1
|
Serneels PJ, De Schutter JD, De Groef L, Moons L, Bergmans S. Oligodendroglial heterogeneity in health, disease, and recovery: deeper insights into myelin dynamics. Neural Regen Res 2025; 20:3179-3192. [PMID: 39665821 PMCID: PMC11881716 DOI: 10.4103/nrr.nrr-d-24-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 12/13/2024] Open
Abstract
Decades of research asserted that the oligodendroglial lineage comprises two cell types: oligodendrocyte precursor cells and oligodendrocytes. However, recent studies employing single-cell RNA sequencing techniques have uncovered novel cell states, prompting a revision of the existing terminology. Going forward, the oligodendroglial lineage should be delineated into five distinct cell states: oligodendrocyte precursor cells, committed oligodendrocyte precursor cells, newly formed oligodendrocytes, myelin-forming oligodendrocytes, and mature oligodendrocytes. This new classification system enables a deeper understanding of the oligodendroglia in both physiological and pathological contexts. Adopting this uniform terminology will facilitate comparison and integration of data across studies. This, including the consolidation of findings from various demyelinating models, is essential to better understand the pathogenesis of demyelinating diseases. Additionally, comparing injury models across species with varying regenerative capacities can provide insights that may lead to new therapeutic strategies to overcome remyelination failure. Thus, by standardizing terminology and synthesizing data from diverse studies across different animal models, we can enhance our understanding of myelin pathology in central nervous system disorders such as multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis, all of which involve oligodendroglial and myelin dysfunction.
Collapse
Affiliation(s)
- Pieter-Jan Serneels
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Julie D. De Schutter
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Lies De Groef
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Cellular Communication & Neurodegeneration Research Group, Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Steven Bergmans
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| |
Collapse
|
2
|
Latham AS, Geer CE, Ackart DF, Weninger KN, Gross CC, Podell BK, Basaraba RJ, Moreno JA. Immune cell infiltration and modulation of the blood-brain barrier in a guinea pig model of tuberculosis: Observations without evidence of bacterial dissemination to the brain. PLoS One 2024; 19:e0307577. [PMID: 39739680 DOI: 10.1371/journal.pone.0307577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 07/08/2024] [Indexed: 01/02/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, is a chronic inflammatory disease. Although typically associated with inflammation of the lungs and other peripheral tissues, increasing evidence has uncovered neurological consequences attributable to Mtb infection. These include deficits in memory and cognition, increased risk for neurodegenerative disease, and progressive neuropathology. Although the neurological effects of the disease, without CNS infection, have been characterized, the mechanism of neurotoxicity is unknown. We hypothesized that alterations to the blood-brain barrier (BBB) allows peripheral immune cells to enter the brain, initiating a neuroinflammatory response. To test this hypothesis, guinea pigs were exposed by aerosol to a laboratory and a clinical Mtb strain for 15 days. Following Mtb infection, proteins critical to BBB function, including claudin V and collagen IV, are modulated without evidence of bacterial dissemination to the brain. This is correlated with increased contact of astrocytic processes to vessels in the brain, as well as increased expression of the water channel protein aquaporin 4 (AQP4) on endfeet. Upon further investigation, we discovered the potential role of glial reactivity, which is increased following infection with both bacterial strains, in the progression of BBB changes and, ultimately, the permeability of peripheral immune cells into the brain. Through these data, we have obtained a preliminary understanding of the mechanisms of cellular stress in the brain following pulmonary Mtb infection which should be further investigated in future studies.
Collapse
Affiliation(s)
- Amanda S Latham
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Brain Research Center, Colorado State University, Fort Collins, Colorado, United States of America
| | - Charlize E Geer
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - David F Ackart
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kristin N Weninger
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Biomedical Science, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Chase C Gross
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Randall J Basaraba
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- Brain Research Center, Colorado State University, Fort Collins, Colorado, United States of America
- Center for Healthy Aging, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
3
|
Rêgo DSB, Calió ML, Filev R, Mello LE, Leslie ATFS. Long-term Effects of Cannabidiol and/or Fentanyl Exposure in Rats Submitted to Neonatal Pain. THE JOURNAL OF PAIN 2024; 25:715-729. [PMID: 37820846 DOI: 10.1016/j.jpain.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/13/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
The current study aimed to evaluate anxiety behavior, hippocampal ionized calcium-binding adaptor molecule 1 (Iba1) and cannabinoid receptor 1 (CB1) gene expression, and nociceptive response in adulthood after a combination of fentanyl and cannabidiol (CBD) for nociceptive stimuli induced during the first week of life in rats. Complete Freund's adjuvant-induced inflammatory nociceptive insult on postnatal day (PN) 1 and PN3. Both fentanyl and CBD were used alone or in combination from PN1 to PN7. Behavioral and nociceptive tests were performed at PN60 and PN62. The expression of the microglial calcium-binding proteins Iba1 and CB1 was detected in the hippocampus using reverse Quantitative polymerase chain reaction (qPCR) and immunohistochemistry. Our results suggest that the anxiety behavior response and immune activation in adult life depend on the CBD dose combined with fentanyl for the nociceptive stimuli induced during the first week of life. Treatment of neonatal nociceptive insult with CBD and opioids showed significant dose-dependent and male-female differences. The increased gene expression in the hippocampus of the analyzed cannabinoid gene supports this data. In addition, treatment with fentanyl led to an increase in CB1 protein expression. Moreover, the expression of Iba1 varied according to the administered dose of CBD and may or may not be associated with the opioid. A lower dose of CBD during the inflammatory period was associated with enhanced anxiety in adult life. PERSPECTIVE: The treatment of nociceptive stimuli with CBD and opioids during the first week of life demonstrated significant sex differences in adult life on anxiety behavior and supraspinal pain sensitivity.
Collapse
Affiliation(s)
- Débora S B Rêgo
- Department of Physiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Michele Longoni Calió
- Department of Physiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Renato Filev
- Programa de Orientação e Atendimento a Dependentes (PROAD), Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Luiz E Mello
- Department of Physiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil; Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Ana T F S Leslie
- Department of Physiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
4
|
Diebold M, Fehrenbacher L, Frosch M, Prinz M. How myeloid cells shape experimental autoimmune encephalomyelitis: At the crossroads of outside-in immunity. Eur J Immunol 2023; 53:e2250234. [PMID: 37505465 DOI: 10.1002/eji.202250234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/21/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of central nervous system (CNS) autoimmunity. It is most commonly used to mimic aspects of multiple sclerosis (MS), a demyelinating disorder of the human brain and spinal cord. The innate immune response displays one of the core pathophysiological features linked to both the acute and chronic stages of MS. Hence, understanding and targeting the innate immune response is essential. Microglia and other CNS resident MUs, as well as infiltrating myeloid cells, diverge substantially in terms of both their biology and their roles in EAE. Recent advances in the field show that antigen presentation, as well as disease-propagating and regulatory interactions with lymphocytes, can be attributed to specific myeloid cell types and cell states in EAE lesions, following a distinct temporal pattern during disease initiation, propagation and recovery. Furthermore, single-cell techniques enable the assessment of characteristic proinflammatory as well as beneficial cell states, and identification of potential treatment targets. Here, we discuss the principles of EAE induction and protocols for varying experimental paradigms, the composition of the myeloid compartment of the CNS during health and disease, and systematically review effects on myeloid cells for therapeutic approaches in EAE.
Collapse
Affiliation(s)
- Martin Diebold
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Luca Fehrenbacher
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Maximilian Frosch
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
6
|
Phillips O, Tubre T, Lorenco H, Batish SD, Brown W. Limbic encephalitis in a child with ovarian teratoma and influenza B. Case report and critical review of the history of autoimmune anti-N-methyl-d-aspartate receptor encephalitis. J Neuroimmunol 2021; 360:577716. [PMID: 34517152 DOI: 10.1016/j.jneuroim.2021.577716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/29/2021] [Accepted: 09/05/2021] [Indexed: 11/17/2022]
Abstract
We report the appearance of clinical symptoms and signs of N-methyl-d-Aspartate (NMDA) receptor encephalitis in a patient presenting just days after contraction of influenza B. The offending mature ovarian teratoma was identified and removed on the 10th day after the appearance of symptoms, with subsequent nearly complete resolution of symptoms over the subsequent 6 months. We provide a focused literature review of the clinical and pathophysiologic literature of anti-NMDA receptor encephalitis pertaining to influenza B virus and the pediatric population. Taken together, this study contributes to the pathophysiological understanding of anti-NMDA receptor encephalitis and aids clinicians in its early recognition and management.
Collapse
Affiliation(s)
- Oliver Phillips
- Cleveland Clinic Center for Neurological Restoration, Cleveland, OH, USA.
| | - Teddi Tubre
- Rhode Island Hospital, Department of Pathology, Providence, RI, USA
| | - Hillary Lorenco
- Rhode Island Hospital, Department of Pediatric Surgery, Providence, RI, USA
| | | | - William Brown
- Rhode Island Hospital, Department of Pediatrics, Providence, RI, USA
| |
Collapse
|
7
|
Cavalli J, Freitas MA, Gonçalves ECD, Fadanni GP, Santos AA, Raposo NRB, Dutra RC. Chia oil prevents chemical and immune-mediated inflammatory responses in mice: Evidence for the underlying mechanisms. Food Res Int 2021; 149:110703. [PMID: 34600695 DOI: 10.1016/j.foodres.2021.110703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Chia (Salvia hispanica L.) is an herbaceous plant used as omega-3 polyunsaturated fatty acid (ω-3 PUFA) source that presents a range of beneficial effects on human health. Herein, it was used a chia oil containing over than 62% of α-linolenic acid (ALA), a compound widely related to anti-inflammatory actions. Chia oil effect was tested using paw edema and mechanical hyperalgesia induced by carrageenan, and ear edema induced by croton oil, histamine, and capsaicin. Croton oil was used in both preventive and therapeutic treatment schedules of chia oil while histamine and capsaicin were used only in preventive treatment schedule. Chia oil mechanism of action was investigated using nociception and paw edema response induced by intraplantar injection of acidified saline (ASIC activator), PGE2 (prostaglandin pathway), cinnamaldehyde (TRPA1 activator), bradykinin (BK pathway), menthol (TRPM8 activator), and capsaicin (TRPV1 activator). Further, RT-PCR for inflammatory mediators (TRPA1, NF-κB, PPAR-γ, COX-2, IL-6, TNF, FPR2, FAAH, MAGL, and IL-12A) induced by carrageenan, NLRP3 inflammasome activation, and the cell viability were then accessed. Later, chia oil actions were evaluated in the experimental autoimmune encephalomyelitis (EAE), a multiple sclerosis (MS) model. Chia oil showed anti-edematogenic and anti-hyperalgesic effects when administered 1 h before pro-inflammatory stimulus - particularly carrageenan and croton oil. Moreover, chia oil upregulated the mRNA levels of COX-2 and formyl peptide receptor 2 (FPR2) while reduced IL-6 expression in the spinal cord of mice submitted to i.pl. injection of carrageenan. Interestingly, chia oil mediates antinociceptive effects in mice decreasing the nociceptive response induced by acidified saline, PGE2, and cinnamaldehyde, but not by bradykinin, menthol, and capsaicin. On the EAE model, chia oil preventively administered attenuated EAE-induced motor deficits and mechanical hyperalgesia in mice, suggesting a valuable effect of chia oil supplementation in regulating inflammatory responses and some immune functions during immune-mediated inflammatory disorders (IMID). Nonetheless, additional reports will need to assess the effect of chia oil in well-controlled clinical trials performed in MS patients.
Collapse
Affiliation(s)
- Juliana Cavalli
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil
| | - Mariana A Freitas
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil
| | - Elaine C D Gonçalves
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil; Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Guilherme P Fadanni
- Center of Innovation and Preclinical Research, 88056-000 Florianópolis, SC, Brazil
| | - Adara A Santos
- Center of Innovation and Preclinical Research, 88056-000 Florianópolis, SC, Brazil
| | - Nádia R B Raposo
- Center for Research and Innovation in Health Sciences (NUPICS), Faculty of Pharmacy, Universidade Federal de Juiz de Fora, 36036-330 Juiz de Fora, MG, Brazil
| | - Rafael C Dutra
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil; Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil; Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil.
| |
Collapse
|
8
|
Abstract
Optic neuritis (ON) is an inflammatory attack of the optic nerve that leads to visual disability. It is the most common optic neuropathy affecting healthy young adults, most commonly women aged 20-45 years. It can be idiopathic and monophasic or as part of a neurologic disease such as multiple sclerosis with recurrence and cumulative damage. Currently, there is no therapy to repair the damage from optic neuritis. Animal models are an essential tool for the understanding of the pathogenesis of optic neuritis and for the development of potential treatment strategies. Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental rodent model for human autoimmune inflammatory demyelinating diseases of the central nervous system (CNS). In this review, we discuss the latest rodent models regarding optic neuritis, focusing on EAE model, and on its recent achievements and developments.
Collapse
Affiliation(s)
- Yael Redler
- Department of Neuro-Ophthalmology, Massachusetts Eye & Ear Infirmary/Harvard Medical School, Boston, MA, United States
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Hoshino K, Hasegawa K, Kamiya H, Morimoto Y. Synapse-specific effects of IL-1β on long-term potentiation in the mouse hippocampus. Biomed Res 2018. [PMID: 28637953 DOI: 10.2220/biomedres.38.183] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Interleukin-1β (IL-1β) is a key molecule in the inflammatory responses elicited during infection and injury. It exerts local effects on synaptic plasticity by binding to IL-1 receptors that are expressed at high levels in the hippocampus. We examined the effects of IL-1β on synaptic plasticity in different hippocampal regions in acute mouse brain slices by measuring long-term potentiation (LTP). IL-1β (1 ng/mL) was applied for 30 min before LTP was induced with high-frequency stimulation (HFS). LTP was significantly impaired by either IL-1β application to the Schaffer collateral-CA1 synapses or the associational/commissural (A/C) fiber-CA3 synapses, which are both dependent on N-methyl-D-aspartate (NMDA) receptor activation. However, mossy fiber-CA3 LTP, which is expressed presynaptically in an NMDA-independent manner, was not impaired by IL-1β. Our results demonstrate that IL-1β exerts variable effects on LTP at different kinds of synapses, indicating that IL-1β has synapse-specific effects on hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Koji Hoshino
- Department of Anesthesiology and Cristical Care Medicine, Hokkaido University Graduate School of medicine.,Department of Neurobiology, Hokkaido University Graduate School of Medicine
| | - Kan Hasegawa
- Department of Anesthesiology and Cristical Care Medicine, Hokkaido University Graduate School of medicine.,Department of Neurobiology, Hokkaido University Graduate School of Medicine
| | - Haruyuki Kamiya
- Department of Neurobiology, Hokkaido University Graduate School of Medicine
| | - Yuji Morimoto
- Department of Anesthesiology and Cristical Care Medicine, Hokkaido University Graduate School of medicine
| |
Collapse
|
10
|
Bjelobaba I, Begovic-Kupresanin V, Pekovic S, Lavrnja I. Animal models of multiple sclerosis: Focus on experimental autoimmune encephalomyelitis. J Neurosci Res 2018; 96:1021-1042. [PMID: 29446144 DOI: 10.1002/jnr.24224] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressive disorder of the central nervous system (CNS) that affects more than two million people worldwide. Several animal models resemble MS pathology; the most employed are experimental autoimmune encephalomyelitis (EAE) and toxin- and/or virus-induced demyelination. In this review we will summarize our knowledge on the utility of different animal models in MS research. Although animal models cannot replicate the complexity and heterogeneity of the MS pathology, they have proved to be useful for the development of several drugs approved for treatment of MS patients. This review focuses on EAE because it represents both clinical and pathological features of MS. During the past decades, EAE has been effective in illuminating various pathological processes that occur during MS, including inflammation, CNS penetration, demyelination, axonopathy, and neuron loss mediated by immune cells.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | | | - Sanja Pekovic
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
11
|
Jandu AS, Odor PM, Vidgeon SD. Status epilepticus and anti-NMDA receptor encephalitis after resection of an ovarian teratoma. J Intensive Care Soc 2017; 17:346-352. [PMID: 28979520 DOI: 10.1177/1751143716638371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Anti-N-methyl-D-aspartate receptor encephalitis is a recently recognised autoimmune, paraneoplastic syndrome that typically presents with psychiatric disturbance, reduced conscious level and seizures. The disorder has been previously associated with ovarian teratomas. We present the case of a 35-year-old female, with a previous surgical history for resection of an ovarian teratoma, who later developed status epilepticus and anti-N-methyl-D-aspartate receptor encephalitis requiring intensive care management. Her presentation, treatment and early follow-up are described, alongside an overview of anti-N-methyl-D-aspartate receptor encephalitis pathophysiology and intensive care management.
Collapse
Affiliation(s)
- Amritpal S Jandu
- Intensive Care Department, Croydon University Hospital, Croydon, UK
| | - Peter M Odor
- Intensive Care Department, Croydon University Hospital, Croydon, UK
| | - Steven D Vidgeon
- Intensive Care Department, Croydon University Hospital, Croydon, UK
| |
Collapse
|
12
|
Raphael I, Webb J, Gomez-Rivera F, Chase Huizar CA, Gupta R, Arulanandam BP, Wang Y, Haskins WE, Forsthuber TG. Serum Neuroinflammatory Disease-Induced Central Nervous System Proteins Predict Clinical Onset of Experimental Autoimmune Encephalomyelitis. Front Immunol 2017; 8:812. [PMID: 28769926 PMCID: PMC5512177 DOI: 10.3389/fimmu.2017.00812] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/27/2017] [Indexed: 11/24/2022] Open
Abstract
There is an urgent need in multiple sclerosis (MS) patients to develop biomarkers and laboratory tests to improve early diagnosis, predict clinical relapses, and optimize treatment responses. In healthy individuals, the transport of proteins across the blood–brain barrier (BBB) is tightly regulated, whereas, in MS, central nervous system (CNS) inflammation results in damage to neuronal tissues, disruption of BBB integrity, and potential release of neuroinflammatory disease-induced CNS proteins (NDICPs) into CSF and serum. Therefore, changes in serum NDICP abundance could serve as biomarkers of MS. Here, we sought to determine if changes in serum NDICPs are detectable prior to clinical onset of experimental autoimmune encephalomyelitis (EAE) and, therefore, enable prediction of disease onset. Importantly, we show in longitudinal serum specimens from individual mice with EAE that pre-onset expression waves of synapsin-2, glutamine synthetase, enolase-2, and synaptotagmin-1 enable the prediction of clinical disease with high sensitivity and specificity. Moreover, we observed differences in serum NDICPs between active and passive immunization in EAE, suggesting hitherto not appreciated differences for disease induction mechanisms. Our studies provide the first evidence for enabling the prediction of clinical disease using serum NDICPs. The results provide proof-of-concept for the development of high-confidence serum NDICP expression waves and protein biomarker candidates for MS.
Collapse
Affiliation(s)
- Itay Raphael
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States.,Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Johanna Webb
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Francisco Gomez-Rivera
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Carol A Chase Huizar
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Rishein Gupta
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Bernard P Arulanandam
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Yufeng Wang
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - William E Haskins
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
13
|
Islam MA, Alam F, Gan SH, Cavestro C, Wong KK. Coexistence of antiphospholipid antibodies and cephalalgia. Cephalalgia 2017; 38:568-580. [DOI: 10.1177/0333102417694881] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The occurrence of antiphospholipid antibodies (aPLs) and headache comorbidity in the presence or absence of underlying autoimmune diseases remains unclear. Aim The aim of this review was to summarize the relationship between headache and aPLs based on evidences from cohort studies and case reports, in addition to examining the treatment strategies that resolved headache in aPLs-positive individuals. Methods A comprehensive literature search was conducted through PubMed, ISI Web of Science and Google Scholar. A total of 559 articles were screened and the appropriate articles were selected based on quality and level of evidence. Results Cohort studies (n = 27) from Europe, North America and Asia demonstrated comorbidity of aPLs and headache in antiphospholipid syndrome, systemic lupus erythematosus (SLE) and neuropsychiatric SLE patients. Significantly higher association between migraine and aPLs was observed (n = 170/779; p < 0.0001) in individuals without any underlying diseases. Our analysis of shortlisted case reports (n = 17) showed that a higher frequency of anticardiolipin antibodies were present in subjects with different autoimmune disorders (70.6%). Corticosteroids were highly effective in resolving headache in aPLs-positive individuals. Conclusion Higher frequency of comorbidity between aPLs and headache was observed in healthy individuals and patient cases. Therefore, experimental studies are warranted to evaluate the aPLs-induced pathogenic mechanism of headache.
Collapse
Affiliation(s)
- Md. Asiful Islam
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Fahmida Alam
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Siew Hua Gan
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | | | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
14
|
Vaitaitis GM, Yussman MG, Waid DM, Wagner DH. Th40 cells (CD4+CD40+ Tcells) drive a more severe form of Experimental Autoimmune Encephalomyelitis than conventional CD4 T cells. PLoS One 2017; 12:e0172037. [PMID: 28192476 PMCID: PMC5305068 DOI: 10.1371/journal.pone.0172037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/30/2017] [Indexed: 12/31/2022] Open
Abstract
CD40-CD154 interaction is critically involved in autoimmune diseases, and CD4 T cells play a dominant role in the Experimental Autoimmune Encephalomyelitis (EAE) model of Multiple Sclerosis (MS). CD4 T cells expressing CD40 (Th40) are pathogenic in type I diabetes but have not been evaluated in EAE. We demonstrate here that Th40 cells drive a rapid, more severe EAE disease course than conventional CD4 T cells. Adoptively transferred Th40 cells are present in lesions in the CNS and are associated with wide spread demyelination. Primary Th40 cells from EAE-induced donors adoptively transfer EAE without further in-vitro expansion and without requiring the administration of the EAE induction regimen to the recipient animals. This has not been accomplished with primary, non-TCR-transgenic donor cells previously. If co-injection of Th40 donor cells with Freund's adjuvant (CFA) in the recipient animals is done, the disease course is more severe. The CFA component of the EAE induction regimen causes generalized inflammation, promoting expansion of Th40 cells and infiltration of the CNS, while MOG-antigen shapes the antigen-specific TCR repertoire. Those events are both necessary to precipitate disease. In MS, viral infections or trauma may induce generalized inflammation in susceptible individuals with subsequent disease onset. It will be important to further understand the events leading up to disease onset and to elucidate the contributions of the Th40 T cell subset. Also, evaluating Th40 levels as predictors of disease onset would be highly useful because if either the generalized inflammation event or the TCR-honing can be interrupted, disease onset may be prevented.
Collapse
Affiliation(s)
- Gisela M. Vaitaitis
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Martin G. Yussman
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Dan M. Waid
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - David H. Wagner
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
15
|
Nourbakhsh B, Graves J, Casper TC, Lulu S, Waldman A, Belman A, Greenberg B, Weinstock-Guttman B, Aaen G, Tillema JM, Hart J, Ness J, Rubin J, Krupp L, Gorman M, Benson L, Rodriguez M, Chitnis T, Rose J, Barcellos L, Waubant E. Dietary salt intake and time to relapse in paediatric multiple sclerosis. J Neurol Neurosurg Psychiatry 2016; 87:1350-1353. [PMID: 27343226 PMCID: PMC5370574 DOI: 10.1136/jnnp-2016-313410] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/09/2016] [Accepted: 06/05/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Salt intake was reported to be associated with increased clinical and MRI activity in adult patients with relapsing-remitting multiple sclerosis (MS). OBJECTIVE To determine if salt intake is associated with time to relapse in patients with paediatric-onset MS. METHODS Paediatric-onset MS and patients with clinically isolated syndrome (CIS) within 4 years of disease onset were recruited from 15 paediatric MS centres in the USA as part of a case-control study. Patients with available prospective relapse data subsequent to enrolment were included in this project. Dietary sodium intake was assessed by self-report questionnaire using the validated Block Kids Food Screener. Cox proportional-hazards regression models were employed to determine the association of sodium density, excess sodium intake and sodium density tertiles with time to relapse following study enrolment, adjusting for several confounders. RESULTS 174 relapsing-remitting MS/CIS patients were included in this analysis (mean age of 15.0 years, and 64.9% females). Median duration of follow-up was 1.8 years. In an unadjusted analysis, density of daily sodium intake was not associated with time to relapse, and patients with excess sodium intake had no decrease in time to relapse as compared with patients with non-excess sodium intake. The multivariable analysis demonstrated that patients in the medium and high tertile of sodium density had a HR of 0.69 (95% CI 0.37 to 1.30, p=0.25) and 1.37 (95% CI 0.74 to 2.51, p=0.32) compared with patients in the lowest tertile, respectively. CONCLUSIONS Higher salt intake was not associated with decreased time to relapse in patients with paediatric-onset MS.
Collapse
Affiliation(s)
| | - Jennifer Graves
- UCSF Regional Paediatric MS Center, San Francisco, California, USA
| | - T Charles Casper
- Department of Paediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Sabeen Lulu
- UCSF Regional Paediatric MS Center, San Francisco, California, USA
| | - Amy Waldman
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anita Belman
- Department of Neurology, SUNY Stony Brook, Stony Brook, New York, USA
| | | | | | - Gregory Aaen
- Department of Child Neurology, Loma Linda University, Loma Linda, California, USA
| | | | - Janace Hart
- UCSF Regional Paediatric MS Center, San Francisco, California, USA
| | - Jayne Ness
- Alabama Paediatric MS Center, Birmingham, Alabama, USA
| | - Jennifer Rubin
- Department of Paediatric Neurology, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lauren Krupp
- Department of Neurology, New York University, New York, New York, USA
| | - Mark Gorman
- Partners Paediatric MS Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Leslie Benson
- Partners Paediatric MS Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tanuja Chitnis
- Partners Paediatric MS Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John Rose
- Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Lisa Barcellos
- Department of Epidemiology, UC Berkeley, Berkeley, California, USA
| | | | | |
Collapse
|
16
|
El Temsahy MM, El Kerdany EDH, Eissa MM, Shalaby TI, Talaat IM, Mogahed NMFH. The effect of chitosan nanospheres on the immunogenicity of Toxoplasma lysate vaccine in mice. J Parasit Dis 2016; 40:611-626. [PMID: 27605755 PMCID: PMC4996159 DOI: 10.1007/s12639-014-0546-z] [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: 05/18/2014] [Accepted: 08/22/2014] [Indexed: 12/13/2022] Open
Abstract
Toxoplasmosis, a zoonotic parasitic disease, is a huge challenge for which there is no effective vaccine up till now. In this study, chitosan nanospheres encapsulated with Toxoplasma lysate vaccine was evaluated for its ability to protect mice against both acute and chronic toxoplasmosis models of infection. Results showed that chitosan nanospheres were equally effective to Freund's incomplete adjuvant (FIA) in enhancing the efficacy of Toxoplasma lysate vaccine. The effectiveness was demonstrated by the delayed death of vaccinated mice following challenge either with virulent RH or avirulent Me49 strains, the significant decrease in parasite density in different organs, significant increase in the humoral and cellular immune response (IgG and IFN γ) with a marked reduction of pathological changes in the different organs. However chitosan nanospheres were superior to FIA due to their cost effective preparation and much less necrotic changes induced in the studied organs. The success of chitosan polymer as an alternative to commonly used adjuvants paves the way for the use of other newly developed polymers to be used in the field of vaccine development.
Collapse
Affiliation(s)
- Mona M. El Temsahy
- Department of Medical Parasitology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Eman D. H. El Kerdany
- Department of Medical Parasitology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Maha M. Eissa
- Department of Medical Parasitology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Thanaa I. Shalaby
- Department of Biophysics and Biomedics, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Iman M. Talaat
- Department of Pathology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Nermine M. F. H. Mogahed
- Department of Medical Parasitology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| |
Collapse
|
17
|
Vistoropsky Y, Heiblum R, Smorodinsky NI, Barnea A. Active immunization against vasoactive intestinal polypeptide decreases neuronal recruitment and inhibits reproduction in zebra finches. J Comp Neurol 2016; 524:2516-28. [PMID: 26801210 DOI: 10.1002/cne.23971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/26/2015] [Accepted: 01/19/2016] [Indexed: 01/21/2023]
Abstract
Neurogenesis and neuronal recruitment occur in adult brains of many vertebrates, and the hypothesis is that these phenomena contribute to the brain plasticity that enables organisms to adjust to environmental changes. In mammals, vasoactive intestinal polypeptide (VIP) is known to have many neuroprotective properties, but in the avian brain, although widely distributed, its role in neuronal recruitment is not yet understood. In the present study we actively immunized adult zebra finches against VIP conjugated to KLH and compared neuronal recruitment in their brains, with brains of control birds, which were immunized against KLH. We looked at two forebrain regions: the nidopallium caudale (NC), which plays a role in vocal communication, and the hippocampus (HC), which is involved in the processing of spatial information. Our data demonstrate that active immunization against VIP reduces neuronal recruitment, inhibits reproduction, and induces molting, with no change in plasma prolactin levels. Thus, our observations suggest that VIP has a direct positive role in neuronal recruitment and reproduction in birds. J. Comp. Neurol. 524:2516-2528, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yulia Vistoropsky
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, 43107, Israel
| | - Rachel Heiblum
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, 43107, Israel
| | - Nechama-Ina Smorodinsky
- Department of Cell Research & Immunology, The George Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, 61391, Israel
| | - Anat Barnea
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, 43107, Israel
| |
Collapse
|
18
|
Spinello C, Laviola G, Macrì S. Pediatric Autoimmune Disorders Associated with Streptococcal Infections and Tourette's Syndrome in Preclinical Studies. Front Neurosci 2016; 10:310. [PMID: 27445678 PMCID: PMC4928151 DOI: 10.3389/fnins.2016.00310] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/20/2016] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence suggests that Tourette's Syndrome (TS) - a multifactorial pediatric disorder characterized by the recurrent exhibition of motor tics and/or vocal utterances - can partly depend on immune dysregulation provoked by early repeated streptococcal infections. The natural and adaptive antibody-mediated reaction to streptococcus has been proposed to potentially turn into a pathological autoimmune response in vulnerable individuals. Specifically, in conditions of increased permeability of the blood brain barrier (BBB), streptococcus-induced antibodies have been proposed to: (i) reach neuronal targets located in brain areas responsible for motion control; and (ii) contribute to the exhibition of symptoms. This theoretical framework is supported by indirect evidence indicating that a subset of TS patients exhibit elevated streptococcal antibody titers upon tic relapses. A systematic evaluation of this hypothesis entails preclinical studies providing a proof of concept of the aforementioned pathological sequelae. These studies shall rest upon individuals characterized by a vulnerable immune system, repeatedly exposed to streptococcus, and carefully screened for phenotypes isomorphic to the pathological signs of TS observed in patients. Preclinical animal models may thus constitute an informative, useful tool upon which conducting targeted, hypothesis-driven experiments. In the present review we discuss the available evidence in preclinical models in support of the link between TS and pediatric autoimmune neuropsychiatric disorders associated with streptococcus infections (PANDAS), and the existing gaps that future research shall bridge. Specifically, we report recent preclinical evidence indicating that the immune responses to repeated streptococcal immunizations relate to the occurrence of behavioral and neurological phenotypes reminiscent of TS. By the same token, we discuss the limitations of these studies: limited evidence of behavioral phenotypes isomorphic to tics and scarce knowledge about the immunological phenomena favoring the transition from natural adaptive immunity to pathological outcomes.
Collapse
Affiliation(s)
- Chiara Spinello
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| | - Giovanni Laviola
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| | - Simone Macrì
- Section of Behavioural Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità Roma, Italy
| |
Collapse
|
19
|
A leading role for NADPH oxidase in an in-vitro study of experimental autoimmune encephalomyelitis. Mol Immunol 2016; 72:19-27. [DOI: 10.1016/j.molimm.2016.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/09/2016] [Accepted: 02/12/2016] [Indexed: 01/24/2023]
|
20
|
Poon C, McMahon D, Hynynen K. Noninvasive and targeted delivery of therapeutics to the brain using focused ultrasound. Neuropharmacology 2016; 120:20-37. [PMID: 26907805 DOI: 10.1016/j.neuropharm.2016.02.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/13/2016] [Accepted: 02/15/2016] [Indexed: 11/24/2022]
Abstract
The range of therapeutic treatment options for central nervous system (CNS) diseases is greatly limited by the blood-brain barrier (BBB). While a variety of strategies to circumvent the blood-brain barrier for drug delivery have been investigated, little clinical success has been achieved. Focused ultrasound (FUS) is a unique approach whereby the transcranial application of acoustic energy to targeted brain areas causes a noninvasive, safe, transient, and targeted opening of the BBB, providing an avenue for the delivery of therapeutic agents from the systemic circulation into the brain. There is a great need for viable treatment strategies for CNS diseases, and we believe that the preclinical success of this technique should encourage a rapid movement towards clinical testing. In this review, we address the versatile applications of FUS-mediated BBB opening, the safety profile of the technique, and the physical and biological mechanisms that drive this process. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Charissa Poon
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Dallan McMahon
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kullervo Hynynen
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
21
|
Jain A, Balice-Gordon R. Cellular, synaptic, and circuit effects of antibodies in autoimmune CNS synaptopathies. HANDBOOK OF CLINICAL NEUROLOGY 2016; 133:77-93. [PMID: 27112672 DOI: 10.1016/b978-0-444-63432-0.00005-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Recently, clinicians have identified overlapping but distinguishable encephalitides, each associated with antibodies in serum and cerebrospinal fluid directed against specific cell surface proteins. The antibody targets identified to date are proteins that modulate cell physiology, synaptic transmission, and circuit function. Clinical and laboratory evidence suggests that the anti-cell surface antibodies are not simply markers of disease, but are pathogenic. Patient antibodies to N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), or gamma-aminobutyric acid-A (GABAA) receptors cause a loss of cognate receptors from synapses, while recent work has shown that antibodies to GABAB receptors directly antagonize receptor activity. Despite the distinct mechanisms by which patient antibodies abrogate the function of their targets, the resulting pathophysiology leads to abnormal circuit activity and plasticity, which manifests as patient signs and symptoms. Understanding the underlying synaptic and circuit mechanisms of patient autoantibody action may enable clinicians to develop diagnostics and therapies unique to each synaptic autoimmunity subtype, thereby improving patient identification and outcomes.
Collapse
Affiliation(s)
- Ankit Jain
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rita Balice-Gordon
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Neuroscience and Pain Research Unit, Pfizer, Inc., Cambridge, MA, USA.
| |
Collapse
|
22
|
Hoogland ICM, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflammation 2015; 12:114. [PMID: 26048578 PMCID: PMC4470063 DOI: 10.1186/s12974-015-0332-6] [Citation(s) in RCA: 659] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022] Open
Abstract
Background Animal studies show that peripheral inflammatory stimuli may activate microglial cells in the brain implicating an important role for microglia in sepsis-associated delirium. We systematically reviewed animal experiments related to the effects of systemic inflammation on the microglial and inflammatory response in the brain. Methods We searched PubMed between January 1, 1950 and December 1, 2013 and Embase between January 1, 1988 and December 1, 2013 for animal studies on the influence of peripheral inflammatory stimuli on microglia and the brain. Identified studies were systematically scored on methodological quality. Two investigators extracted independently data on animal species, gender, age, and genetic background; number of animals; infectious stimulus; microglial cells; and other inflammatory parameters in the brain, including methods, time points after inoculation, and brain regions. Results Fifty-one studies were identified of which the majority was performed in mice (n = 30) or in rats (n = 19). Lipopolysaccharide (LPS) (dose ranging between 0.33 and 200 mg/kg) was used as a peripheral infectious stimulus in 39 studies (76 %), and live or heat-killed pathogens were used in 12 studies (24 %). Information about animal characteristics such as species, strain, sex, age, and weight were defined in 41 studies (80 %), and complete methods of the disease model were described in 35 studies (68 %). Studies were also heterogeneous with respect to methods used to assess microglial activation; markers used mostly were the ionized calcium binding adaptor molecule-1 (Iba-1), cluster of differentiation 68 (CD68), and CD11b. After LPS challenge microglial activation was seen 6 h after challenge and remained present for at least 3 days. Live Escherichia coli resulted in microglial activation after 2 days, and heat-killed bacteria after 2 weeks. Concomitant with microglial response, inflammatory parameters in the brain were reviewed in 23 of 51 studies (45 %). Microglial activation was associated with an increase in Toll-like receptor (TLR-2 and TLR-4), tumor necrosis factor alpha (TNF-α), and interleukin 1 beta (IL-1β) messenger ribonucleic acid (mRNA) expression or protein levels. Interpretation Animal experiments robustly showed that peripheral inflammatory stimuli cause microglial activation. We observed distinct differences in microglial activation between systemic stimulation with (supranatural doses) LPS and live or heat-killed bacteria.
Collapse
Affiliation(s)
- Inge C M Hoogland
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Carin Houbolt
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | - Willem A van Gool
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Diederik van de Beek
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Muro S. Strategies for delivery of therapeutics into the central nervous system for treatment of lysosomal storage disorders. Drug Deliv Transl Res 2015; 2:169-86. [PMID: 24688886 DOI: 10.1007/s13346-012-0072-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lysosomal storage disorders (LSDs) are a group of about fifty life-threatening conditions caused by genetic defects affecting lysosomal components. The underscoring molecular deficiency leads to widespread cellular dysfunction through most tissues in the body, including peripheral organs and the central nervous system (CNS). Efforts during the last few decades have rendered a remarkable advance regarding our knowledge, medical awareness, and early detection of these genetic defects, as well as development of several treatment modalities. Clinical and experimental strategies encompassing enzyme replacement, gene and cell therapies, substrate reduction, and chemical chaperones are showing considerable potential in attenuating the peripheral pathology. However, a major drawback has been encountered regarding the suboptimal impact of these approaches on the CNS pathology. Particular anatomical and biochemical constraints of this tissue pose a major obstacle to the delivery of therapeutics into the CNS. Approaches to overcome these obstacles include modalities of local administration, strategies to enhance the blood-CNS permeability, intranasal delivery, use of exosomes, and those exploiting targeting of transporters and transcytosis pathways in the endothelial lining. The later two approaches are being pursued at the time by coupling therapeutic agents to affinity moieties and drug delivery systems capable of targeting these natural transport routes. This approach is particularly promising, as using paths naturally active at this interface may render safe and effective delivery of LSD therapies into the CNS.
Collapse
Affiliation(s)
- Silvia Muro
- Institute for Bioscience and Biotechnology Research University of Maryland, College Park, MD, 20742, USA ; Fischell Dept. of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| |
Collapse
|
24
|
Ayzenberg I, Schlevogt S, Metzdorf J, Stahlke S, Pedreitturia X, Hunfeld A, Couillard-Despres S, Kleiter I. Analysis of neurogenesis during experimental autoimmune encephalomyelitis reveals pitfalls of bioluminescence imaging. PLoS One 2015; 10:e0118550. [PMID: 25780928 PMCID: PMC4363373 DOI: 10.1371/journal.pone.0118550] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 01/20/2015] [Indexed: 12/11/2022] Open
Abstract
Bioluminescence imaging is a sensitive approach for longitudinal neuroimaging. Transgenic mice expressing luciferase under the promoter of doublecortin (DCX-luc), a specific marker of neuronal progenitor cells (NPC), allow monitoring of neurogenesis in living mice. Since the extent and time course of neurogenesis during autoimmune brain inflammation are controversial, we investigated neurogenesis in MOG-peptide induced experimental allergic encephalomyelitis (EAE) using DCX-luc reporter mice. We observed a marked, 2- to 4-fold increase of the bioluminescence signal intensity 10 days after EAE induction and a gradual decline 1–2 weeks thereafter. In contrast, immunostaining for DCX revealed no differences between EAE and control mice 2 and 4 weeks after immunization in zones of adult murine neurogenesis such as the dentate gyrus. Ex vivo bioluminescence imaging showed similar luciferase expression in brain homogenates of EAE and control animals. Apart from complete immunization including MOG-peptide also incomplete immunization with complete Freund´s adjuvant and pertussis toxin resulted in a rapid increase of the in vivo bioluminescence signal. Blood-brain barrier (BBB) leakage was demonstrated 10 days after both complete and incomplete immunization and might explain the increased bioluminescence signal in vivo. We conclude, that acute autoimmune inflammation in EAE does not alter neurogenesis, at least at the stage of DCX-expressing NPC. Effects of immunization on the BBB integrity must be considered when luciferase is used as a reporter within the CNS during the active stage of EAE. Models with stable CNS-restricted luciferase expression could serve as technically convenient way to evaluate BBB integrity in a longitudinal manner.
Collapse
Affiliation(s)
- Ilya Ayzenberg
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Sibylle Schlevogt
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Judith Metzdorf
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Sarah Stahlke
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | | | - Anika Hunfeld
- Department of Animal Physiology, Ruhr-University, Bochum, Germany
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Ingo Kleiter
- Department of Neurology, St. Josef-Hospital, Ruhr-University, Bochum, Germany
- * E-mail:
| |
Collapse
|
25
|
Kozai TDY, Jaquins-Gerstl AS, Vazquez AL, Michael AC, Cui XT. Brain tissue responses to neural implants impact signal sensitivity and intervention strategies. ACS Chem Neurosci 2015; 6:48-67. [PMID: 25546652 PMCID: PMC4304489 DOI: 10.1021/cn500256e] [Citation(s) in RCA: 387] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
![]()
Implantable biosensors are valuable
scientific tools for basic
neuroscience research and clinical applications. Neurotechnologies
provide direct readouts of neurological signal and neurochemical processes.
These tools are generally most valuable when performance capacities
extend over months and years to facilitate the study of memory, plasticity,
and behavior or to monitor patients’ conditions. These needs
have generated a variety of device designs from microelectrodes for
fast scan cyclic voltammetry (FSCV) and electrophysiology to microdialysis
probes for sampling and detecting various neurochemicals. Regardless
of the technology used, the breaching of the blood–brain barrier
(BBB) to insert devices triggers a cascade of biochemical pathways
resulting in complex molecular and cellular responses to implanted
devices. Molecular and cellular changes in the microenvironment surrounding
an implant include the introduction of mechanical strain, activation
of glial cells, loss of perfusion, secondary metabolic injury, and
neuronal degeneration. Changes to the tissue microenvironment surrounding
the device can dramatically impact electrochemical and electrophysiological
signal sensitivity and stability over time. This review summarizes
the magnitude, variability, and time course of the dynamic molecular
and cellular level neural tissue responses induced by state-of-the-art
implantable devices. Studies show that insertion injuries and foreign
body response can impact signal quality across all implanted central
nervous system (CNS) sensors to varying degrees over both acute (seconds
to minutes) and chronic periods (weeks to months). Understanding the
underlying biological processes behind the brain tissue response to
the devices at the cellular and molecular level leads to a variety
of intervention strategies for improving signal sensitivity and longevity.
Collapse
Affiliation(s)
- Takashi D. Y. Kozai
- Department
of Bioengineering, ‡Center for the Neural Basis of Cognition, §McGowan Institute
for Regenerative Medicine, ∥Department of Chemistry, and ⊥Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Andrea S. Jaquins-Gerstl
- Department
of Bioengineering, ‡Center for the Neural Basis of Cognition, §McGowan Institute
for Regenerative Medicine, ∥Department of Chemistry, and ⊥Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alberto L. Vazquez
- Department
of Bioengineering, ‡Center for the Neural Basis of Cognition, §McGowan Institute
for Regenerative Medicine, ∥Department of Chemistry, and ⊥Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Adrian C. Michael
- Department
of Bioengineering, ‡Center for the Neural Basis of Cognition, §McGowan Institute
for Regenerative Medicine, ∥Department of Chemistry, and ⊥Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - X. Tracy Cui
- Department
of Bioengineering, ‡Center for the Neural Basis of Cognition, §McGowan Institute
for Regenerative Medicine, ∥Department of Chemistry, and ⊥Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
26
|
IgG accumulates in inhibitory hippocampal neurons of experimental antiphospholipid syndrome. J Autoimmun 2014; 55:86-93. [DOI: 10.1016/j.jaut.2014.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 11/18/2022]
|
27
|
Kozai TDY, Li X, Bodily LM, Caparosa EM, Zenonos GA, Carlisle DL, Friedlander RM, Cui XT. Effects of caspase-1 knockout on chronic neural recording quality and longevity: insight into cellular and molecular mechanisms of the reactive tissue response. Biomaterials 2014; 35:9620-34. [PMID: 25176060 DOI: 10.1016/j.biomaterials.2014.08.006] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/01/2014] [Indexed: 12/17/2022]
Abstract
Chronic implantation of microelectrodes into the cortex has been shown to lead to inflammatory gliosis and neuronal loss in the microenvironment immediately surrounding the probe, a hypothesized cause of neural recording failure. Caspase-1 (aka Interleukin 1β converting enzyme) is known to play a key role in both inflammation and programmed cell death, particularly in stroke and neurodegenerative diseases. Caspase-1 knockout (KO) mice are resistant to apoptosis and these mice have preserved neurologic function by reducing ischemia-induced brain injury in stroke models. Local ischemic injury can occur following neural probe insertion and thus in this study we investigated the hypothesis that caspase-1 KO mice would have less ischemic injury surrounding the neural probe. In this study, caspase-1 KO mice were implanted with chronic single shank 3 mm Michigan probes into V1m cortex. Electrophysiology recording showed significantly improved single-unit recording performance (yield and signal to noise ratio) of caspase-1 KO mice compared to wild type C57B6 (WT) mice over the course of up to 6 months for the majority of the depth. The higher yield is supported by the improved neuronal survival in the caspase-1 KO mice. Impedance fluctuates over time but appears to be steadier in the caspase-1 KO especially at longer time points, suggesting milder glia scarring. These findings show that caspase-1 is a promising target for pharmacologic interventions.
Collapse
Affiliation(s)
- Takashi D Y Kozai
- Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, USA.
| | - Xia Li
- Bioengineering, University of Pittsburgh, USA
| | - Lance M Bodily
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, USA
| | - Ellen M Caparosa
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, USA
| | - Georgios A Zenonos
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, USA
| | - Diane L Carlisle
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, USA
| | - Robert M Friedlander
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, USA
| | - X Tracy Cui
- Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, USA.
| |
Collapse
|
28
|
Sagar D, Masih S, Schell T, Jacobson S, Comber JD, Philip R, Wigdahl B, Jain P, Khan ZK. In vivo immunogenicity of Tax(11-19) epitope in HLA-A2/DTR transgenic mice: implication for dendritic cell-based anti-HTLV-1 vaccine. Vaccine 2014; 32:3274-84. [PMID: 24739247 DOI: 10.1016/j.vaccine.2014.03.087] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/20/2014] [Accepted: 03/26/2014] [Indexed: 01/10/2023]
Abstract
Viral oncoprotein Tax plays key roles in transformation of human T-cell leukemia virus (HTLV-1)-infected T cells leading to adult T-cell leukemia (ATL), and is the key antigen recognized during HTLV-associated myelopathy (HAM). In HLA-A2+ asymptomatic carriers as well as ATL and HAM patients, Tax(11-19) epitope exhibits immunodominance. Here, we evaluate CD8 T-cell immune response against this epitope in the presence and absence of dendritic cells (DCs) given the recent encouraging observations made with Phase 1 DC-based vaccine trial for ATL. To facilitate these studies, we first generated an HLA-A2/DTR hybrid mouse strain carrying the HLA-A2.1 and CD11c-DTR genes. We then studied CD8 T-cell immune response against Tax(11-19) epitope delivered in the absence or presence of Freund's adjuvant and/or DCs. Overall results demonstrate that naturally presented Tax epitope could initiate an antigen-specific CD8T cell response in vivo but failed to do so upon DC depletion. Presence of adjuvant potentiated Tax(11-19)-specific response. Elevated serum IL-6 levels coincided with depletion of DCs whereas decreased TGF-β was associated with adjuvant use. Thus, Tax(11-19) epitope is a potential candidate for the DC-based anti-HTLV-1 vaccine and the newly hybrid mouse strain could be used for investigating DC involvement in human class-I-restricted immune responses.
Collapse
Affiliation(s)
- Divya Sagar
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Shet Masih
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Todd Schell
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Steven Jacobson
- Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Brian Wigdahl
- Department of Microbiology and Immunology, and the Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Zafar K Khan
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology & Virology Research, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
29
|
Maggio N, Shavit-Stein E, Dori A, Blatt I, Chapman J. Prolonged systemic inflammation persistently modifies synaptic plasticity in the hippocampus: modulation by the stress hormones. Front Mol Neurosci 2013; 6:46. [PMID: 24363642 PMCID: PMC3850065 DOI: 10.3389/fnmol.2013.00046] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/15/2013] [Indexed: 12/18/2022] Open
Abstract
Transient systemic inflammation has been shown to cause altered behavior both in humans and in laboratory animals through activation of microglia and heightened level of cytokines detected in the brain and in the body. Furthermore, both activated microglia and the increased cytokines level have been associated with the sudden clinical deterioration in demented people or in aged patients upon systemic inflammation. Whilst it is increasingly becoming clear the role of transient systemic inflammation in promoting dementia in aged individuals, it is still a matter of debate whether prolonged systemic inflammation might persistently modify the brain. In this study, we examined the influence of a systemic long term inflammatory event on synaptic plasticity. We report that while a short exposure to LPS produces transient deficit in long term potentiation (LTP) expression, systemic prolonged inflammation impairs LTP in slices of animals previously primed by a Complete Freund's adjuvant injection. Interestingly, steroids are able to modulate this effect: whereas glucocorticosteroid (GR) activation further reduces LTP, mineralocorticosteroid receptors (MR) activation promotes the full recovery of LTP. We believe that this research advances the current understandings on the role of the immune system in the onset and progression of cognitive deficits following long lasting systemic inflammation, and proposes possible insights on future strategies in order to prevent early dementia in these predisposed individuals.
Collapse
Affiliation(s)
- Nicola Maggio
- Department of Neurology, The Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center Tel HaShomer, Israel ; Talpiot Medical Leadership Program, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Amir Dori
- Department of Neurology, The Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center Tel HaShomer, Israel ; Talpiot Medical Leadership Program, The Chaim Sheba Medical Center Tel HaShomer, Israel ; Department of Neurology, Washington University School of Medicine Saint Louis, MO, USA
| | - Ilan Blatt
- Department of Neurology, The Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center Tel HaShomer, Israel ; Department of Neurology, Sackler Faculty of Medicine, Tel Aviv University Tel Aviv, Israel
| | - Joab Chapman
- Department of Neurology, The Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center Tel HaShomer, Israel ; Department of Neurology, Sackler Faculty of Medicine, Tel Aviv University Tel Aviv, Israel
| |
Collapse
|
30
|
Marshall SA, McClain JA, Kelso ML, Hopkins DM, Pauly JR, Nixon K. Microglial activation is not equivalent to neuroinflammation in alcohol-induced neurodegeneration: The importance of microglia phenotype. Neurobiol Dis 2013; 54:239-51. [PMID: 23313316 PMCID: PMC3629000 DOI: 10.1016/j.nbd.2012.12.016] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 12/21/2012] [Accepted: 12/28/2012] [Indexed: 12/19/2022] Open
Abstract
Excessive alcohol intake, a defining characteristic of an alcohol use disorder (AUD), results in neurodegeneration in the hippocampus and entorhinal cortex that has been linked to a variety of cognitive deficits. Neuroinflammation is thought to be a factor in alcohol-induced neurodegeneration, and microglia activation is a key but not sole component of an inflammatory response. These experiments investigate the effects of ethanol exposure in a well-accepted model of an AUD on both microglial activation and blood brain barrier disruption (BBB) in order to understand their relationship to classical definitions of inflammation and alcohol-induced neurodegeneration. Following a four-day binge ethanol paradigm, rat hippocampal and entorhinal cortex tissue was examined using three distinct approaches to determine microglia phenotype and BBB disruption: immunohistochemistry, autoradiography, and ELISA. After ethanol exposure, there was an increase in [(3)H]-PK-11195 binding and OX-42 immunoreactivity indicative of microglial activation; however, microglia were not fully activated since both OX-6 and ED-1 immunoreactive microglia were absent. This data was supported by functional evidence as there was no increase in the proinflammatory cytokines IL-6 or TNF-α, but a 26% increase in the anti-inflammatory cytokine, IL-10, and a 38% increase in the growth factor, TGF-β, seven days after exposure. Furthermore, there was no evidence of a disruption of the BBB. These data suggest that the four-day binge model of an AUD, which produces neurodegeneration in corticolimbic regions, does not elicit classical neuroinflammation but instead produces partially activated microglia. Partial activation of microglia following binge ethanol exposure suggest that microglia in this model have beneficial or homeostatic roles rather than directly contributing to neurodegeneration and are a consequence of alcohol-induced-damage instead of the source of damage.
Collapse
Affiliation(s)
- S. Alex Marshall
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536-0596
| | - Justin A. McClain
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536-0596
| | | | - Deann M. Hopkins
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536-0596
| | - James R. Pauly
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536-0596
| | - Kimberly Nixon
- Department of Pharmaceutical Sciences, The University of Kentucky, Lexington, KY 40536-0596
| |
Collapse
|
31
|
Lutz NW, Fernandez C, Pellissier JF, Cozzone PJ, Béraud E. Cerebral biochemical pathways in experimental autoimmune encephalomyelitis and adjuvant arthritis: a comparative metabolomic study. PLoS One 2013; 8:e56101. [PMID: 23457507 PMCID: PMC3573043 DOI: 10.1371/journal.pone.0056101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 01/09/2013] [Indexed: 01/17/2023] Open
Abstract
Many diseases, including brain disorders, are associated with perturbations of tissue metabolism. However, an often overlooked issue is the impact that inflammations outside the brain may have on brain metabolism. Our main goal was to study similarities and differences between brain metabolite profiles of animals suffering from experimental autoimmune encephalomyelitis (EAE) and adjuvant arthritis (AA) in Lewis rat models. Our principal objective was the determination of molecular protagonists involved in the metabolism underlying these diseases. EAE was induced by intraplantar injection of complete Freund’s adjuvant (CFA) and spinal-cord homogenate (SC-H), whereas AA was induced by CFA only. Naive rats served as controls (n = 9 for each group). Two weeks after inoculation, animals were sacrificed, and brains were removed and processed for metabolomic analysis by NMR spectroscopy or for immunohistochemistry. Interestingly, both inflammatory diseases caused similar, though not identical, changes in metabolites involved in regulation of brain cell size and membrane production: among the osmolytes, taurine and the neuronal marker, N-acetylaspartate, were decreased, and the astrocyte marker, myo-inositol, slightly increased in both inoculated groups compared with controls. Also ethanolamine-containing phospholipids, sources of inflammatory agents, and several glycolytic metabolites were increased in both inoculated groups. By contrast, the amino acids, aspartate and isoleucine, were less concentrated in CFA/SC-H and control vs. CFA rats. Our results suggest that inflammatory brain metabolite profiles may indicate the existence of either cerebral (EAE) or extra-cerebral (AA) inflammation. These inflammatory processes may act through distinct pathways that converge toward similar brain metabolic profiles. Our findings open new avenues for future studies aimed at demonstrating whether brain metabolic effects provoked by AA are pain/stress-mediated and/or due to the presence of systemic proinflammatory molecules. Regardless of the nature of these mechanisms, our findings may be of interest for future clinical studies, e.g. by in-vivo magnetic resonance spectroscopy.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Brain/metabolism
- Brain/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Freund's Adjuvant
- Metabolic Networks and Pathways
- Metabolomics
- Phospholipids/metabolism
- Rats
- Rats, Inbred Lew
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Water/metabolism
Collapse
Affiliation(s)
- Norbert W Lutz
- Center for Biological and Medical Magnetic Resonance CRMBM, National Center for Scientific Research Joint Research Unit 7339, Aix-Marseille University, Marseille, France.
| | | | | | | | | |
Collapse
|
32
|
Immunoglobulin-Mediated Neuro-Cognitive Impairment: New Data and a Comprehensive Review. Clin Rev Allergy Immunol 2013; 45:248-55. [DOI: 10.1007/s12016-013-8357-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Ahn M, Yang W, Kim H, Jin JK, Moon C, Shin T. Immunohistochemical study of arginase-1 in the spinal cords of Lewis rats with experimental autoimmune encephalomyelitis. Brain Res 2012; 1453:77-86. [PMID: 22483960 DOI: 10.1016/j.brainres.2012.03.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 02/11/2012] [Accepted: 03/09/2012] [Indexed: 11/30/2022]
Abstract
Arginase-1, a marker for M2 phenotype alternatively activated macrophages, inhibits inflammation and is associated with phagocytosis of cell debris and apoptotic cells. We analyzed the expression of arginase-1, a competitive enzyme of inducible nitric oxide synthase (iNOS), in the spinal cords of Lewis rats with experimental autoimmune encephalomyelitis (EAE). Western blot analysis showed that both arginase-1 and iNOS significantly increased in the spinal cords of rats at the peak stage of EAE compared with the expression level in control animals (p<0.05) and declined thereafter. Immunofluorescent staining demonstrated that increased expression of arginase-1 in EAE spinal cords was confirmed in macrophages as well as in some neurons and astrocytes that were constitutively positive for arginase-1 in normal spinal cords. A semiquantitative analysis by immunofluorescence showed that in EAE lesions, an increased level of arginase-1 immunoreactivity was matched with ED1-positive macrophages, which were also positive for activin A, a marker for the M2 phenotype. Taking all of these findings into consideration, we postulate that the increased level of arginase-1, which is partly from M2 macrophages, contributes to the modulation of neuroinflammation in EAE lesions, possibly through the reduction of nitric oxide in the lesion via competition with iNOS for the use of L-arginine.
Collapse
Affiliation(s)
- Meejung Ahn
- Department of Anatomy, School of Medicine, Jeju National University, Jeju 690-756, South Korea
| | | | | | | | | | | |
Collapse
|
34
|
Simultaneous analysis of plasma and CSF by NMR and hierarchical models fusion. Anal Bioanal Chem 2012; 403:947-59. [PMID: 22395451 PMCID: PMC3336062 DOI: 10.1007/s00216-012-5871-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 01/28/2012] [Accepted: 02/14/2012] [Indexed: 12/31/2022]
Abstract
Because cerebrospinal fluid (CSF) is the biofluid which interacts most closely with the central nervous system, it holds promise as a reporter of neurological disease, for example multiple sclerosis (MScl). To characterize the metabolomics profile of neuroinflammatory aspects of this disease we studied an animal model of MScl—experimental autoimmune/allergic encephalomyelitis (EAE). Because CSF also exchanges metabolites with blood via the blood–brain barrier, malfunctions occurring in the CNS may be reflected in the biochemical composition of blood plasma. The combination of blood plasma and CSF provides more complete information about the disease. Both biofluids can be studied by use of NMR spectroscopy. It is then necessary to perform combined analysis of the two different datasets. Mid-level data fusion was therefore applied to blood plasma and CSF datasets. First, relevant information was extracted from each biofluid dataset by use of linear support vector machine recursive feature elimination. The selected variables from each dataset were concatenated for joint analysis by partial least squares discriminant analysis (PLS-DA). The combined metabolomics information from plasma and CSF enables more efficient and reliable discrimination of the onset of EAE. Second, we introduced hierarchical models fusion, in which previously developed PLS-DA models are hierarchically combined. We show that this approach enables neuroinflamed rats (even on the day of onset) to be distinguished from either healthy or peripherally inflamed rats. Moreover, progression of EAE can be investigated because the model separates the onset and peak of the disease. Graphical representation of Hierarchical Models Fusion applied to concatenated plasma and CSF datasets. ![]()
Collapse
|
35
|
Ko MK, Saraswathy S, Parikh JG, Rao NA. The role of TLR4 activation in photoreceptor mitochondrial oxidative stress. Invest Ophthalmol Vis Sci 2011; 52:5824-35. [PMID: 21666244 DOI: 10.1167/iovs.10-6357] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Herein the authors investigated whether the activation of Toll-like receptors (TLRs) in the innate immune response causes retinal photoreceptor oxidative stress and mitochondrial DNA (mtDNA) damage. METHODS On day 5 after injection of complete Freund's adjuvant containing heat-killed Mycobacterium tuberculosis (CFA), retinas were submitted to polymerase chain reaction (PCR) array focused on the TLR signaling, or apoptosis, pathway. CFA-mediated TLR4 activation, oxidative stress, and mtDNA damage were determined in B10.RIII and knockout (KO) mice (recombination activation gene [Rag] 1(KO), TLR4(KO), myeloid differentiation primary response gene 88 [MyD88](KO), tumor necrosis factor [TNF]-α(KO), or caspase 7(KO) mice) using quantitative real-time PCR, enzyme-linked immunosorbent assay, Western blot analysis, and immunohistochemistry. The mycobacterial DNA load on the retina, brain, liver, and spleen was determined by real-time PCR after intracardiac perfusion. RESULTS PCR array demonstrated the upregulation of TLRs and their signaling molecules in retinas of CFA-injected mice compared with those of control animals without inflammatory cell infiltration in the retina and uvea. Mycobacterial DNA was detected in the retinas of CFA-injected mice. Retinas of CFA-injected animals showed oxidative stress and mtDNA damage, primarily in the photoreceptor inner segments. Upregulated TLR4 was localized with CD11b(+)MHCII(+) cells but not with GFAP(+) astrocytes. This oxidative stress/damage was similar in CFA-injected Rag1(KO) mice compared with wild-type controls. Such damage was absent in the retinas of CFA-injected TLR4(KO), MyD88(KO), and TNF-α(KO) mice. CFA-mediated inducible nitric oxide synthase expression in the retina was significantly decreased in TNF-α(KO) mice. CONCLUSIONS Retinal photoreceptors are susceptible to mitochondrial oxidative stress/mtDNA damage in robust TLR4-mediated innate immune response.
Collapse
Affiliation(s)
- MinHee K Ko
- Doheny Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | |
Collapse
|
36
|
Blanchet L, Smolinska A, Attali A, Stoop MP, Ampt KAM, van Aken H, Suidgeest E, Tuinstra T, Wijmenga SS, Luider T, Buydens LMC. Fusion of metabolomics and proteomics data for biomarkers discovery: case study on the experimental autoimmune encephalomyelitis. BMC Bioinformatics 2011; 12:254. [PMID: 21696593 PMCID: PMC3225201 DOI: 10.1186/1471-2105-12-254] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 06/22/2011] [Indexed: 01/22/2023] Open
Abstract
Background Analysis of Cerebrospinal Fluid (CSF) samples holds great promise to diagnose neurological pathologies and gain insight into the molecular background of these pathologies. Proteomics and metabolomics methods provide invaluable information on the biomolecular content of CSF and thereby on the possible status of the central nervous system, including neurological pathologies. The combined information provides a more complete description of CSF content. Extracting the full combined information requires a combined analysis of different datasets i.e. fusion of the data. Results A novel fusion method is presented and applied to proteomics and metabolomics data from a pre-clinical model of multiple sclerosis: an Experimental Autoimmune Encephalomyelitis (EAE) model in rats. The method follows a mid-level fusion architecture. The relevant information is extracted per platform using extended canonical variates analysis. The results are subsequently merged in order to be analyzed jointly. We find that the combined proteome and metabolome data allow for the efficient and reliable discrimination between healthy, peripherally inflamed rats, and rats at the onset of the EAE. The predicted accuracy reaches 89% on a test set. The important variables (metabolites and proteins) in this model are known to be linked to EAE and/or multiple sclerosis. Conclusions Fusion of proteomics and metabolomics data is possible. The main issues of high-dimensionality and missing values are overcome. The outcome leads to higher accuracy in prediction and more exhaustive description of the disease profile. The biological interpretation of the involved variables validates our fusion approach.
Collapse
Affiliation(s)
- Lionel Blanchet
- Radboud University Nijmegen, Institute for Molecules and Materials, Heyendaalseweg, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Armentero MT, Levandis G, Bazzini E, Cerri S, Ghezzi C, Blandini F. Adhesion molecules as potential targets for neuroprotection in a rodent model of Parkinson's disease. Neurobiol Dis 2011; 43:663-8. [PMID: 21684338 DOI: 10.1016/j.nbd.2011.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 04/28/2011] [Accepted: 05/28/2011] [Indexed: 01/10/2023] Open
Abstract
Cell adhesion molecules might play an important role in the inflammatory mechanisms associated with neurodegeneration. We have previously observed, in rats, that subcutaneous injection of complete Freund's adjuvant (CFA), a pro-inflammatory agent that induces a peripheral inflammatory stimulus, reduces the nigrostriatal degeneration and microglial activation caused by stereotaxic injection of 6-hydroxydopamine (6-OHDA). Here we further investigated the effects of CFA in 6-OHDA-lesioned rats by evaluating the expression of selected adhesion molecules, both at central and peripheral levels. Male, Sprague-Dawley rats received a subcutaneous injection of CFA followed, 10 days later, by intrastriatal injection of 6-OHDA. Animals were sacrificed at various time points and changes affecting intercellular (ICAM-1), vascular (VCAM-1), platelet endothelial (PECAM-1) and neural (NCAM-1) cell adhesion molecules were analyzed in striatum, ventral midbrain (containing the substantia nigra) and sera. Our results confirmed the protective effect of systemic CFA on 6-OHDA-induced nigrostriatal degeneration. Injection of 6-OHDA increased striatal ICAM-1 and PECAM-1 expression, while opposite changes (decreased expression) were detected in the ventral midbrain, particularly for VCAM-1 and NCAM-1. Pretreatment with CFA counteracted these changes. Nigrostriatal degeneration also affected peripheral immune function, with lesioned animals showing increased sPECAM levels with respect to intact animals. Also in this case, CFA pretreatment blocked the 6-OHDA induced increase of sPECAM. Our findings confirm that a pre-existing, peripheral pro-inflammatory condition reduces the neuroinflammatory response and associated neurodegeneration provoked by centrally-administered 6-OHDA, with a mechanism that seems to involve selected adhesion molecules. The link between peripheral and central immune responses may, therefore, represent a target for new therapeutic strategies aimed at reducing the neuroinflammatory component associated with neurodegeneration.
Collapse
Affiliation(s)
- Marie-Therese Armentero
- Laboratory of Functional Neurochemistry, Interdepartmental Research Center for Parkinson's Disease, CRIMP, IRCCS National Institute of Neurology C Mondino, Pavia, Italy.
| | | | | | | | | | | |
Collapse
|
38
|
Strategy for effective brain drug delivery. Eur J Pharm Sci 2010; 40:385-403. [DOI: 10.1016/j.ejps.2010.05.003] [Citation(s) in RCA: 257] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 04/15/2010] [Accepted: 05/10/2010] [Indexed: 12/20/2022]
|
39
|
Moscato EH, Jain A, Peng X, Hughes EG, Dalmau J, Balice-Gordon RJ. Mechanisms underlying autoimmune synaptic encephalitis leading to disorders of memory, behavior and cognition: insights from molecular, cellular and synaptic studies. Eur J Neurosci 2010; 32:298-309. [PMID: 20646055 PMCID: PMC2955837 DOI: 10.1111/j.1460-9568.2010.07349.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recently, several novel, potentially lethal and treatment-responsive syndromes that affect hippocampal and cortical function have been shown to be associated with auto-antibodies against synaptic antigens, notably glutamate or GABA-B receptors. Patients with these auto-antibodies, sometimes associated with teratomas and other neoplasms, present with psychiatric symptoms, seizures, memory deficits and decreased levels of consciousness. These symptoms often improve dramatically after immunotherapy or tumor resection. Here we review studies of the cellular and synaptic effects of these antibodies in hippocampal neurons in vitro and preliminary work in rodent models. Our work suggests that patient antibodies lead to rapid and reversible removal of neurotransmitter receptors from synaptic sites, leading to changes in synaptic and circuit function that in turn are likely to lead to behavioral deficits. We also discuss several of the many questions raised by these and related disorders. Determining the mechanisms underlying these novel anti-neurotransmitter receptor encephalopathies will provide insights into the cellular and synaptic bases of the memory and cognitive deficits that are hallmarks of these disorders, and potentially suggest avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Emilia H Moscato
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6074, USA
| | | | | | | | | | | |
Collapse
|
40
|
Pasha S, Gupta K. Various drug delivery approaches to the central nervous system. Expert Opin Drug Deliv 2010; 7:113-35. [PMID: 20017662 DOI: 10.1517/17425240903405581] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IMPORTANCE OF THE FIELD The presence of the blood-brain barrier (BBB), an insurmountable obstacle, in particular, and other barriers in brain and periphery contribute to hindrance of the successful diagnosis and treatment of a myriad of central nervous system pathologies. This review discusses several strategies adopted to define a rational drug delivery approach to the CNS along with a short description of the strategies implemented by the authors' group to enhance the analgesic activity, a CNS property, of chimeric peptide of Met-enkephalin and FMRFa (YGGFMKKKFMRFa-YFa). AREAS COVERED IN THIS REVIEW Various approaches for drug delivery to the CNS with their beneficial and non-beneficial aspects, supported by an extensive literature survey published recently, up to August 2009. WHAT THE READER WILL GAIN The reader will have the privilege of gaining an understanding of previous as well as recent approaches to breaching the CNS barriers. TAKE HOME MESSAGE Among the various strategies discussed, the potential for efficacious CNS drug targeting in future lies either with the non-invasively administered multifunctional nanosystems or these nanosystems without characterstics such as long systemic circulating capability and avoiding reticuloendothelial system scavenging system of the body, endogenous transporters and efflux inhibitors administered by convection-enhanced delivery.
Collapse
Affiliation(s)
- Santosh Pasha
- Institute of Genomics and Integrative Biology, Peptide Synthesis Laboratory, Mall Road, Delhi-110007, India.
| | | |
Collapse
|
41
|
Celia C, Cosco D, Paolino D, Fresta M. Nanoparticulate devices for brain drug delivery. Med Res Rev 2010; 31:716-56. [DOI: 10.1002/med.20201] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
42
|
Pardridge WM. Alzheimer's disease drug development and the problem of the blood-brain barrier. Alzheimers Dement 2009; 5:427-32. [PMID: 19751922 DOI: 10.1016/j.jalz.2009.06.003] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 06/26/2009] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) drug development is limited by the presence of the blood-brain barrier (BBB). More than 98% of all small-molecule drugs, and approximately 100% of all large-molecule drugs, do not cross the BBB. Although the vast majority of AD drug candidates do not cross the BBB, the present-day AD drug-development effort is characterized by an imbalance wherein >99% of the drug-development effort is devoted to central nervous system (CNS) drug discovery, and <1% of drug development is devoted to CNS drug delivery. Future AD drug development needs a concerted effort to incorporate BBB sciences early in the CNS drug discovery process. This goal can be achieved by a reallocation of resources, and an expansion of research efforts in the pure science of BBB biology and the applied science of brain drug-targeting technology.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
43
|
Hsuchou H, Pan W, Wu X, Kastin AJ. Cessation of blood-to-brain influx of interleukin-15 during development of EAE. J Cereb Blood Flow Metab 2009; 29:1568-78. [PMID: 19536071 PMCID: PMC3599788 DOI: 10.1038/jcbfm.2009.79] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Regulatory changes in cytokine permeation across the blood-brain barrier (BBB) may have crucial roles in central nervous system (CNS) autoimmune disease. Accordingly, we examined the interactions of interleukin (IL)-15 with the cerebral vasculature after induction of experimental autoimmune encephalomyelitis (EAE). In contrast to the influx of (125)I-IL15 from blood to the CNS in normal mice and the persistence of IL15 influx in the spinal cord of EAE mice, influx was reduced in the EAE brain. Analyses of disappearance kinetics, FITC (fluorescein isothiocyanate)-albumin space, and delivery of IL15 by in situ perfusion, all indicate that the changes were not caused by BBB disruption but by the rapid availability (high volume of distribution) of IL15 and albumin. Although there was no significant change in the BBB permeation of IL15 in either direction in EAE mice, there was an upregulation of its specific receptor, IL15Ralpha, and an increased in situ production of IL15 mRNA that showed regional variation in both basal and EAE states. Overall, for IL15, its increased cerebral vascular space in the brain was equally as important as its persistent influx across the blood-spinal cord barrier, indicating that it is fully capable of activating the upregulated IL15Ralpha in the brain along with the intrinsic CNS source of IL15 in EAE mice.
Collapse
Affiliation(s)
- Hung Hsuchou
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | | | | | | |
Collapse
|
44
|
Streit WJ, Xue QS. Life and death of microglia. J Neuroimmune Pharmacol 2009; 4:371-9. [PMID: 19680817 DOI: 10.1007/s11481-009-9163-5] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Accepted: 06/30/2009] [Indexed: 01/05/2023]
Abstract
The importance of microglial cells in the maintenance of a well-functioning central nervous system (CNS) cannot be overstated. As descendants of the myelomonocytic lineage they are industrious housekeepers and watchful sentries that safeguard a homeostatic environment through a number of mechanisms designed to provide protection of fastidious neurons at all times. Microglia become particularly active after homeostasis has been perturbed by physical injury or other insults and they enter into a state of activation which is determined largely by the nature and severity of the lesion. Microglial activation is the main cellular event in acute neuroinflammation and essential for wound healing in the CNS. Recent studies from this laboratory have been focused on microglia in the aging brain and identified structural abnormalities, termed microglial dystrophy, that are consistent with cell senescence and progress to a form of accidental cell death that is marked by cytoplasmic degeneration and has been termed cytorrhexis. Cytorrhexis of microglia is infrequent in the normally aged human brain and non-detectable in aged rodents, but its occurrence increases dramatically during neurodegenerative conditions, including Alzheimer's disease (AD) in humans and motoneuron disease in transgenic rats. The identification of degenerating microglia has given rise to a novel theory of AD pathogenesis, the microglial dysfunction hypothesis, which views the loss of microglial neuroprotection as a central event in neurodegenerative disease development.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| | | |
Collapse
|
45
|
Decreased inflammation and augmented expression of trophic factors correlate with MOG-induced neuroprotection of the injured nigrostriatal system in the murine MPTP model of Parkinson's disease. Int Immunopharmacol 2009; 9:781-91. [PMID: 19286483 DOI: 10.1016/j.intimp.2009.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 02/06/2009] [Accepted: 03/02/2009] [Indexed: 11/20/2022]
Abstract
The response of the immune system during injury of the central nervous system may play a role in protecting neurons. We have previously reported that immunization with MOG 35-55 prior to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced injury of the dopaminergic system promotes less dopamine depletion and less dopaminergic damage of neurons in mice. In this study, we evaluate the influence of MOG immunization on the inflammatory reaction that occurs at the place of injury. C57Bl male mice, 2 and 12 months old, received i.p. injections of MPTP (40 mg/kg) and some groups animals also received an additional injection with myelin oligodendrocyte glycoprotein (MOG) 35-55 in CFA 6 days before MPTP administration. MPTP caused a common inflammatory reaction characterized by microglial activation, infiltration of T cells into the substantia nigra and striatum and increased expression of mRNA encoding pro-inflammatory cytokines (IL-1 beta, TNFalpha, INF gamma) and trophic factors (TGFbeta, GDNF). MOG immunization prior to MPTP administration significantly diminished the microglial reaction and reduced the levels of infiltrating CD8+ lymphocytes. The number of CD4+ T cells remained at the same level as in the MPTP group. Expression of pro-inflammatory cytokines was diminished. The mRNA expression of GDNF was significantly higher in the MOG pretreated mice relative to the MPTP group, both in the 2 month old and 12 month old groups. Since MOG immunization prior to MPTP intoxication appears to prevent nigrostriatal injury, the observed decrease of inflammation and increase of GDNF mRNA expression in the injured areas might represent one of the mechanisms of observed neuroprotection.
Collapse
|
46
|
Lavrnja I, Bjelobaba I, Stojiljkovic M, Pekovic S, Mostarica-Stojkovic M, Stosic-Grujicic S, Nedeljkovic N. Time-course changes in ectonucleotidase activities during experimental autoimmune encephalomyelitis. Neurochem Int 2009; 55:193-8. [PMID: 19524108 DOI: 10.1016/j.neuint.2009.02.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 02/24/2009] [Accepted: 02/25/2009] [Indexed: 12/20/2022]
Abstract
The aim of the present study was to analyze the activities of extracellular purine metabolizing enzymes, CD39 (apyrase, EC 3.6.1.5) and CD73 (ecto-5' nucleotidase, EC 3.1.3.5) in experimental autoimmune encephalomyelitis (EAE). The levels of ATP, ADP and AMP hydrolysis were analyzed in the blood serum and in the rat spinal cord plasma membrane preparation 8, 15 and 25 days after induction of EAE. The animals were divided in three groups: control (saline), CFA (adjuvant-only) and EAE (CFA and homogenate of spinal cords). Eight days after immunization, ATP, ADP and AMP hydrolysis in the blood serum and spinal cord membrane preparations were unaffected in EAE compared to both, control and CFA group. In the peak of disease, ATP, ADP and AMP hydrolysis in EAE group showed significant decrease in the blood serum and prominent increase in the spinal cord membrane preparation compared to CFA and control group. At the end of illness, as judged by disappearance of clinical manifestation of EAE, ATP, ADP and AMP hydrolysis, although closer to CFA levels, were still significantly different in respect to the CFA group. Modulation of ATP, ADP and AMP hydrolysis suggests that they operate during EAE and might represent the basis of novel therapeutic strategies in immune-mediated diseases, such as MS.
Collapse
Affiliation(s)
- Irena Lavrnja
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic" University of Belgrade, Bulevar Despota Stevana 142, 11000 Belgrade, Serbia.
| | | | | | | | | | | | | |
Collapse
|
47
|
Delivery of peptide and protein drugs over the blood-brain barrier. Prog Neurobiol 2009; 87:212-51. [PMID: 19395337 DOI: 10.1016/j.pneurobio.2008.12.002] [Citation(s) in RCA: 184] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 11/11/2008] [Accepted: 12/17/2008] [Indexed: 12/12/2022]
Abstract
Peptide and protein (P/P) drugs have been identified as showing great promises for the treatment of various neurodegenerative diseases. A major challenge in this regard, however, is the delivery of P/P drugs over the blood-brain barrier (BBB). Intense research over the last 25 years has enabled a better understanding of the cellular and molecular transport mechanisms at the BBB, and several strategies for enhanced P/P drug delivery over the BBB have been developed and tested in preclinical and clinical-experimental research. Among them, technology-based approaches (comprising functionalized nanocarriers and liposomes) and pharmacological strategies (such as the use of carrier systems and chimeric peptide technology) appear to be the most promising ones. This review combines a comprehensive overview on the current understanding of the transport mechanisms at the BBB with promising selected strategies published so far that can be applied to facilitate enhanced P/P drug delivery over the BBB.
Collapse
|
48
|
Mitchell K, Yang HYT, Berk JD, Tran JH, Iadarola MJ. Monocyte chemoattractant protein-1 in the choroid plexus: a potential link between vascular pro-inflammatory mediators and the CNS during peripheral tissue inflammation. Neuroscience 2009; 158:885-95. [PMID: 19032979 PMCID: PMC2668531 DOI: 10.1016/j.neuroscience.2008.10.047] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 10/17/2008] [Accepted: 10/28/2008] [Indexed: 12/22/2022]
Abstract
During peripheral tissue inflammation, inflammatory processes in the CNS can be initiated by blood-borne pro-inflammatory mediators. The choroid plexus, the site of cerebrospinal fluid (CSF) production, is a highly specialized interface between the vascular system and CNS, and thus, this structure may be an important element in communication between the vascular compartment and the CNS during peripheral tissue inflammation. We investigated the potential participation of the choroid plexus in this process during peripheral tissue inflammation by examining expression of the small inducible cytokine A2 (SCYA2) gene which codes for monocyte chemoattractant protein-1 (MCP-1). MCP-1 protein was previously reported to be induced in a variety of cells during peripheral tissue inflammation. In the basal state, SCYA2 is highly expressed in the choroid plexus as compared with other rat CNS tissues. During hind paw inflammation, SCYA2 expression was significantly elevated in choroid plexus, whereas it remained unchanged in a variety of brain regions. The SCYA2-expressing cells were strongly associated with the choroid plexus as vascular depletion of blood cells by whole-body saline flush did not significantly alter SCYA2 expression in the choroid plexus. In situ hybridization suggested that the SCYA2-expressing cells were localized to the choroid plexus stroma. To elucidate potential molecular mechanisms of SCYA2 increase, we examined genes in the nuclear factor-kappa B (NF-kappaB) signaling cascade including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta) and inhibitor of kappa B alpha (IkappaBalpha) in choroid tissue. Given that we also detected increased levels of MCP-1 protein by ELISA, we sought to identify potential downstream targets of MCP-1 and observed altered expression levels of mRNAs encoding tight junction proteins TJP2 and claudin 5. Finally, we detected a substantial up-regulation of the transcript encoding endothelial leukocyte adhesion molecule 1 (E-selectin), a molecule which could participate in leukocyte recruitment to the choroid plexus along with MCP-1. Together, these results suggest that profound changes occur in the choroid plexus during peripheral tissue inflammation, likely initiated by blood-borne inflammatory mediators, which may modify events in CNS.
Collapse
Affiliation(s)
- K. Mitchell
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - H.-Y. T. Yang
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - J. D. Berk
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - J. H. Tran
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| | - M. J. Iadarola
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Building 49, Room 1C20, 49 Convent Drive, MSC 4410, Bethesda, MD 20892-4410, USA
| |
Collapse
|
49
|
Vykhodtseva N, McDannold N, Hynynen K. Progress and problems in the application of focused ultrasound for blood-brain barrier disruption. ULTRASONICS 2008; 48:279-96. [PMID: 18511095 PMCID: PMC2569868 DOI: 10.1016/j.ultras.2008.04.004] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 03/25/2008] [Accepted: 04/06/2008] [Indexed: 05/03/2023]
Abstract
Advances in neuroscience have resulted in the development of new diagnostic and therapeutic agents for potential use in the central nervous system (CNS). However, the ability to deliver the majority of these agents to the brain is limited by the blood-brain barrier (BBB), a specialized structure of the blood vessel wall that hampers transport and diffusion from the blood to the brain. Many CNS disorders could be treated with drugs, enzymes, genes, or large-molecule biotechnological products such as recombinant proteins, if they could cross the BBB. This article reviews the problems of the BBB presence in treating the vast majority of CNS diseases and the efforts to circumvent the BBB through the design of new drugs and the development of more sophisticated delivery methods. Recent advances in the development of noninvasive, targeted drug delivery by MRI-guided ultrasound-induced BBB disruption are also summarized.
Collapse
Affiliation(s)
- Natalia Vykhodtseva
- Department of Radiology, Focused Ultrasound Laboratory, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Room 515, 75 Francis Street, Boston, MA 02115, USA.
| | | | | |
Collapse
|
50
|
Mitchell K, Yang HYT, Tessier PA, Muhly WT, Swaim WD, Szalayova I, Keller JM, Mezey E, Iadarola MJ. Localization of S100A8 and S100A9 expressing neutrophils to spinal cord during peripheral tissue inflammation. Pain 2007; 134:216-31. [PMID: 18063312 DOI: 10.1016/j.pain.2007.10.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 09/24/2007] [Accepted: 10/22/2007] [Indexed: 11/28/2022]
Abstract
Investigation of hyperalgesia at the spinal transcriptome level indicated that carrageenan-induced inflammation of rat hind paws leads to a rapid but sustained increase in S100A8 and S100A9 expression, two genes implicated in the pathology of numerous inflammatory diseases including rheumatoid arthritis and gout. In situ hybridization revealed that the elevation occurred in neutrophils that migrate to the spinal cord vasculature during peripheral inflammation, not in spinal neurons or glial cells. Immunohistochemical analysis suggests, but does not prove, that these neutrophils abundantly release S100A8 and S100A9. Consistent with this, we detected an increase in ICAM and VCAM, both indicators of endothelial activation, a known trigger for secretion of S100A8 and S100A9. Migration of S100A8- and S100A9-expressing neutrophils to spinal cord is selective, since MCP-1- and CD68-expressing leukocytes do not increase in spinal cord vasculature during hind paw inflammation. Examination of many neutrophil granule mediators in spinal cord indicated that they are not regulated to the same degree as S100A8 and S100A9. Neutrophil migration also occurs in the vasculature of brain and pituitary gland during peripheral inflammation. Together, these findings suggest an interaction between a subpopulation of leukocytes and the CNS during peripheral tissue inflammation, as implied by an apparent release and possible diffusion of S100A8 and S100A9 through the endothelial blood-brain barrier. Although the present findings do not establish the neurophysiological or behavioral relevance of these observations to nociceptive processing, the data raise the possibility that selective populations of leukocytes may communicate the presence of disease or tissue damage from the periphery to cells in the central nervous system.
Collapse
Affiliation(s)
- Kendall Mitchell
- Neurobiology and Pain Therapeutics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, 49 Convent Drive, Bethesda, MD 20892, United States
| | | | | | | | | | | | | | | | | |
Collapse
|