1
|
Wei D, Zhang N, Qu S, Wang H, Li J. Advances in nanotechnology for the treatment of GBM. Front Neurosci 2023; 17:1180943. [PMID: 37214394 PMCID: PMC10196029 DOI: 10.3389/fnins.2023.1180943] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/05/2023] [Indexed: 05/24/2023] Open
Abstract
Glioblastoma (GBM), a highly malignant glioma of the central nervous system, is the most dread and common brain tumor with a high rate of therapeutic resistance and recurrence. Currently, the clinical treatment methods are surgery, radiotherapy, and chemotherapy. However, owning to the highly invasive nature of GBM, it is difficult to completely resect them due to the unclear boundary between the edges of GBM and normal brain tissue. Traditional radiotherapy and the combination of alkylating agents and radiotherapy have significant side effects, therapeutic drugs are difficult to penetrate the blood brain barrier. Patients receiving treatment have a high postoperative recurrence rate and a median survival of less than 2 years, Less than 5% of patients live longer than 5 years. Therefore, it is urgent to achieve precise treatment through the blood brain barrier and reduce toxic and side effects. Nanotechnology exhibit great potential in this area. This article summarizes the current treatment methods and shortcomings of GBM, and summarizes the research progress in the diagnosis and treatment of GBM using nanotechnology.
Collapse
Affiliation(s)
- Dongyan Wei
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, China
- College of Life Sciences, Tarim University, Alar, China
| | - Ni Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuang Qu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Hao Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jin Li
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Liu X, Zhang Y, Cui X, Fan T, Shu J, Li H, Huo X, Lu C. Gadopentetate meglumine activates mast cells to cause IgE-independent allergic reactions both in vitro and in vivo. Int Immunopharmacol 2022; 106:108602. [DOI: 10.1016/j.intimp.2022.108602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/19/2022] [Accepted: 01/30/2022] [Indexed: 11/05/2022]
|
3
|
Pashkunova-Martic I, Manzano-Szalai K, Friske J, Aszmann O, Theiner S, Klose MHM, Baurecht D, Trattnig S, Keppler BK, Helbich TH. Modified amino-dextrans as carriers of Gd-chelates for retrograde transport and visualization of peripheral nerves by magnetic resonance imaging (MRI). J Inorg Biochem 2021; 222:111495. [PMID: 34098348 DOI: 10.1016/j.jinorgbio.2021.111495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/22/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022]
Abstract
Amino-dextrans (AD) conjugated with gadolinium (Gd3+) were developed as neuro-specific contrast agents (CA) for the visualization of the sciatic nerve in rats by magnetic resonance imaging (MRI). AD with 3, 10, and 70 kDa molecular weights were assessed as carrier molecules known to be transported with various speed by axonal microtubules. Detailed spectroscopic characterizations, analyses by Fast Protein Liquid Chromatography (FPLC), Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis (SDS-PAGE), and inductively coupled plasma-mass spectrometry (ICP-MS), were carried out. For MRI, the paramagnetic Gd3+ ion was coupled as a T1 signal enhancer. The well-established linear chelator, diethylenetriaminepentaacetic acid (DTPA), was used and subsequently replaced by the more stable cyclic chelator 1,4,7,10-Tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA). In addition, a fluorescently labeled AD-DTPA-Gd was prepared to demonstrate an active transport to the spinal cord by histochemistry. After successful synthesis and characterization, molecular migration of the AD-DTPA-Gd in the sciatic nerve of healthy Sprague Dawley rats was monitored by MRI for up to seven days. Enhancement of nerve structures was evaluated by MRI and correlated with ICP-MS analyses. To investigate the distribution of CA along the neuraxis, all animals were sacrificed after the final MRI monitoring. Nerves, spinal ganglions, and corresponding spinal cord sections were harvested, to determine the localization and concentration of the paramagnetic element. This is the first report that demonstrates the active uptake and transport of AD-Gd conjugates within the sciatic nerve. This new concept may serve as a potential diagnostic tool for the direct visualization and monitoring of the continuity of injured nerves.
Collapse
Affiliation(s)
- Irena Pashkunova-Martic
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna & General Hospital of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria.
| | - Krisztina Manzano-Szalai
- Department of Surgery, Division of Plastic & Reconstructive Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Joachim Friske
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna & General Hospital of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, High Field MR Center, Lazarettgasse 14, 1090 Vienna, Austria
| | - Oskar Aszmann
- Department of Surgery, Division of Plastic & Reconstructive Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Sarah Theiner
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Matthias H M Klose
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, 1090 Vienna, Austria
| | - Dieter Baurecht
- Department of Physical Chemistry, University of Vienna, Vienna, Austria
| | - Siegfried Trattnig
- Department of Biomedical Imaging and Image-guided Therapy, High Field MR Center, Lazarettgasse 14, 1090 Vienna, Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna & General Hospital of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
4
|
Pol S, Schweser F, Bertolino N, Preda M, Sveinsson M, Sudyn M, Babek N, Zivadinov R. Characterization of leptomeningeal inflammation in rodent experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Exp Neurol 2019; 314:82-90. [PMID: 30684521 DOI: 10.1016/j.expneurol.2019.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 01/14/2019] [Accepted: 01/21/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Leptomeningeal inflammation, as evidenced by leptomeningeal contrast enhancement (LMCE), is associated to cortical pathology in multiple sclerosis. The temporal pattern of LMCE in experimental autoimmune encephalomyelitis (EAE) myelin oligodendrocyte glycoprotein (MOG) is unknown. OBJECTIVE To investigate LMCE using serial MRI in the EAE model of MS, and its association with clinical disease progression. To characterize the relationship between LMCE and underlying histological correlates. DESIGN Thirteen C57BL/6J mice, MOG-immunized (35-55 amino acid) and 8 saline injected animals were assessed at pre-induction and at 3, 6, 10, 20, 27, 32, 45 and 63 days post induction (dPI). LMCE scan was obtained using FLAIR-RARE sequence after post-contrast gadolinium administration on 9.4 T scanner. Brain cryo-sections were assessed for measuring cellular density of Iba1 positive macrophage/microglia at 10 dPI and 32 dPI, and for the presence of T, B and macrophage cells in the meningeal layer at 10 dPI and 63 dPI. RESULTS All EAE-MOG animals showed presence of LMCE and none of the control mice. The peak signal intensity of LMCE was evidenced at 10dPI in the meninges and decreased through 10-63 dPI. The peak of LMCE was associated with a weight loss starting at 1 week PI and with clinical symptoms starting at 2 weeks PI. Histological analysis of the brain tissue showed a higher density of Iba1 positive microglial cells in the EAE-MOG animals, corresponding to the areas of LMCE. Meninges of EAE mice showed higher density of Iba1 stained macrophage cells relative to saline animals. EAE animals also showed the presence of T and B cells in the meninges which were absent in the saline animals. CONCLUSIONS LMCE peak intensity in the meninges corresponds to the acute inflammatory phase of EAE-MOG disease progression, and is associated with clinical symptoms and higher inflammatory cell density.
Collapse
Affiliation(s)
- Suyog Pol
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Ferdinand Schweser
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Nicola Bertolino
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Marilena Preda
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Michele Sveinsson
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Michelle Sudyn
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Natan Babek
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
5
|
Acton PD. Multimodality Preclinical Imaging in Inflammatory Diseases. IMAGE FUSION IN PRECLINICAL APPLICATIONS 2019:135-160. [DOI: 10.1007/978-3-030-02973-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Boiziau C, Nikolski M, Mordelet E, Aussudre J, Vargas-Sanchez K, Petry KG. A Peptide Targeting Inflammatory CNS Lesions in the EAE Rat Model of Multiple Sclerosis. Inflammation 2018. [PMID: 29516383 DOI: 10.1007/s10753-018-0748-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis is characterized by inflammatory lesions dispersed throughout the central nervous system (CNS) leading to severe neurological handicap. Demyelination, axonal damage, and blood brain barrier alterations are hallmarks of this pathology, whose precise processes are not fully understood. In the experimental autoimmune encephalomyelitis (EAE) rat model that mimics many features of human multiple sclerosis, the phage display strategy was applied to select peptide ligands targeting inflammatory sites in CNS. Due to the large diversity of sequences after phage display selection, a bioinformatics procedure called "PepTeam" designed to identify peptides mimicking naturally occurring proteins was used, with the goal to predict peptides that were not background noise. We identified a circular peptide CLSTASNSC called "Ph48" as an efficient binder of inflammatory regions of EAE CNS sections including small inflammatory lesions of both white and gray matter. Tested on human brain endothelial cells hCMEC/D3, Ph48 was able to bind efficiently when these cells were activated with IL1β to mimic inflammatory conditions. The peptide is therefore a candidate for further analyses of the molecular alterations in inflammatory lesions.
Collapse
Affiliation(s)
- Claudine Boiziau
- INSERM, UMR 1049, F-33076, Bordeaux, France. .,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France. .,INSERM, UMR 1026, BioTis, F-33 076, Bordeaux, France.
| | - Macha Nikolski
- Univ. Bordeaux, CBiB, F-33076, Bordeaux, France.,CNRS, LaBRI UMR 5800, F-33400, Talence, France
| | - Elodie Mordelet
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France
| | - Justine Aussudre
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France
| | - Karina Vargas-Sanchez
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France.,Biomedical Sciences Research Group, GRINCIBIO, School of Medicine, Universidad Antonio Nariño, Bogotà, Colombia
| | - Klaus G Petry
- INSERM, UMR 1049, F-33076, Bordeaux, France.,Univ. Bordeaux, Neuroinflammation Imaging and Therapy of Multiple Sclerosis, F-33076, Bordeaux, France.,INSERM, UMR1029, F-33076, Bordeaux, France
| |
Collapse
|
7
|
Schafflick D, Kieseier BC, Wiendl H, Meyer Zu Horste G. Novel pathomechanisms in inflammatory neuropathies. J Neuroinflammation 2017; 14:232. [PMID: 29179723 PMCID: PMC5704548 DOI: 10.1186/s12974-017-1001-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/13/2017] [Indexed: 12/19/2022] Open
Abstract
Inflammatory neuropathies are rare autoimmune-mediated disorders affecting the peripheral nervous system. Considerable progress has recently been made in understanding pathomechanisms of these disorders which will be essential for developing novel diagnostic and therapeutic strategies in the future. Here, we summarize our current understanding of antigenic targets and the relevance of new immunological concepts for inflammatory neuropathies. In addition, we provide an overview of available animal models of acute and chronic variants and how new diagnostic tools such as magnetic resonance imaging and novel therapeutic candidates will benefit patients with inflammatory neuropathies in the future. This review thus illustrates the gap between pre-clinical and clinical findings and aims to outline future directions of development.
Collapse
Affiliation(s)
- David Schafflick
- Department of Neurology, Westfälische Wilhems-University, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Bernd C Kieseier
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology, Westfälische Wilhems-University, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Gerd Meyer Zu Horste
- Department of Neurology, Westfälische Wilhems-University, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
8
|
Finn JP, Nguyen KL, Hu P. Ferumoxytol vs. Gadolinium agents for contrast-enhanced MRI: Thoughts on evolving indications, risks, and benefits. J Magn Reson Imaging 2017; 46:919-923. [PMID: 28160356 PMCID: PMC10156572 DOI: 10.1002/jmri.25580] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/21/2016] [Indexed: 12/27/2022] Open
Affiliation(s)
- J Paul Finn
- Department of Radiological Sciences, UCLA, Los Angeles, California, USA.,Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,Department of Biomedical Physics, UCLA, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Peng Hu
- Department of Radiological Sciences, UCLA, Los Angeles, California, USA.,Department of Biomedical Physics, UCLA, Los Angeles, California, USA
| |
Collapse
|
9
|
Koppe T, Doneda D, Siebert M, Paskulin L, Camargo M, Tirelli KM, Vairo F, Daudt L, Schwartz IVD. The prognostic value of the serum ferritin in a southern Brazilian cohort of patients with Gaucher disease. Genet Mol Biol 2016; 39:30-4. [PMID: 27007895 PMCID: PMC4807389 DOI: 10.1590/1678-4685-gmb-2015-0125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 09/28/2015] [Indexed: 01/17/2023] Open
Abstract
The clinical utility of serum ferritin as a biomarker of disease severity and prognosis in Gaucher disease (GD) is still debated. Here, we aimed to evaluate ferritin and its relation to clinicolaboratory parameters of GD patients seen at the Reference Center for Gaucher Disease of Rio Grande do Sul, Brazil, so as to gather evidence on the utility of ferritin as a biomarker of this condition. A retrospective chart review was performed collecting pre-and posttreatment data from GD patients. Eighteen patients with ferritin levels available before and after treatment were included in the study. Nine of these participants were males, and seventeen had type I GD. All patients were given either enzyme replacement (n = 16) or substrate reduction therapy (n = 2), and ferritin was found to decrease from 756 [318-1441] ng/mL at baseline to 521 [227-626] ng/mL (p=0.025) after 28.8 month soft treatment. Serum ferritin levels did not correlate with measures of disease severity, but showed an association with age at onset of treatment (ρ= 0.880; n = 18; p < 0.001). In conclusion, although serum ferritin did not correlate with disease severity, after a median 28.8 months of treatment, clinical outcomes had clearly improved, and ferritin levels had decreased.
Collapse
Affiliation(s)
- Tiago Koppe
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Divair Doneda
- Laboratório de Técnica Dietética, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Marina Siebert
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Livia Paskulin
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Matheus Camargo
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Filippo Vairo
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Liane Daudt
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ida Vanessa D Schwartz
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
10
|
Gobbo OL, Sjaastad K, Radomski MW, Volkov Y, Prina-Mello A. Magnetic Nanoparticles in Cancer Theranostics. Theranostics 2015; 5:1249-63. [PMID: 26379790 PMCID: PMC4568452 DOI: 10.7150/thno.11544] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 07/15/2015] [Indexed: 12/18/2022] Open
Abstract
In a report from 2008, The International Agency for Research on Cancer predicted a tripled cancer incidence from 1975, projecting a possible 13-17 million cancer deaths worldwide by 2030. While new treatments are evolving and reaching approval for different cancer types, the main prevention of cancer mortality is through early diagnosis, detection and treatment of malignant cell growth. The last decades have seen a development of new imaging techniques now in widespread clinical use. The development of nano-imaging through fluorescent imaging and magnetic resonance imaging (MRI) has the potential to detect and diagnose cancer at an earlier stage than with current imaging methods. The characteristic properties of nanoparticles result in their theranostic potential allowing for simultaneous detection of and treatment of the disease. This review provides state of the art of the nanotechnological applications for cancer therapy. Furthermore, it advances a novel concept of personalized nanomedical theranostic therapy using iron oxide magnetic nanoparticles in conjunction with MRI imaging. Regulatory and industrial perspectives are also included to outline future perspectives in nanotechnological cancer research.
Collapse
Affiliation(s)
- Oliviero L. Gobbo
- 1. School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
- 2. Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
- 4. Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Kristine Sjaastad
- 3. Department of Clinical Medicine Trinity Centre for Health Science, St. James's Hospital, Dublin, Ireland
| | - Marek W. Radomski
- 1. School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
- 4. Trinity Biomedical Sciences Institute, Dublin, Ireland
- 5. Silesian Medical University and Kardio-Med Silesia, Zabrze, Poland
| | - Yuri Volkov
- 3. Department of Clinical Medicine Trinity Centre for Health Science, St. James's Hospital, Dublin, Ireland
- 6. Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Ireland
| | - Adriele Prina-Mello
- 3. Department of Clinical Medicine Trinity Centre for Health Science, St. James's Hospital, Dublin, Ireland
- 6. Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Ireland
| |
Collapse
|
11
|
Mouhieddine TH, Itani MM, Nokkari A, Ren C, Daoud G, Zeidan A, Mondello S, Kobeissy FH. Nanotheragnostic applications for ischemic and hemorrhagic strokes: improved delivery for a better prognosis. Curr Neurol Neurosci Rep 2015; 15:505. [PMID: 25394858 DOI: 10.1007/s11910-014-0505-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stroke is the second leading cause of death worldwide and a major cause of long-term severe disability representing a global health burden and one of the highly researched medical conditions. Nanostructured material synthesis and engineering have been recently developed and have been largely integrated into many fields including medicine. Recent studies have shown that nanoparticles might be a valuable tool in stroke. Different types, shapes, and sizes of nanoparticles have been used for molecular/biomarker profiling and imaging to help in early diagnosis and prevention of stroke and for drug/RNA delivery for improved treatment and neuroprotection. However, these promising applications have limitations, including cytotoxicity, which hindered their adoption into clinical use. Future research is warranted to fully develop and effectively and safely translate nanoparticles for stroke diagnosis and treatment into the clinic. This work will discuss the emerging role of nanotheragnostics in stroke diagnosis and treatment applications.
Collapse
Affiliation(s)
- Tarek H Mouhieddine
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon,
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Kollmer J, Bendszus M, Pham M. MR Neurography: Diagnostic Imaging in the PNS. Clin Neuroradiol 2015; 25 Suppl 2:283-9. [DOI: 10.1007/s00062-015-0412-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022]
|
13
|
Tagliafico A, Bignotti B, Tagliafico G, Martinoli C. Peripheral nerve MRI: precision and reproducibility of T2*-derived measurements at 3.0-T : a feasibility study. Skeletal Radiol 2015; 44:679-686. [PMID: 25631357 DOI: 10.1007/s00256-015-2106-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To prospectively evaluate the precision and reproducibility of T2*-derived measurements of the peripheral nerves. MATERIALS AND METHODS The study was approved by the local ethics committee and written informed consent was obtained. Bilateral upper and lower limb MRI examination was performed in 40 healthy subjects on a 3.0-T scanner. MRI protocol included T1-turbo spin-echo, T2-turbo spin-echo with fat suppression, and multiecho gradient recalled echo. Measurements of T2* times on T2* maps at different anatomical levels were performed. Three authors measured independently and in different sessions at baseline and after 4 weeks. Non-parametric tests and Bland-Altman statistics were used. RESULTS Minimum and maximum percentage variability were 10 % and 19 % for T2* (84-91 % of reproducibility). Maximum values of minimum detectable differences between limbs was 16 % (with 95 % CI: 2-37). Intra- and inter-observer agreement of the three radiologists for T2* was considered good. Evaluating the combined influence of the observer and of the repeated measurements the reproducibility was 87-98 %. CONCLUSIONS T2* measurement of the peripheral nerves is precise and reproducible. The healthy contralateral side can be used as an internal control. Variations in T2* values up to 16 % have to be considered.
Collapse
Affiliation(s)
- Alberto Tagliafico
- Institute of Anatomy, Department of Experimental Medicine, University of Genoa, Via L.B. Alberti, 16132, Genoa, Italy,
| | | | | | | |
Collapse
|
14
|
Rangavajla G, Mokarram N, Masoodzadehgan N, Pai SB, Bellamkonda RV. Noninvasive imaging of peripheral nerves. Cells Tissues Organs 2015; 200:69-77. [PMID: 25766202 DOI: 10.1159/000369451] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2014] [Indexed: 12/19/2022] Open
Abstract
Recent developments in the field of peripheral nerve imaging extend the capabilities of imaging modalities to assist in the diagnosis and treatment of patients with peripheral nerve maladies. Methods such as magnetic resonance imaging (MRI) and its derivative diffusion tensor imaging (DTI), ultrasound (US) and positron emission tomography (PET) are capable of assessing nerve structure and function following injury and relating the state of the nerve to electrophysiological and histological analysis. Of the imaging methods surveyed here, each offered unique and interesting advantages related to the field. MRI offered the opportunity to visualize immune activity on the injured nerve throughout the course of the regeneration process, and DTI offered numerical characterization of the injury and the ability to develop statistical bases for diagnosing injury. US extends imaging to the treatment phase by enabling more precise analgesic applications following surgery, and PET represents a novel method of assessing nerve injury through analysis of relative metabolism rates in injured and healthy tissue. Exciting new possibilities to enhance and extend the abilities of imaging methods are also discussed, including innovative contrast agents, some of which enable multimodal imaging approaches and present opportunities for treatment application.
Collapse
|
15
|
Su YY, Yang GF, Lu GM, Wu S, Zhang LJ. PET and MR imaging of neuroinflammation in hepatic encephalopathy. Metab Brain Dis 2015; 30:31-45. [PMID: 25514861 DOI: 10.1007/s11011-014-9633-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/17/2014] [Indexed: 12/11/2022]
Abstract
Neurological or psychiatric abnormalities associated with hepatic encephalopathy (HE) range from subclinical findings to coma. HE is commonly accompanied with the accumulation of toxic substances in bloodstream. The toxicity effect of hyperammonemia on astrocyte, such as the alteration in neurotransmission, oxidative stress, astrocyte swelling, is considered as an important factor in the pathogenesis of HE. Besides, neuroinflammation has captured more attention in the process of HE, but the mechanism of neuroinflammation leading to HE remains unclear. Molecular imaging techniques such as positron emission tomography (PET) and magnetic resonance imaging (MRI) targeting activated microglia and/ or other mediators appear to be promising noninvasive approaches to assess HE. This review focuses on novel imaging and therapy strategies of neuroinflammation in HE.
Collapse
Affiliation(s)
- Yun Yan Su
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, 305 Zhongshan East Road, Xuanwu District, Nangjing, Jiangsu Province, 210002, China
| | | | | | | | | |
Collapse
|
16
|
Kollmer J, Hund E, Hornung B, Hegenbart U, Schönland SO, Kimmich C, Kristen AV, Purrucker J, Röcken C, Heiland S, Bendszus M, Pham M. In vivo detection of nerve injury in familial amyloid polyneuropathy by magnetic resonance neurography. ACTA ACUST UNITED AC 2014; 138:549-62. [PMID: 25526974 PMCID: PMC4339768 DOI: 10.1093/brain/awu344] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
See Morrow and Reilly (doi:10.1093/awu396) for a scientific commentary on this article. Transthyretin familial amyloid polyneuropathy is a rare, autosomal-dominant multisystem disorder. Kollmer et al. show that high-resolution MR-neurography can quantify and localize lower limb nerve injury in vivo, both in symptomatic patients and in asymptomatic mutation carriers. Lesions appear at thigh-level and are predominantly proximal, although symptoms start and prevail distally. Transthyretin familial amyloid polyneuropathy is a rare, autosomal-dominant inherited multisystem disorder usually manifesting with a rapidly progressive, axonal, distally-symmetric polyneuropathy. The detection of nerve injury by nerve conduction studies is limited, due to preferential involvement of small-fibres in early stages. We investigated whether lower limb nerve-injury can be detected, localized and quantified in vivo by high-resolution magnetic resonance neurography. We prospectively included 20 patients (12 male and eight female patients, mean age 47.9 years, range 26–66) with confirmed mutation in the transthyretin gene: 13 with symptomatic polyneuropathy and seven asymptomatic gene carriers. A large age- and sex-matched cohort of healthy volunteers served as controls (20 male and 20 female, mean age 48.1 years, range 30–73). All patients received detailed neurological and electrophysiological examinations and were scored using the Neuropathy Impairment Score–Lower Limbs, Neuropathy Deficit and Neuropathy Symptom Score. Magnetic resonance neurography (3 T) was performed with large longitudinal coverage from proximal thigh to ankle-level and separately for each leg (140 axial slices/leg) by using axial T2-weighted (repetition time/echo time = 5970/55 ms) and dual echo (repetition time 5210 ms, echo times 12 and 73 ms) turbo spin echo 2D sequences with spectral fat saturation. A 3D T2-weighted inversion-recovery sequence (repetition time/echo time 3000/202 ms) was acquired for imaging of the spinal nerves and lumbar plexus (50 axial slice reformations). Precise manual segmentation of the spinal/sciatic/tibial/common peroneal nerves was performed on each slice. Histogram-based normalization of nerve–voxel signal intensities was performed using the age- and sex-matched control group as normative reference. Nerve-voxels were subsequently classified as lesion-voxels if a threshold of >1.2 (normalized signal-intensity) was exceeded. At distal thigh level, where a predominant nerve–lesion–voxel burden was observed, signal quantification was performed by calculating proton spin density and T2-relaxation time as microstructural markers of nerve tissue integrity. The total number of nerve–lesion voxels (cumulated from proximal-to-distal) was significantly higher in symptomatic patients (20 405 ± 1586) versus asymptomatic gene carriers (12 294 ± 3199; P = 0.036) and versus controls (6536 ± 467; P < 0.0001). It was also higher in asymptomatic carriers compared to controls (P = 0.043). The number of nerve–lesion voxels was significantly higher at thigh level compared to more distal levels (lower leg/ankle) of the lower extremities (f-value = 279.22, P < 0.0001). Further signal-quantification at this proximal site (thigh level) revealed a significant increase of proton-density (P < 0.0001) and T2-relaxation-time (P = 0.0011) in symptomatic patients, whereas asymptomatic gene-carriers presented with a significant increase of proton-density only. Lower limb nerve injury could be detected and quantified in vivo on microstructural level by magnetic resonance neurography in symptomatic familial amyloid polyneuropathy, and also in yet asymptomatic gene carriers, in whom imaging detection precedes clinical and electrophysiological manifestation. Although symptoms start and prevail distally, the focus of predominant nerve injury and injury progression was found proximally at thigh level with strong and unambiguous lesion-contrast. Imaging of proximal nerve lesions, which are difficult to detect by nerve conduction studies, may have future implications also for other distally-symmetric polyneuropathies.
Collapse
Affiliation(s)
- Jennifer Kollmer
- 1 Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Ernst Hund
- 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany 3 Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Benjamin Hornung
- 1 Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany 4 Medical Department V, University of Heidelberg, Heidelberg, Germany
| | - Stefan O Schönland
- 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany 4 Medical Department V, University of Heidelberg, Heidelberg, Germany
| | - Christoph Kimmich
- 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany 4 Medical Department V, University of Heidelberg, Heidelberg, Germany
| | - Arnt V Kristen
- 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany 5 Medical Department III, University of Heidelberg, Heidelberg, Germany
| | - Jan Purrucker
- 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany 3 Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Christoph Röcken
- 6 Department of Pathology, University Hospital Kiel, Kiel, Germany
| | - Sabine Heiland
- 1 Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany 7 Division of Experimental Radiology, Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany
| | - Martin Bendszus
- 1 Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany
| | - Mirko Pham
- 1 Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany 2 Amyloidosis Centre Heidelberg, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Schoknecht K, David Y, Heinemann U. The blood-brain barrier-gatekeeper to neuronal homeostasis: clinical implications in the setting of stroke. Semin Cell Dev Biol 2014; 38:35-42. [PMID: 25444848 DOI: 10.1016/j.semcdb.2014.10.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/24/2014] [Accepted: 10/31/2014] [Indexed: 12/23/2022]
Abstract
The blood-brain barrier is part of the neurovascular unit and serves as a functional and anatomical barrier between the blood and the extracellular space. It controls the flow of solutes in and out of the brain thereby providing an optimal environment for neuronal functioning. Paracellular transport between endothelial cells is restricted by tight junctions and transendothelial transport is reduced and more selective compared to capillaries of other organs. Further, the blood-brain barrier is involved in controlling blood flow and it is the site for signaling damage of the nervous system to the peripheral immune system. As an important player in brain homeostasis, blood-brain barrier dysfunction has been implicated in the pathophysiology of many brain diseases including stroke, traumatic brain injury, brain tumors, epilepsy and neurodegenerative disorders. In this article - highlighting recent advances in basic science - we review the features of the blood-brain barrier and their significance for neuronal homeostasis to discuss clinical implications for neurological complications following cerebral ischemia.
Collapse
Affiliation(s)
- Karl Schoknecht
- Institute for Neurophysiology, Charité - University Medicine Berlin, Germany
| | - Yaron David
- Departments of Physiology & Cell Biology, Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Uwe Heinemann
- Institute for Neurophysiology, Charité - University Medicine Berlin, Germany.
| |
Collapse
|
18
|
Zheng L, Li K, Han Y, Wei W, Zheng S, Zhang G. In vivo targeted peripheral nerve imaging with a nerve-specific nanoscale magnetic resonance probe. Med Hypotheses 2014; 83:588-92. [DOI: 10.1016/j.mehy.2014.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 07/09/2014] [Accepted: 08/11/2014] [Indexed: 11/26/2022]
|
19
|
Amhaoul H, Staelens S, Dedeurwaerdere S. Imaging brain inflammation in epilepsy. Neuroscience 2014; 279:238-52. [DOI: 10.1016/j.neuroscience.2014.08.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 01/15/2023]
|
20
|
Ciccarelli O, Barkhof F, Bodini B, Stefano ND, Golay X, Nicolay K, Pelletier D, Pouwels PJW, Smith SA, Wheeler-Kingshott CAM, Stankoff B, Yousry T, Miller DH. Pathogenesis of multiple sclerosis: insights from molecular and metabolic imaging. Lancet Neurol 2014; 13:807-22. [DOI: 10.1016/s1474-4422(14)70101-2] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
21
|
Golestani R, Sadeghi MM. Emergence of molecular imaging of aortic aneurysm: implications for risk stratification and management. J Nucl Cardiol 2014; 21:251-67; quiz 268-70. [PMID: 24381115 PMCID: PMC3991015 DOI: 10.1007/s12350-013-9845-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 11/19/2013] [Indexed: 12/21/2022]
Abstract
Imaging cellular and molecular processes associated with aneurysm expansion, dissection, and rupture can potentially transform the management of patients with thoracic and abdominal aortic aneurysm. Here, we review recent advances in molecular imaging of aortic aneurysm, focusing on imaging modalities with the greatest potential for clinical translation and application, PET, SPECT, and MRI. Inflammation (e.g., with (18)F-FDG, nanoparticles) and matrix remodeling (e.g., with matrix metalloproteinase-targeted tracers) are highlighted as promising targets for molecular imaging of aneurysm. Potential alternative or complementary approaches to molecular imaging for aneurysm risk stratification are briefly discussed.
Collapse
Affiliation(s)
- Reza Golestani
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
22
|
Ohana M, Moser T, Moussaouï A, Kremer S, Carlier R, Liverneaux P, Dietemann JL. Current and future imaging of the peripheral nervous system. Diagn Interv Imaging 2014; 95:17-26. [DOI: 10.1016/j.diii.2013.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Thurman JM, Serkova NJ. Nanosized contrast agents to noninvasively detect kidney inflammation by magnetic resonance imaging. Adv Chronic Kidney Dis 2013; 20:488-99. [PMID: 24206601 DOI: 10.1053/j.ackd.2013.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 06/02/2013] [Accepted: 06/03/2013] [Indexed: 12/15/2022]
Abstract
Several molecular imaging methods have been developed that use nanosized contrast agents to detect markers of inflammation within tissues. Kidney inflammation contributes to disease progression in a wide range of autoimmune and inflammatory diseases, and a biopsy is currently the only method of definitively diagnosing active kidney inflammation. However, the development of new molecular imaging methods that use contrast agents capable of detecting particular immune cells or protein biomarkers will allow clinicians to evaluate inflammation throughout the kidneys and to assess a patient's response to immunomodulatory drugs. These imaging tools will improve our ability to validate new therapies and to optimize the treatment of individual patients with existing therapies. This review describes the clinical need for new methods of monitoring kidney inflammation and recent advances in the development of nanosized contrast agents for the detection of inflammatory markers of kidney disease.
Collapse
|
24
|
Sillerud LO, Solberg NO, Chamberlain R, Orlando RA, Heidrich JE, Brown DC, Brady CI, Vander Jagt TA, Garwood M, Vander Jagt DL. SPION-enhanced magnetic resonance imaging of Alzheimer's disease plaques in AβPP/PS-1 transgenic mouse brain. J Alzheimers Dis 2013; 34:349-65. [PMID: 23229079 DOI: 10.3233/jad-121171] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In our program to develop non-invasive magnetic resonance imaging (MRI) methods for the diagnosis of Alzheimer's disease (AD), we have synthesized antibody-conjugated, superparamagnetic iron oxide nanoparticles (SPIONs) for use as an in vivo agent for MRI detection of amyloid-β plaques in AD. Here we report studies in AβPP/PS1 transgenic mice, which demonstrate the ability of novel anti-AβPP conjugated SPIONs to penetrate the blood-brain barrier to act as a contrast agent for MR imaging of plaques. The conspicuity of the plaques increased from an average Z-score of 5.1 ± 0.5 to 8.3 ± 0.2 when the plaque contrast to noise ratio was compared in control AD mice with AD mice treated with SPIONs. The number of MRI-visible plaques per brain increased from 347 ± 45 in the control AD mice, to 668 ± 86 in the SPION treated mice. These results indicated that our SPION enhanced amyloid-β detection method delivers an efficacious, non-invasive MRI detection method in transgenic mice.
Collapse
Affiliation(s)
- Laurel O Sillerud
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Navarathna DHMLP, Munasinghe J, Lizak MJ, Nayak D, McGavern DB, Roberts DD. MRI confirms loss of blood-brain barrier integrity in a mouse model of disseminated candidiasis. NMR IN BIOMEDICINE 2013; 26:1125-1134. [PMID: 23606437 PMCID: PMC3744627 DOI: 10.1002/nbm.2926] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 10/25/2012] [Accepted: 01/02/2013] [Indexed: 06/02/2023]
Abstract
Disseminated candidiasis primarily targets the kidneys and brain in mice and humans. Damage to these critical organs leads to the high mortality associated with such infections, and invasion across the blood-brain barrier can result in fungal meningoencephalitis. Candida albicans can penetrate a brain endothelial cell barrier in vitro through transcellular migration, but this mechanism has not been confirmed in vivo. MRI using the extracellular vascular contrast agent gadolinium diethylenetriaminepentaacetic acid demonstrated that integrity of the blood-brain barrier is lost during C. albicans invasion. Intravital two-photon laser scanning microscopy was used to provide the first real-time demonstration of C. albicans colonizing the living brain, where both yeast and filamentous forms of the pathogen were found. Furthermore, we adapted a previously described method utilizing MRI to monitor inflammatory cell recruitment into infected tissues in mice. Macrophages and other phagocytes were visualized in kidney and brain by the administration of ultrasmall iron oxide particles. In addition to obtaining new insights into the passage of C. albicans across the brain microvasculature, these imaging methods provide useful tools to study further the pathogenesis of C. albicans infections, to define the roles of Candida virulence genes in kidney versus brain infection and to assess new therapeutic measures for drug development.
Collapse
Affiliation(s)
- Dhammika H. M. L. P. Navarathna
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jeeva Munasinghe
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Martin J. Lizak
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Debasis Nayak
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Dorian B. McGavern
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
26
|
Harhausen D, Sudmann V, Khojasteh U, Müller J, Zille M, Graham K, Thiele A, Dyrks T, Dirnagl U, Wunder A. Specific imaging of inflammation with the 18 kDa translocator protein ligand DPA-714 in animal models of epilepsy and stroke. PLoS One 2013; 8:e69529. [PMID: 23936336 PMCID: PMC3732268 DOI: 10.1371/journal.pone.0069529] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 06/10/2013] [Indexed: 11/19/2022] Open
Abstract
Inflammation is a pathophysiological hallmark of many diseases of the brain. Specific imaging of cells and molecules that contribute to cerebral inflammation is therefore highly desirable, both for research and in clinical application. The 18 kDa translocator protein (TSPO) has been established as a suitable target for the detection of activated microglia/macrophages. A number of novel TSPO ligands have been developed recently. Here, we evaluated the high affinity TSPO ligand DPA-714 as a marker of brain inflammation in two independent animal models. For the first time, the specificity of radiolabeled DPA-714 for activated microglia/macrophages was studied in a rat model of epilepsy (induced using Kainic acid) and in a mouse model of stroke (transient middle cerebral artery occlusion, tMCAO) using high-resolution autoradiography and immunohistochemistry. Additionally, cold-compound blocking experiments were performed and changes in blood-brain barrier (BBB) permeability were determined. Target-to-background ratios of 2 and 3 were achieved in lesioned vs. unaffected brain tissue in the epilepsy and tMCAO models, respectively. In both models, ligand uptake into the lesion corresponded well with the extent of Ox42- or Iba1-immunoreactive activated microglia/macrophages. In the epilepsy model, ligand uptake was almost completely blocked by pre-injection of DPA-714 and FEDAA1106, another high-affinity TSPO ligand. Ligand uptake was independent of the degree of BBB opening and lesion size in the stroke model. We provide further strong evidence that DPA-714 is a specific ligand to image activated microglia/macrophages in experimental models of brain inflammation.
Collapse
Affiliation(s)
- Denise Harhausen
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Al Faraj A, Luciani N, Kolosnjaj-Tabi J, Mattar E, Clement O, Wilhelm C, Gazeau F. Real-time high-resolution magnetic resonance tracking of macrophage subpopulations in a murine inflammation model: a pilot study with a commercially available cryogenic probe. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:193-203. [PMID: 23281292 DOI: 10.1002/cmmi.1516] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 09/12/2012] [Accepted: 10/10/2012] [Indexed: 12/14/2022]
Abstract
Macrophages present different polarization states exhibiting distinct functions in response to environmental stimuli. However, the dynamic of their migration to sites of inflammation is not fully elucidated. Here we propose a real-time in vivo cell tracking approach, using high-resolution (HR)-MRI obtained with a commercially available cryogenic probe (Cryoprobe™), to monitor trafficking of differently polarized macrophages after systemic injection into mice. Murine bone marrow-derived mononuclear cells were differentiated ex vivo into nonpolarized M0, pro-inflammatory M1 and immunomodulator M2 macrophage subsets and labeled with citrate-coated anionic iron oxide nanoparticles (AMNP). These cells were subsequently intravenously injected to mice bearing calf muscle inflammation. Whole body migration dynamics of macrophage subsets was monitored by MRI at 4.7 T with a volume transmission/reception radiofrequency coil and macrophage infiltration to the inflamed paw was monitored with the cryogenic probe, allowing 3D spatial resolution of 50 µm with a scan time of only 10 min. Capture of AMNP was rapid and efficient regardless of macrophage polarization, with the highest uptake in M2 macrophages. Flow cytometry confirmed that macrophages preserved their polarization hallmarks after labeling. Migration kinetics of labeled cells differed from that of free AMNP. A preferential homing of M2-polarized macrophages to inflammation sites was observed. Our in vivo HR-MRI protocol highlights the extent of macrophage infiltration to the inflammation site. Coupled to whole body imaging, HR-MRI provides quantitative information on the time course of migration of ex vivo-polarized intravenously injected macrophages.
Collapse
Affiliation(s)
- Achraf Al Faraj
- Laboratoire Matière et Systèmes Complexes MSC, CNRS UMR7057, Université Paris Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
28
|
West CA, Ljungberg C, Wiberg M, Hart A. Sensory Neuron Death After Upper Limb Nerve Injury and Protective Effect of Repair. Neurosurgery 2013; 73:632-9; discussion 640. [DOI: 10.1227/neu.0000000000000066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Abstract
BACKGROUND:
Extensive death of sensory neurons after nerve trauma depletes the number of regenerating neurons, contributing to inadequate cutaneous innervation density and poor sensory recovery. Experimentally proven neuroprotective neoadjuvant drugs require noninvasive in vivo measures of neuron death to permit clinical trials. In animal models of nerve transection, magnetic resonance imaging (MRI) proved a valid tool for quantifying sensory neuron loss within dorsal root ganglia (DRG) by measuring consequent proportional shrinkage of respective ganglia.
OBJECTIVE:
This system is investigated for clinical application after upper limb nerve injury and microsurgical nerve repair.
METHODS:
A 3-T clinical magnet was used to image and measure volume (Cavalieri principle) of C7-T1 DRG in uninjured volunteers (controls, n = 14), hand amputees (unrepaired nerve injury, n = 5), and early nerve repair patients (median and ulnar nerves transected, microsurgical nerve repair within 24 hours, n = 4).
RESULTS:
MRI was well tolerated. Volumetric analysis was feasible in 74% of patients. A mean 14% volume reduction was found in amputees' C7 and C8 DRG (P < .001 vs controls). Volume loss was lower in median and ulnar nerve repair patients (mean 3% volume loss, P < .01 vs amputees), and varied among patients. T1 DRG volume remained unaffected.
CONCLUSION:
MRI provides noninvasive in vivo assessment of DRG volume as a proxy clinical measure of sensory neuron death. The significant decrease found after unrepaired nerve injury provides indirect clinical evidence of axotomy-induced neuronal death. This loss was less after nerve repair, indicating a neuroprotective benefit of early repair. Volumetric MRI has potential diagnostic applications and is a quantitative tool for clinical trials of neuroprotective therapies.
Collapse
Affiliation(s)
- Christian Alexander West
- Department of Integrative Medical Biology, Section for Anatomy, Umea University, Umea, Sweden
- Department of Surgical and Perioperative Science, Section for Hand & Plastic Surgery, University Hospital, Umea, Sweden
| | - Christina Ljungberg
- Department of Integrative Medical Biology, Section for Anatomy, Umea University, Umea, Sweden
- Department of Surgical and Perioperative Science, Section for Hand & Plastic Surgery, University Hospital, Umea, Sweden
| | - Mikael Wiberg
- Department of Integrative Medical Biology, Section for Anatomy, Umea University, Umea, Sweden
- Department of Surgical and Perioperative Science, Section for Hand & Plastic Surgery, University Hospital, Umea, Sweden
| | - Andrew Hart
- Department of Surgical and Perioperative Science, Section for Hand & Plastic Surgery, University Hospital, Umea, Sweden
- Plastic Surgery Research, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Canniesburn Plastic Surgery Unit, Scottish National Brachial Plexus Service, Glasgow Royal Infirmary, Glasgow, United Kingdom
| |
Collapse
|
29
|
Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE. Inflammation and adaptive immunity in Parkinson's disease. Cold Spring Harb Perspect Med 2013; 2:a009381. [PMID: 22315722 DOI: 10.1101/cshperspect.a009381] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The immune system is designed to protect the host from infection and injury. However, when an adaptive immune response continues unchecked in the brain, the proinflammatory innate microglial response leads to the accumulation of neurotoxins and eventual neurodegeneration. What drives such responses are misfolded and nitrated proteins. Indeed, the antigen in Parkinson's disease (PD) is an aberrant self-protein, although the adaptive immune responses are remarkably similar in a range of diseases. Ingress of lymphocytes and chronic activation of glial cells directly affect neurodegeneration. With this understanding, new therapies aimed at modulating the immune system's response during PD could lead to decreased neuronal loss and improved clinical outcomes for disease.
Collapse
Affiliation(s)
- R Lee Mosley
- Movement Disorders Program, Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | |
Collapse
|
30
|
Marinescu M, Langer M, Durand A, Olivier C, Chabrol A, Rositi H, Chauveau F, Cho TH, Nighoghossian N, Berthezène Y, Peyrin F, Wiart M. Synchrotron Radiation X-Ray Phase Micro-computed Tomography as a New Method to Detect Iron Oxide Nanoparticles in the Brain. Mol Imaging Biol 2013; 15:552-9. [DOI: 10.1007/s11307-013-0639-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
Sun Z, Yathindranath V, Worden M, Thliveris JA, Chu S, Parkinson FE, Hegmann T, Miller DW. Characterization of cellular uptake and toxicity of aminosilane-coated iron oxide nanoparticles with different charges in central nervous system-relevant cell culture models. Int J Nanomedicine 2013; 8:961-70. [PMID: 23494517 PMCID: PMC3593762 DOI: 10.2147/ijn.s39048] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Aminosilane-coated iron oxide nanoparticles (AmS-IONPs) have been widely used in constructing complex and multifunctional drug delivery systems. However, the biocompatibility and uptake characteristics of AmS-IONPs in central nervous system (CNS)-relevant cells are unknown. The purpose of this study was to determine the effect of surface charge and magnetic field on toxicity and uptake of AmS-IONPs in CNS-relevant cell types. Methods The toxicity and uptake profile of positively charged AmS-IONPs and negatively charged COOH-AmS-IONPs of similar size were examined using a mouse brain microvessel endothelial cell line (bEnd.3) and primary cultured mouse astrocytes and neurons. Cell accumulation of IONPs was examined using the ferrozine assay, and cytotoxicity was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Results No toxicity was observed in bEnd.3 cells at concentrations up to 200 μg/mL for either AmS-IONPs or COOH-AmS-IONPs. AmS-IONPs at concentrations above 200 μg/mL reduced neuron viability by 50% in the presence or absence of a magnetic field, while only 20% reductions in viability were observed with COOH-AmS-IONPs. Similar concentrations of AmS-IONPs in astrocyte cultures reduced viability to 75% but only in the presence of a magnetic field, while exposure to COOH-AmS-IONPs reduced viability to 65% and 35% in the absence and presence of a magnetic field, respectively. Cellular accumulation of AmS-IONPs was greater in all cell types examined compared to COOH-AmS-IONPs. Rank order of cellular uptake for AmS-IONPs was astrocytes > bEnd.3 > neurons. Accumulation of COOH-AmS-IONPs was minimal and similar in magnitude in different cell types. Magnetic field exposure enhanced cellular accumulation of both AmS- and COOH-AmS-IONPs. Conclusion Both IONP compositions were nontoxic at concentrations below 100 μg/mL in all cell types examined. At doses above 100 μg/mL, neurons were more sensitive to AmS-IONPs, whereas astrocytes were more vulnerable toward COOH-AmS-IONPs. Toxicity appears to be dependent on the surface coating as opposed to the amount of iron-oxide present in the cell.
Collapse
Affiliation(s)
- Zhizhi Sun
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Coan AC, Cendes F. Epilepsy as progressive disorders: what is the evidence that can guide our clinical decisions and how can neuroimaging help? Epilepsy Behav 2013; 26:313-21. [PMID: 23127969 DOI: 10.1016/j.yebeh.2012.09.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 09/16/2012] [Indexed: 10/27/2022]
Abstract
There is evidence that some types of epilepsy progress over time, and an important part of this knowledge has derived from neuroimaging studies. Different authors have demonstrated structural damage more pronounced in individuals with a longer duration of epilepsy, and others have been able to quantify this progression over time. However, others have failed to demonstrate progression possibly due to the heterogeneity of individuals evaluated. Currently, temporal lobe epilepsy associated with hippocampal sclerosis is regarded as a progressive disorder. Conversely, for other types of epilepsy, the evidence is not so clear. The causes of this damage progression are also unknown although there is consistent evidence that seizure is one of the mechanisms. The conflicting data about epilepsy progression can be a challenge for clinical decisions for an individual patient. Studies with homogenous groups and longer follow-up are necessary for appropriate conclusions about the real burden of damage progression in epilepsies, and neuroimaging will be essential in this context.
Collapse
Affiliation(s)
- Ana C Coan
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, SP, Brazil
| | | |
Collapse
|
33
|
Tourdias T, Dousset V. Neuroinflammatory imaging biomarkers: relevance to multiple sclerosis and its therapy. Neurotherapeutics 2013; 10:111-23. [PMID: 23132327 PMCID: PMC3557362 DOI: 10.1007/s13311-012-0155-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Magnetic resonance imaging is an established tool in the management of multiple sclerosis (MS). Loss of blood brain barrier integrity assessed by gadolinium (Gd) enhancement is the current standard marker of MS activity. To explore the complex cascade of the inflammatory events, other magnetic resonance imaging, but also positron emission tomographic markers reviewed in this article are being developed to address active neuroinflammation with increased sensitivity and specificity. Alternative magnetic resonance contrast agents, positron emission tomographic tracers and imaging techniques could be more sensitive than Gd to early blood brain barrier alteration, and they could assess the inflammatory cell recruitment and/or the associated edema accumulation. These markers of active neuroinflammation, although some of them are limited to experimental studies, could find great relevance to complete Gd information and thereby increase our understanding of acute lesion pathophysiology and its noninvasive follow-up, especially to monitor treatment efficacy. Furthermore, such accurate markers of inflammation combined with those of neurodegeneration hold promise to provide a more complete picture of MS, which will be of great benefit for future therapeutic strategies.
Collapse
Affiliation(s)
- Thomas Tourdias
- INSERM Unit 1049 Neuroinflammation, Imagerie et Thérapie de la Sclérose en Plaques, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, F-33076, France.
| | | |
Collapse
|
34
|
Stoll G, Wilder-Smith E, Bendszus M. Imaging of the peripheral nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2013; 115:137-153. [PMID: 23931778 DOI: 10.1016/b978-0-444-52902-2.00008-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
This chapter summarizes progress in the evaluation of peripheral nerve (PN) lesions and disorders by imaging techniques encompassing magnetic resonance imaging (MRI) and nerve ultrasound (US). Due to the radiation exposure and limited sensitivity in soft tissue contrast, computed-tomography (CT) plays no significant role in the diagnostic work-up of PN disorders. MRI and US are complementary techniques for the evaluation of peripheral nerves, each having particular advantages and disadvantages. Nerve injury induces intrinsic MRI signal alterations on T2-weighted sequences in degenerating or demyelinating nerve segments as well as in corresponding muscle groups exhibiting denervation which can be exploited diagnostically. Nerve US is based on changes in the nerve echotexture due to tumor formation or focal enlargement caused by entrapment or inflammation. Both MRI and US provide morphological information on the precise site and extent of nerve injury. While US has the advantage of easy accessibility, providing images with superior spatial resolution at low cost, MRI shows better soft tissue contrast and better image quality for deep-lying nerve structures since imaging is not hindered by bone. Recent advances have remarkably increased spatial resolution of both MRI and US making imaging indispensible for the elucidation of causes of nerve compression, peripheral nerve tumors, and focal inflammatory conditions. Both MRI and US further guide neurosurgical exploration and can simplify treatment. Importantly, imaging can reveal treatable conditions even in the absence of gross electrophysiological alterations, illustrating its increasing role in clinical practice. In experimental settings, novel molecular and cellular MRI contrast agents allow in-vivo assessment of nerve regeneration as well as monitoring of neuroinflammation. Depending on further clinical development, contrast-enhanced MRI has the potential to follow cellular responses over time in vivo and to overcome the current limitations of histological assessment of nerve afflictions. Further advances in contrast-enhanced US has the potential for developing into a tool for the assessment of nerve blood perfusion, paving the way for better assessments of ischemic neuropathies.
Collapse
Affiliation(s)
- Guido Stoll
- Department of Neurology, University of Würzburg, Würzburg, Germany.
| | | | | |
Collapse
|
35
|
Lagraoui M, Latoche JR, Cartwright NG, Sukumar G, Dalgard CL, Schaefer BC. Controlled cortical impact and craniotomy induce strikingly similar profiles of inflammatory gene expression, but with distinct kinetics. Front Neurol 2012; 3:155. [PMID: 23118733 PMCID: PMC3484408 DOI: 10.3389/fneur.2012.00155] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 10/09/2012] [Indexed: 11/13/2022] Open
Abstract
An immediate consequence of traumatic brain injury (TBI) is the induction of an inflammatory response. Mounting data suggest that inflammation is a major contributor to TBI-induced brain damage. However, much remains unknown regarding the induction and regulation of the inflammatory response to TBI. In this study we compared the TBI-induced inflammatory response to severe parenchymal injury (controlled cortical impact) vs. mild brain injury (craniotomy) over a 21-day period. Our data show that both severe and mild brain injury induce a qualitatively similar inflammatory response, involving highly overlapping sets of effector molecules. However, kinetic analysis revealed that the inflammatory response to mild brain injury is of much shorter duration than the response to severe TBI. Specifically, the inflammatory response to severe brain injury persists for at least 21 days, whereas the response to mild brain injury returns to near baseline values within 10 days post-injury. Our data therefore imply that the development of accurate diagnostic tests of TBI severity that are based on imaging or biomarker analysis of the inflammatory response may require repeated measures over at least a 10-day period, post-injury.
Collapse
Affiliation(s)
- Mouna Lagraoui
- Department of Microbiology and Immunology, Uniformed Services University Bethesda, MD, USA ; Center for Neuroscience and Regenerative Medicine, Uniformed Services University Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
36
|
Kollmer J, Bäumer P, Milford D, Dombert T, Staub F, Bendszus M, Pham M. T2-signal of ulnar nerve branches at the wrist in guyon's canal syndrome. PLoS One 2012; 7:e47295. [PMID: 23071777 PMCID: PMC3468548 DOI: 10.1371/journal.pone.0047295] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/11/2012] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE To evaluate T2-signal of high-resolution MRI in distal ulnar nerve branches at the wrist as diagnostic sign of guyon's-canal-syndrome (GCS). MATERIALS AND METHODS 11 GCS patients confirmed by clinical/electrophysiological findings, and 20 wrists from 11 asymptomatic volunteers were prospectively included to undergo the following protocol: axial T2-weighted-fat-suppressed and T1-weighted-turbo-spin-echo-sequences (3T-MR-scanner, Magnetom/Verio/Siemens). Patients were examined in prone position with the arm extended and wrist placed in an 8-channel surface-array-coil. Nerve T2-signal was evaluated as contrast-to-noise-ratios (CNR) from proximal-to-distal in ulnar nerve trunk, its superficial/sensory and deep/motor branch. Distal motor-nerve-conduction (distal-motor-latency (dml)) to first dorsal-interosseus (IOD I) and abductor digiti minimi muscles was correlated with T2-signal. Approval by the institutional review-board and written informed consent was given by all participants. RESULTS In GCS, mean nerve T2-signal was strongly increased within the deep/motor branch (11.7±4.8 vs.controls:-5.3±2.4;p = 0.001) but clearly less and not significantly increased in ulnar nerve trunk (6.8±6.4vs.-7.4±2.5;p = 0.07) and superficial/sensory branch (-2.1±4.9vs.-9.7±2.9;p = 0.08). Median nerve T2-signal did not differ between patients and controls (-9.8±2.5vs.-6.7±4.2;p = 0.45). T2-signal of deep/motor branch correlated strongly with motor-conduction-velocity to IOD I in non-linear fashion (R(2) = -0.8;p<0.001). ROC-analysis revealed increased nerve T2-signal of the deep/motor branch to be a sign of excellent diagnostic performance (area-under-the-curve 0.94, 95% CI: 0.85-1.00; specificity 90%, sensitivity 89.5%). CONCLUSIONS Nerve T2-signal increase of distal ulnar nerve branches and in particular of the deep/motor branch is highly accurate for the diagnostic determination of GCS. Furthermore, for the first time it was found in nerve entrapment injury that T2-signal strongly correlates with electrical-conduction-velocity.
Collapse
Affiliation(s)
- Jennifer Kollmer
- Department of Neuroradiology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
37
|
Tourdias T, Roggerone S, Filippi M, Kanagaki M, Rovaris M, Miller DH, Petry KG, Brochet B, Pruvo JP, Radüe EW, Dousset V. Assessment of disease activity in multiple sclerosis phenotypes with combined gadolinium- and superparamagnetic iron oxide-enhanced MR imaging. Radiology 2012; 264:225-33. [PMID: 22723563 DOI: 10.1148/radiol.12111416] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To compare magnetic resonance (MR) imaging features of multiple sclerosis (MS) lesions after the administration of a gadolinium-based contrast agent and ultrasmall superparamagnetic iron oxide (USPIO) particles among the clinical phenotypes of MS and over time. MATERIALS AND METHODS This study was approved by the local ethics committee, and written informed consent was obtained from all patients. Twenty-four patients with MS (10 with relapsing and 14 with progressive forms) underwent clinical and gadolinium- and USPIO-enhanced MR examinations at baseline and 6-month follow-up. The number of lesions that enhanced with gadolinium alone, USPIO alone, or both was compared with the Pearson χ2 or Fisher exact test, and lesion sizes were compared with the Wilcoxon Mann-Whitney U test. At 6-month follow-up, the lesion signal intensity on precontrast T1-weighted images and the enhancement after repeat injection of the contrast agent were compared with the baseline postcontrast imaging features by using the McNemar test. RESULTS Fifty-six lesions were considered active owing to contrast enhancement at baseline; 37 lesions (66%) in 10 patients enhanced with gadolinium. The use of USPIO helped detect 19 additional lesions (34%), and two additional patients were classified as having active disease. Thus, the use of both agents enabled detection of 51% (19 of 37 lesions) more lesions than with gadolinium alone. Enhanced lesions were more frequently observed in the relapsing compared with the progressive forms of MS (P<.0001). USPIO enhancement, in the form of ringlike patterns, could also be observed on T1-weighted images in patients with progressive MS, enabling the detection of five lesions in addition to the five detected with gadolinium in this phenotype. Lesions that enhanced with both contrast agents at baseline were larger (mean size, 6.5 mm±3.8; P=.001) and were more likely to persistently enhance at 6-month follow-up (seven of 27 lesions, P<.0001) compared with those that enhanced only with gadolinium (mean size, 4.9 mm±2.2; one of nine lesions) or USPIO (mean size, 3.5 mm±1.5; 0 of 17 lesions). CONCLUSION The combination of gadolinium and USPIO in patients with MS can help identify additional active lesions compared with the current standard, the gadolinium-only approach, even in progressive forms of MS. Lesions that enhance with both agents may exhibit a more aggressive evolution than those that enhance with only one contrast agent.
Collapse
Affiliation(s)
- Thomas Tourdias
- Department of Neuroradiology and INSERM U1049, CHU de Bordeaux, Université Bordeaux Segalen, Bordeaux, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Boltze J, Kleinschnitz C, Reymann KG, Reiser G, Wagner DC, Kranz A, Michalski D. Neurovascular pathophysiology in cerebral ischemia, dementia and the ageing brain - current trends in basic, translational and clinical research. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:14. [PMID: 22883324 PMCID: PMC3431988 DOI: 10.1186/2040-7378-4-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 08/03/2012] [Indexed: 06/01/2023]
Abstract
The 7th International Symposium on Neuroprotection and Neurorepair was held from May 2nd to May 5th, 2012 in Potsdam, Germany. The symposium, which directly continues the successful Magdeburg meeting series, attracted over 330 colleagues from 29 countries to discuss recent findings and advances in the field. The focus of the 2012 symposium was widened from stroke and traumatic brain injury to neurodegenerative diseases, notably dementia, and more generally the ageing brain. Thereby, emphasis was given on neurovascular aspects of neurodegeneration and stroke including the blood-brain barrier, recent findings regarding the pathomechanism of Alzheimer's disease, and brain imaging approaches. In addition, neurobiochemical aspects of neuroprotection, the role of astrogliosis, the clinical progress of cell-based approaches as well as translational hurdles and opportunities were discussed in-depth. This review summarizes some of the most stimulating discussions and reports from the meeting.
Collapse
Affiliation(s)
- Johannes Boltze
- Fraunhofer-Institute for Cell Therapy and Immunology, Perlickstr. 1, D-04103 Leipzig, Germany
- Translational Centre for Regenerative Medicine, University of Leipzig, Philipp-Rosenthal-Str. 55, D-04103 Leipzig, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, D-97080 Würzburg, Germany
| | - Klaus G Reymann
- Leibniz Institute for Neurobiology, Project Group Neuropharmacology, Brenneckestr. 6, D-39118 Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), site Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Georg Reiser
- Institute of Neurobiochemistry, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Daniel-Christoph Wagner
- Fraunhofer-Institute for Cell Therapy and Immunology, Perlickstr. 1, D-04103 Leipzig, Germany
- Translational Centre for Regenerative Medicine, University of Leipzig, Philipp-Rosenthal-Str. 55, D-04103 Leipzig, Germany
| | - Alexander Kranz
- Fraunhofer-Institute for Cell Therapy and Immunology, Perlickstr. 1, D-04103 Leipzig, Germany
- Translational Centre for Regenerative Medicine, University of Leipzig, Philipp-Rosenthal-Str. 55, D-04103 Leipzig, Germany
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, Liebigstr. 20, D-04103, Leipzig Germany
| |
Collapse
|
39
|
In vivo USPIO magnetic resonance imaging shows that minocycline mitigates macrophage recruitment to a peripheral nerve injury. Mol Pain 2012; 8:49. [PMID: 22742763 PMCID: PMC3472277 DOI: 10.1186/1744-8069-8-49] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 06/18/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Minocycline has proven anti-nociceptive effects, but the mechanism by which minocycline delays the development of allodynia and hyperalgesia after peripheral nerve injury remains unclear. Inflammatory cells, in particular macrophages, are critical components of the response to nerve injury. Using ultrasmall superparamagnetic iron oxide-magnetic resonance imaging (USPIO-MRI) to monitor macrophage trafficking, the purpose of this project is to determine whether minocycline modulates macrophage trafficking to the site of nerve injury in vivo and, in turn, results in altered pain thresholds. RESULTS Animal experiments were approved by Stanford IACUC. A model of neuropathic pain was created using the Spared Nerve Injury (SNI) model that involves ligation of the left sciatic nerve in the left thigh of adult Sprague-Dawley rats. Animals with SNI and uninjured animals were then injected with/without USPIOs (300 μmol/kg i.v.) and with/without minocycline (50 mg/kg i.p.). Bilateral sciatic nerves were scanned with a volume coil in a 7 T magnet 7 days after USPIO administration. Fluid-sensitive MR images were obtained, and ROIs were placed on bilateral sciatic nerves to quantify signal intensity. Pain behavior modulation by minocycline was measured using the Von Frey filament test. Sciatic nerves were ultimately harvested at day 7, fixed in 10% buffered formalin and stained for the presence of iron oxide-laden macrophages. Behavioral measurements confirmed the presence of allodynia in the neuropathic pain model while the uninjured and minocycline-treated injured group had significantly higher paw withdrawal thresholds (p < 0.011). Decreased MR signal is observed in the SNI group that received USPIOs (3.3+/-0.5%) compared to the minocycline-treated SNI group that received USPIOs (15.2+/-4.5%) and minocycline-treated group that did not receive USPIOs (41.2+/-2.3%) (p < 0.04). Histology of harvested sciatic nerve specimens confirmed the presence USPIOs at the nerve injury site in the SNI group without minocycline treatment. CONCLUSION Animals with neuropathic pain in the left hindpaw show increased trafficking of USPIO-laden macrophages to the site of sciatic nerve injury. Minocycline to retards the migration of macrophages to the nerve injury site, which may partly explain its anti-nociceptive effects. USPIO-MRI is an effective in vivo imaging tool to study the role of macrophages in the development of neuropathic pain.
Collapse
|
40
|
Evans MC, Modo M, Talbot K, Sibson N, Turner MR. Magnetic resonance imaging of pathological processes in rodent models of amyotrophic lateral sclerosis. ACTA ACUST UNITED AC 2012; 13:288-301. [DOI: 10.3109/17482968.2011.623300] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Matthew C. Evans
- Oxford University Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital,
Oxford, UK
| | - Michel Modo
- University of Pittsburgh Department of Radiology & McGowan Center for Regenerative Medicine,
Pittsburgh, USA
| | - Kevin Talbot
- Oxford University Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital,
Oxford, UK
| | - Niki Sibson
- Oxford University Gray Institute for Radiation Oncology and Biology, Churchill Hospital,
Oxford, UK
| | - Martin R. Turner
- Oxford University Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital,
Oxford, UK
| |
Collapse
|
41
|
van Tilborg GAF, Cormode DP, Jarzyna PA, van der Toorn A, van der Pol SMA, van Bloois L, Fayad ZA, Storm G, Mulder WJM, de Vries HE, Dijkhuizen RM. Nanoclusters of iron oxide: effect of core composition on structure, biocompatibility, and cell labeling efficacy. Bioconjug Chem 2012; 23:941-50. [PMID: 22471239 DOI: 10.1021/bc200543k] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inorganic nanocrystals have a variety of applications in medicine. They may serve as contrast agents, therapeutics, and for in vitro diagnostics. Frequently, the synthesis route yields hydrophobically capped nanocrystals, which necessitates their subsequent coating to render a water-soluble and biocompatible probe. Biocompatibility is crucial for cellular imaging applications, which require large quantities of diagnostically active nanoparticles to be loaded into cells. We have previously reported the design and synthesis of a fluorescent and magnetic resonance imaging-detectable core-shell nanoparticle that encapsulates hydrophobically coated iron oxide nanocrystals. The core of soybean oil and iron oxide is covered by a shell mixture of phospholipids, some of which contained polyethylene glycol. Despite the biocompatibility of these components, we hypothesize that we can improve this formulation with respect to in vitro toxicity. To this aim, we measured the effect of six different core compositions on nanoparticle structure, cell labeling efficacy, and cell viability, as well as cell tracking potential. We methodically investigated the causes of toxicity and conclude that, even when combining biocompatible materials, the resulting formulation is not guaranteed to be biocompatible.
Collapse
Affiliation(s)
- Geralda A F van Tilborg
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht , Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Perron H, Germi R, Bernard C, Garcia-Montojo M, Deluen C, Farinelli L, Faucard R, Veas F, Stefas I, Fabriek BO, Van-Horssen J, Van-der-Valk P, Gerdil C, Mancuso R, Saresella M, Clerici M, Marcel S, Creange A, Cavaretta R, Caputo D, Arru G, Morand P, Lang AB, Sotgiu S, Ruprecht K, Rieckmann P, Villoslada P, Chofflon M, Boucraut J, Pelletier J, Hartung HP. Human endogenous retrovirus type W envelope expression in blood and brain cells provides new insights into multiple sclerosis disease. Mult Scler 2012; 18:1721-36. [PMID: 22457345 PMCID: PMC3573672 DOI: 10.1177/1352458512441381] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: The envelope protein from multiple sclerosis (MS) associated retroviral element (MSRV), a member of the Human Endogenous Retroviral family ‘W’ (HERV-W), induces dysimmunity and inflammation. Objective: The objective of this study was to confirm and specify the association between HERV-W/MSRV envelope (Env) expression and MS. Methods: 103 MS, 199 healthy controls (HC) and controls with other neurological diseases (28), chronic infections (30) or autoimmunity (30) were analysed with an immunoassay detecting Env in serum. Env RNA or DNA copy numbers in peripheral blood mononuclear cells (PBMC) were determined by a quantitative polymerase chain reaction (PCR). Env was detected by immunohistology in the brains of patients with MS with three specific monoclonals. Results: Env antigen was detected in a serum of 73% of patients with MS with similar prevalence in all clinical forms, and not in chronic infection, systemic lupus, most other neurological diseases and healthy donors (p<0.01). Cases with chronic inflammatory demyelinating polyneuropathy (5/8) and rare HC (4/103) were positive. RNA expression in PBMC and DNA copy numbers were significantly elevated in patients with MS versus HC (p<0.001). In patients with MS, DNA copy numbers were significantly increased in chronic progressive MS (secondary progressive MS vs relapsing–remitting MS (RRMS) p<0.001; primary progressive MS vs RRMS –<0.02). Env protein was evidenced in macrophages within MS brain lesions with particular concentrations around vascular elements. Conclusion: The association between MS disease and the MSRV-type HERV-W element now appears quite strong, as evidenced ex-vivo from serum and PBMC with post-mortem confirmation in brain lesions. Chronic progressive MS, RRMS and clinically isolated syndrome show different ELISA (Enzyme-Linked Immunosorbent Assay) and/or PCR profiles suggestive of an increase with disease evolution, and amplicon sequencing confirms the association with particular HERV-W elements.
Collapse
|
43
|
Stoll G, Basse-Lüsebrink T, Weise G, Jakob P. Visualization of inflammation using19F-magnetic resonance imaging and perfluorocarbons. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2012; 4:438-47. [DOI: 10.1002/wnan.1168] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
44
|
Robbins ME, Brunso-Bechtold JK, Peiffer AM, Tsien CI, Bailey JE, Marks LB. Imaging radiation-induced normal tissue injury. Radiat Res 2012; 177:449-66. [PMID: 22348250 DOI: 10.1667/rr2530.1] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Technological developments in radiation therapy and other cancer therapies have led to a progressive increase in five-year survival rates over the last few decades. Although acute effects have been largely minimized by both technical advances and medical interventions, late effects remain a concern. Indeed, the need to identify those individuals who will develop radiation-induced late effects, and to develop interventions to prevent or ameliorate these late effects is a critical area of radiobiology research. In the last two decades, preclinical studies have clearly established that late radiation injury can be prevented/ameliorated by pharmacological therapies aimed at modulating the cascade of events leading to the clinical expression of radiation-induced late effects. These insights have been accompanied by significant technological advances in imaging that are moving radiation oncology and normal tissue radiobiology from disciplines driven by anatomy and macrostructure to ones in which important quantitative functional, microstructural, and metabolic data can be noninvasively and serially determined. In the current article, we review use of positron emission tomography (PET), single photon emission tomography (SPECT), magnetic resonance (MR) imaging and MR spectroscopy to generate pathophysiological and functional data in the central nervous system, lung, and heart that offer the promise of, (1) identifying individuals who are at risk of developing radiation-induced late effects, and (2) monitoring the efficacy of interventions to prevent/ameliorate them.
Collapse
Affiliation(s)
- Mike E Robbins
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Temme S, Bönner F, Schrader J, Flögel U. 19
F magnetic resonance imaging of endogenous macrophages in inflammation. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2012; 4:329-43. [DOI: 10.1002/wnan.1163] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Weise G, Basse-Lüsebrink TC, Kleinschnitz C, Kampf T, Jakob PM, Stoll G. In vivo imaging of stepwise vessel occlusion in cerebral photothrombosis of mice by 19F MRI. PLoS One 2011; 6:e28143. [PMID: 22194810 PMCID: PMC3240623 DOI: 10.1371/journal.pone.0028143] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Accepted: 11/02/2011] [Indexed: 01/20/2023] Open
Abstract
Background 19F magnetic resonance imaging (MRI) was recently introduced as a promising technique for in vivo cell tracking. In the present study we compared 19F MRI with iron-enhanced MRI in mice with photothrombosis (PT) at 7 Tesla. PT represents a model of focal cerebral ischemia exhibiting acute vessel occlusion and delayed neuroinflammation. Methods/Principal Findings Perfluorocarbons (PFC) or superparamagnetic iron oxide particles (SPIO) were injected intravenously at different time points after photothrombotic infarction. While administration of PFC directly after PT induction led to a strong 19F signal throughout the entire lesion, two hours delayed application resulted in a rim-like 19F signal at the outer edge of the lesion. These findings closely resembled the distribution of signal loss on T2-weighted MRI seen after SPIO injection reflecting intravascular accumulation of iron particles trapped in vessel thrombi as confirmed histologically. By sequential administration of two chemically shifted PFC compounds 0 and 2 hours after illumination the different spatial distribution of the 19F markers (infarct core/rim) could be visualized in the same animal. When PFC were applied at day 6 the fluorine marker was only detected after long acquisition times ex vivo. SPIO-enhanced MRI showed slight signal loss in vivo which was much more prominent ex vivo indicative for neuroinflammation at this late lesion stage. Conclusion Our study shows that vessel occlusion can be followed in vivo by 19F and SPIO-enhanced high-field MRI while in vivo imaging of neuroinflammation remains challenging. The timing of contrast agent application was the major determinant of the underlying processes depicted by both imaging techniques. Importantly, sequential application of different PFC compounds allowed depiction of ongoing vessel occlusion from the core to the margin of the ischemic lesions in a single MRI measurement.
Collapse
Affiliation(s)
- Gesa Weise
- Department of Neurology, University of Würzburg, Würzburg, Germany
- Interdisciplinary Center for Clinical Research Würzburg, University of Würzburg, Würzburg, Germany
| | - Thomas C. Basse-Lüsebrink
- Department of Neurology, University of Würzburg, Würzburg, Germany
- Department of Physics, EPV, University of Würzburg, Würzburg, Germany
- Interdisciplinary Center for Clinical Research Würzburg, University of Würzburg, Würzburg, Germany
| | | | - Thomas Kampf
- Department of Physics, EPV, University of Würzburg, Würzburg, Germany
| | - Peter M. Jakob
- Department of Physics, EPV, University of Würzburg, Würzburg, Germany
- Interdisciplinary Center for Clinical Research Würzburg, University of Würzburg, Würzburg, Germany
| | - Guido Stoll
- Department of Neurology, University of Würzburg, Würzburg, Germany
- Interdisciplinary Center for Clinical Research Würzburg, University of Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
47
|
MRI stem cell tracking for therapy in experimental cerebral ischemia. Transl Stroke Res 2011; 3:22-35. [PMID: 24323753 DOI: 10.1007/s12975-011-0111-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/20/2011] [Accepted: 09/28/2011] [Indexed: 12/22/2022]
Abstract
Magnetic resonance has an established role in investigations on the evolution of stroke and the assessment of therapeutic strategies in experimental animals. Here we show that the technique has also an important place for the study of stem cell-mediated regenerative therapies after stroke. We review the literature by bridging from the methodological aspects of stem cell labeling via grafting and monitoring of cell dynamics after implantation into the brain all the way to MRI's role in analyzing the stem cell-mediated functional improvement. Thus, we have aimed at a view combining the focus on the monitoring of the cell activities with the aspect of lesion evolution while including also the essence of a potential functional improvement by the implantation of stem cells following stroke.
Collapse
|
48
|
Vezzani A, Aronica E, Mazarati A, Pittman QJ. Epilepsy and brain inflammation. Exp Neurol 2011; 244:11-21. [PMID: 21985866 DOI: 10.1016/j.expneurol.2011.09.033] [Citation(s) in RCA: 421] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 09/15/2011] [Accepted: 09/26/2011] [Indexed: 02/06/2023]
Abstract
During the last decade, experimental research has demonstrated a prominent role of glial cells, activated in brain by various injuries, in the mechanisms of seizure precipitation and recurrence. In particular, alterations in the phenotype and function of activated astrocytes and microglial cells have been described in experimental and human epileptic tissue, including modifications in potassium and water channels, alterations of glutamine/glutamate cycle, changes in glutamate receptor expression and transporters, release of neuromodulatory molecules (e.g. gliotransmitters, neurotrophic factors), and induction of molecules involved in inflammatory processes (e.g. cytokines, chemokines, prostaglandins, complement factors, cell adhesion molecules) (Seifert et al., 2006; Vezzani et al., 2011; Wetherington et al., 2008). In particular, brain injury or proconvulsant events can activate microglia and astrocytes to release a number of proinflammatory mediators, thus initiating a cascade of inflammatory processes in brain tissue. Proinflammatory molecules can alter neuronal excitability and affect the physiological functions of glia by paracrine or autocrine actions, thus perturbing the glioneuronal communications. In experimental models, these changes contribute to decreasing the threshold to seizures and may compromise neuronal survival (Riazi et al., 2010; Vezzani et al., 2008). In this context, understanding which are the soluble mediators and the molecular mechanisms crucially involved in glio-neuronal interactions is instrumental to shed light on how brain inflammation may contribute to neuronal hyperexcitability in epilepsy. This review will report the clinical observations in drug-resistant human epilepsies and the experimental findings in adult and immature rodents linking brain inflammation to the epileptic process in a causal and reciprocal manner. By confronting the clinical evidence with the experimental findings, we will discuss the role of specific soluble inflammatory mediators in the etiopathogenesis of seizures, reporting evidence for both their acute and long term effects on seizure threshold. The possible contribution of these mediators to co-morbidities often described in epilepsy patients will be also discussed. Finally, we will report on the anti-inflammatory treatments with anticonvulsant actions in experimental models highlighting possible therapeutic options for treating drug-resistant seizures and for prevention of epileptogenesis.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Via G. La Masa 19, 20156 Milano, Italy.
| | | | | | | |
Collapse
|
49
|
Kumata K, Yui J, Hatori A, Fujinaga M, Yanamoto K, Yamasaki T, Kawamura K, Wakizaka H, Nengaki N, Yoshida Y, Ogawa M, Fukumura T, Zhang MR. Synthesis and Evaluation of Novel Carbon-11 Labeled Oxopurine Analogues for Positron Emission Tomography Imaging of Translocator Protein (18 kDa) in Peripheral Organs. J Med Chem 2011; 54:6040-9. [DOI: 10.1021/jm200516a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Katsushi Kumata
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Joji Yui
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Akiko Hatori
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masayuki Fujinaga
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kazuhiko Yanamoto
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Tomoteru Yamasaki
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kazunori Kawamura
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hidekatsu Wakizaka
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Nobuki Nengaki
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- SHI Accelerator Service Co. Ltd., 5-9-11 Kitashinagawa, Shinagawa-ku, Tokyo 141-8686, Japan
| | - Yuichiro Yoshida
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- SHI Accelerator Service Co. Ltd., 5-9-11 Kitashinagawa, Shinagawa-ku, Tokyo 141-8686, Japan
| | - Masanao Ogawa
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- SHI Accelerator Service Co. Ltd., 5-9-11 Kitashinagawa, Shinagawa-ku, Tokyo 141-8686, Japan
| | - Toshimitsu Fukumura
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
50
|
Hohlfeld R, Toyka KV. [Multiple sclerosis: updates on pathogenesis and treatment: report from the 9th MS Symposium held by the German Multiple Sclerosis Society]. DER NERVENARZT 2011; 82:1026-1035. [PMID: 21424411 DOI: 10.1007/s00115-011-3264-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- R Hohlfeld
- Institut für Klinische Neuroimmunologie, Klinikum der LMU München, Campus Großhadern, Marchioninistr. 15, 81377 München, Deutschland.
| | | |
Collapse
|