailed review including pros and cons of current and prospective technologies for modelling the BBB in vitro including ex situ, cell based and computational (in silico) models. A special section is dedicated to microfluidic systems including micro-BBB, BBB-on-a-chip, Neurovascular Unit-on-a-Chip and Synthetic Microvasculature Blood-brain Barrier.
Collapse
Affiliation(s)
| | - Shikha Prasad
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Pooja Naik
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Kaisar MD Abul
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ravi K. Sajja
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | | | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|