1
|
Amanat M, Nemeth CL, Fine AS, Leung DG, Fatemi A. Antisense Oligonucleotide Therapy for the Nervous System: From Bench to Bedside with Emphasis on Pediatric Neurology. Pharmaceutics 2022; 14:2389. [PMID: 36365206 PMCID: PMC9695718 DOI: 10.3390/pharmaceutics14112389] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 09/05/2023] Open
Abstract
Antisense oligonucleotides (ASOs) are disease-modifying agents affecting protein-coding and noncoding ribonucleic acids. Depending on the chemical modification and the location of hybridization, ASOs are able to reduce the level of toxic proteins, increase the level of functional protein, or modify the structure of impaired protein to improve function. There are multiple challenges in delivering ASOs to their site of action. Chemical modifications in the phosphodiester bond, nucleotide sugar, and nucleobase can increase structural thermodynamic stability and prevent ASO degradation. Furthermore, different particles, including viral vectors, conjugated peptides, conjugated antibodies, and nanocarriers, may improve ASO delivery. To date, six ASOs have been approved by the US Food and Drug Administration (FDA) in three neurological disorders: spinal muscular atrophy, Duchenne muscular dystrophy, and polyneuropathy caused by hereditary transthyretin amyloidosis. Ongoing preclinical and clinical studies are assessing the safety and efficacy of ASOs in multiple genetic and acquired neurological conditions. The current review provides an update on underlying mechanisms, design, chemical modifications, and delivery of ASOs. The administration of FDA-approved ASOs in neurological disorders is described, and current evidence on the safety and efficacy of ASOs in other neurological conditions, including pediatric neurological disorders, is reviewed.
Collapse
Affiliation(s)
- Man Amanat
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christina L. Nemeth
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amena Smith Fine
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Doris G. Leung
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Genetic Muscle Disorders, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Ohyagi M, Nagata T, Ihara K, Yoshida-Tanaka K, Nishi R, Miyata H, Abe A, Mabuchi Y, Akazawa C, Yokota T. DNA/RNA heteroduplex oligonucleotide technology for regulating lymphocytes in vivo. Nat Commun 2021; 12:7344. [PMID: 34937876 PMCID: PMC8695577 DOI: 10.1038/s41467-021-26902-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 10/19/2021] [Indexed: 11/30/2022] Open
Abstract
Manipulating lymphocyte functions with gene silencing approaches is promising for treating autoimmunity, inflammation, and cancer. Although oligonucleotide therapy has been proven to be successful in treating several conditions, efficient in vivo delivery of oligonucleotide to lymphocyte populations remains a challenge. Here, we demonstrate that intravenous injection of a heteroduplex oligonucleotide (HDO), comprised of an antisense oligonucleotide (ASO) and its complementary RNA conjugated to α-tocopherol, silences lymphocyte endogenous gene expression with higher potency, efficacy, and longer retention time than ASOs. Importantly, reduction of Itga4 by HDO ameliorates symptoms in both adoptive transfer and active experimental autoimmune encephalomyelitis models. Our findings reveal the advantages of HDO with enhanced gene knockdown effect and different delivery mechanisms compared with ASO. Thus, regulation of lymphocyte functions by HDO is a potential therapeutic option for immune-mediated diseases.
Collapse
MESH Headings
- Administration, Intravenous
- Adoptive Transfer
- Animals
- Demyelinating Diseases/genetics
- Demyelinating Diseases/immunology
- Demyelinating Diseases/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endocytosis/drug effects
- Female
- Gene Expression Regulation
- Gene Silencing
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Humans
- Integrin alpha4/genetics
- Integrin alpha4/metabolism
- Jurkat Cells
- Lymphocytes/metabolism
- Male
- Mice, Inbred C57BL
- Nucleic Acid Heteroduplexes/administration & dosage
- Nucleic Acid Heteroduplexes/metabolism
- Nucleic Acid Heteroduplexes/pharmacokinetics
- Nucleic Acid Heteroduplexes/pharmacology
- Oligonucleotides/administration & dosage
- Oligonucleotides/metabolism
- Oligonucleotides/pharmacokinetics
- Oligonucleotides/pharmacology
- RNA/metabolism
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Spinal Cord/pathology
- Tissue Distribution/drug effects
- Mice
Collapse
Affiliation(s)
- Masaki Ohyagi
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuya Nagata
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Kensuke Ihara
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kie Yoshida-Tanaka
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rieko Nishi
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Haruka Miyata
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aya Abe
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chihiro Akazawa
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
3
|
Akkari L, Bowman RL, Tessier J, Klemm F, Handgraaf SM, de Groot M, Quail DF, Tillard L, Gadiot J, Huse JT, Brandsma D, Westerga J, Watts C, Joyce JA. Dynamic changes in glioma macrophage populations after radiotherapy reveal CSF-1R inhibition as a strategy to overcome resistance. Sci Transl Med 2021; 12:12/552/eaaw7843. [PMID: 32669424 DOI: 10.1126/scitranslmed.aaw7843] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 01/31/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAMs) and microglia (MG) are potent regulators of glioma development and progression. However, the dynamic alterations of distinct TAM populations during the course of therapeutic intervention, response, and recurrence have not yet been fully explored. Here, we investigated how radiotherapy changes the relative abundance and phenotypes of brain-resident MG and peripherally recruited monocyte-derived macrophages (MDMs) in glioblastoma. We identified radiation-specific, stage-dependent MG and MDM gene expression signatures in murine gliomas and confirmed altered expression of several genes and proteins in recurrent human glioblastoma. We found that targeting these TAM populations using a colony-stimulating factor-1 receptor (CSF-1R) inhibitor combined with radiotherapy substantially enhanced survival in preclinical models. Our findings reveal the dynamics and plasticity of distinct macrophage populations in the irradiated tumor microenvironment, which has translational relevance for enhancing the efficacy of standard-of-care treatment in gliomas.
Collapse
Affiliation(s)
- Leila Akkari
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland.,Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Robert L Bowman
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jeremy Tessier
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Florian Klemm
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland.,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland
| | - Shanna M Handgraaf
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Marnix de Groot
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Daniela F Quail
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lucie Tillard
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland.,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland
| | - Jules Gadiot
- Tumor Biology and Immunology Division, Netherlands Cancer Institute, Oncode Institute, Amsterdam 1066CX, Netherlands
| | - Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dieta Brandsma
- Departments of Neuro-Oncology and Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam 1066CX, Netherlands
| | - Johan Westerga
- Departments of Neuro-Oncology and Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam 1066CX, Netherlands
| | - Colin Watts
- Birmingham Brain Cancer Program, Institute of Cancer Genome Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland. .,Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland
| |
Collapse
|
4
|
Reduction of integrin alpha 4 activity through splice modulating antisense oligonucleotides. Sci Rep 2019; 9:12994. [PMID: 31506448 PMCID: PMC6736852 DOI: 10.1038/s41598-019-49385-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/19/2019] [Indexed: 12/20/2022] Open
Abstract
With recent approvals of antisense oligonucleotides as therapeutics, there is an increasing interest in expanding the application of these compounds to many other diseases. Our laboratory focuses on developing therapeutic splice modulating antisense oligonucleotides to treat diseases potentially amendable to intervention during pre-mRNA processing, and here we report the use of oligomers to down-regulate integrin alpha 4 protein levels. Over one hundred antisense oligonucleotides were designed to induce skipping of individual exons of the ITGA4 transcript and thereby reducing protein expression. Integrin alpha 4-mediated activities were evaluated in human dermal fibroblasts and Jurkat cells, an immortalised human T lymphocyte cell line. Peptide conjugated phosphorodiamidate morpholino antisense oligomers targeting ITGA4 were also assessed for their effect in delaying disease progression in the experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. With the promising results in ameliorating disease progression, we are optimistic that the candidate oligomer may also be applicable to many other diseases associated with integrin alpha 4 mediated inflammation. This highly specific strategy to down-regulate protein expression through interfering with normal exon selection during pre-mRNA processing should be applicable to many other gene targets that undergo splicing during expression.
Collapse
|
5
|
Evers MM, Toonen LJ, van Roon-Mom WM. Antisense oligonucleotides in therapy for neurodegenerative disorders. Adv Drug Deliv Rev 2015; 87:90-103. [PMID: 25797014 DOI: 10.1016/j.addr.2015.03.008] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 03/02/2015] [Accepted: 03/12/2015] [Indexed: 12/14/2022]
Abstract
Antisense oligonucleotides are synthetic single stranded strings of nucleic acids that bind to RNA and thereby alter or reduce expression of the target RNA. They can not only reduce expression of mutant proteins by breakdown of the targeted transcript, but also restore protein expression or modify proteins through interference with pre-mRNA splicing. There has been a recent revival of interest in the use of antisense oligonucleotides to treat several neurodegenerative disorders using different approaches to prevent disease onset or halt disease progression and the first clinical trials for spinal muscular atrophy and amyotrophic lateral sclerosis showing promising results. For these trials, intrathecal delivery is being used but direct infusion into the brain ventricles and several methods of passing the blood brain barrier after peripheral administration are also under investigation.
Collapse
|
6
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1016] [Impact Index Per Article: 84.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
7
|
Zalachoras I, Evers MM, van Roon-Mom WMC, Aartsma-Rus AM, Meijer OC. Antisense-mediated RNA targeting: versatile and expedient genetic manipulation in the brain. Front Mol Neurosci 2011; 4:10. [PMID: 21811437 PMCID: PMC3142880 DOI: 10.3389/fnmol.2011.00010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 07/08/2011] [Indexed: 12/28/2022] Open
Abstract
A limiting factor in brain research still is the difficulty to evaluate in vivo the role of the increasing number of proteins implicated in neuronal processes. We discuss here the potential of antisense-mediated RNA targeting approaches. We mainly focus on those that manipulate splicing (exon skipping and exon inclusion), but will also briefly discuss mRNA targeting. Classic knockdown of expression by mRNA targeting is only one possible application of antisense oligonucleotides (AON) in the control of gene function. Exon skipping and inclusion are based on the interference of AONs with splicing of pre-mRNAs. These are powerful, specific and particularly versatile techniques, which can be used to circumvent pathogenic mutations, shift splice variant expression, knock down proteins, or to create molecular models using in-frame deletions. Pre-mRNA targeting is currently used both as a research tool, e.g., in models for motor neuron disease, and in clinical trials for Duchenne muscular dystrophy and amyotrophic lateral sclerosis. AONs are particularly promising in relation to brain research, as the modified AONs are taken up extremely fast in neurons and glial cells with a long residence, and without the need for viral vectors or other delivery tools, once inside the blood brain barrier. In this review we cover (1). The principles of antisense-mediated techniques, chemistry, and efficacy. (2) The pros and cons of AON approaches in the brain compared to other techniques of interfering with gene function, such as transgenesis and short hairpin RNAs, in terms of specificity of the manipulation, spatial, and temporal control over gene expression, toxicity, and delivery issues. (3) The potential applications for Neuroscience. We conclude that there is good evidence from animal studies that the central nervous system can be successfully targeted, but the potential of the diverse AON-based approaches appears to be under-recognized.
Collapse
Affiliation(s)
- Ioannis Zalachoras
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research Leiden, Netherlands
| | | | | | | | | |
Collapse
|
8
|
Farooqi N, Gran B, Constantinescu CS. Are current disease-modifying therapeutics in multiple sclerosis justified on the basis of studies in experimental autoimmune encephalomyelitis? J Neurochem 2010; 115:829-44. [DOI: 10.1111/j.1471-4159.2010.06982.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
9
|
Abstract
Antisense oligonucleotides as a therapeutic platform have been slow to progress since the approval of the first antisense drug in 1998. Recently, there have been several examples of convincing antisense interventions in animal models and promising clinical trial data. This review considers the factors determining the success of antisense oligonucleotides as therapeutic agents. In order to produce target knockdown after systemic delivery, antisense oligonucleotides must avoid nuclease degradation, reticuloendothelial-system uptake and rapid renal excretion, and extravasate to the target cell type outside the vasculature. They then must enter the target cell, and escape the endosome-lysosome pathway so as to be free to interact with the target mRNA. We consider the significance of these limiting factors based on the literature and our own experience using systemic administration of antisense oligonucleotides.
Collapse
|
10
|
Leaman DW. Recent progress in oligonucleotide therapeutics: antisense to aptamers. Expert Opin Drug Discov 2008; 3:997-1009. [DOI: 10.1517/17460441.3.9.997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
|
12
|
Actively induced EAE in Lewis rats: characterization of spleen and spinal cord infiltrating lymphocytes by flow cytometry during the course of the disease. J Neuroimmunol 2008; 199:67-74. [PMID: 18572253 DOI: 10.1016/j.jneuroim.2008.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 05/15/2008] [Accepted: 05/16/2008] [Indexed: 11/21/2022]
Abstract
Actively induced Lewis rat Experimental Autoimmune Encephalomyelitis (EAE) is a highly reproducible model for portraying the acute phase of multiple sclerosis. Our aim was to get more information about this model by means of flow cytometry looking at potential markers for tracing new treatments' efficacy. Thus we characterized the changes occurring in encephalitogenic TCR Vbeta8.2(+) frequency and the adhesion molecule alpha4 integrin expression in both spleen and spinal cord T cells. The increase in both these parameters was observed only in spinal cord infiltrating T cells while relevant changes in spleen cell composition were observed as early as disease onset.
Collapse
|
13
|
Tsunoda I, Terry EJ, Marble BJ, Lazarides E, Woods C, Fujinami RS. Modulation of experimental autoimmune encephalomyelitis by VLA-2 blockade. Brain Pathol 2007; 17:45-55. [PMID: 17493037 PMCID: PMC8095550 DOI: 10.1111/j.1750-3639.2006.00042.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Adhesion molecules play important roles in cell-cell and cell-extracellular matrix (ECM) interactions in inflammation. Blocking the interaction between inflammatory cells and vascular endothelia can prevent cell entry into tissues and harmful inflammatory responses, that is, autoimmunity, but could also limit immunosurveillance by anti-viral T cells in sites of infection or latency. Development of progressive multifocal leukoencephalopathy in patients treated with antibody against very late antigen (VLA)-4 prompted us to explore an alternative therapeutic approach. We used an antibody against the integrin alpha2, VLA-2, that interacts with ECM, not vascular endothelium. SJL/J mice were sensitized with myelin proteolipid protein (PLP)(139-151) peptide to induce experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Treatment of mice with VLA-2 antibody suppressed clinical signs and CNS inflammation of EAE, when antibody was given immediately after disease onset. In contrast, VLA-4 or VLA-2 antibody treatment of mice during the priming or remission phase of EAE had minor effects on the disease's clinical course. No differences were found in lymphoproliferative responses to PLP(139-151) among treatment groups. Data suggest that blocking cell-ECM interactions can be an alternative therapy for MS.
Collapse
Affiliation(s)
- Ikuo Tsunoda
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Emily Jane Terry
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Benjamin J. Marble
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Elias Lazarides
- (Formerly of Targeted Molecules Corporation) Chromos Molecular Systems, Burnaby, British Columbia, Canada
| | - Catherine Woods
- (Formerly of Targeted Molecules Corporation) Chromos Molecular Systems, Burnaby, British Columbia, Canada
| | - Robert S. Fujinami
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
14
|
Kleiter I, Pedré X, Mueller AM, Poeschl P, Couillard-Despres S, Spruss T, Bogdahn U, Giegerich G, Steinbrecher A. Inhibition of Smad7, a negative regulator of TGF-beta signaling, suppresses autoimmune encephalomyelitis. J Neuroimmunol 2007; 187:61-73. [PMID: 17553571 DOI: 10.1016/j.jneuroim.2007.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 04/12/2007] [Indexed: 02/07/2023]
Abstract
We studied the role of the Transforming growth factor (TGF)-beta signaling antagonist Smad7 in autoimmune central nervous system (CNS) inflammation by using specific antisense oligonucleotides (Smad7-as). Elevated Smad7 protein expression was found in the spinal cord of SJL/J mice and DA rats with experimental autoimmune encephalomyelitis (EAE) and in effector T cells upon antigen stimulation. Smad7-as specifically decreased Smad7 mRNA and protein in cell lines and in ex-vivo-treated primary mouse lymph node cells (LNC). LNC exposed to Smad7-as during secondary activation showed reduced proliferation and encephalitogenicity. After systemic administration, Smad7-as ameliorated clinical signs of active and adoptively transferred EAE, diminished CNS inflammation, and reduced Smad7 protein levels in the brain. Smad7-as was found to be incorporated by peritoneal macrophages as well as by cells of the liver, kidneys, and peripheral lymph nodes. Importantly, Smad7-as treatment was not toxic and did not increase extracellular matrix formation. Smad7 inhibition thus represents a novel systemic treatment strategy for autoimmune CNS inflammation, targeting TGF-beta signaling without TGF-beta-associated toxicity.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Animals
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Gene Expression Regulation/drug effects
- Lymphocytes/drug effects
- Lymphocytes/metabolism
- Mice
- Mice, Inbred Strains
- Oligonucleotides, Antisense/pharmacology
- Oligonucleotides, Antisense/therapeutic use
- RNA, Messenger/biosynthesis
- Rats
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Smad7 Protein/chemistry
- Smad7 Protein/metabolism
- Thionucleotides/pharmacology
- Thionucleotides/therapeutic use
- Time Factors
- Transforming Growth Factor beta/physiology
Collapse
Affiliation(s)
- Ingo Kleiter
- Department of Neurology, University of Regensburg, Universitaetsstr. 84, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Adams JE, Webb MS, Hu J, Staunton D, Barnum SR. Disruption of the beta2-integrin CD11d (alphaDbeta2) gene fails to protect against experimental autoimmune encephalomyelitis. J Neuroimmunol 2007; 184:180-7. [PMID: 17254640 PMCID: PMC2747331 DOI: 10.1016/j.jneuroim.2006.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 12/05/2006] [Accepted: 12/15/2006] [Indexed: 12/17/2022]
Abstract
The fourth member of the beta(2)-integrin family of adhesion molecules, CD11d (alpha(D)beta(2)), is expressed on a wide variety of immune cells, however its function in autoimmune diseases, including EAE remains unknown. We induced EAE in wild-type and CD11d(-/-) C57BL/6 mice using myelin oligodendrocyte glycoprotein (MOG(35-55)) peptide. The clinical course and histopathology of EAE were identical in both groups of mice throughout the disease course. There were no significant differences in the infiltration of leukocyte subsets into the central nervous system or in the production of cytokines from T cells isolated from the spleen or spinal cord from both groups of mice. Our data demonstrate that CD11d is not required for the development of EAE and, to date, is the only beta(2)-integrin molecule whose deletion does not result in attenuated disease.
Collapse
MESH Headings
- Animals
- CD18 Antigens/genetics
- CD18 Antigens/metabolism
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Flow Cytometry/methods
- Glycoproteins/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/pharmacology
- Spinal Cord/pathology
- Spleen/pathology
- Statistics, Nonparametric
- T-Lymphocytes/metabolism
- Time Factors
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Jillian E. Adams
- Department of Microbiology, University of Alabama at Birmingham, 845 19 St. S., BBRB/842, Birmingham, AL, 35294 USA
| | - Matthew S. Webb
- Department of Microbiology, University of Alabama at Birmingham, 845 19 St. S., BBRB/842, Birmingham, AL, 35294 USA
| | - Jane Hu
- Department of Microbiology, University of Alabama at Birmingham, 845 19 St. S., BBRB/842, Birmingham, AL, 35294 USA
| | - Don Staunton
- ICOS Pharmaceuticals, 22021 20 Ave. SE Bothell, WA, 98021 USA
| | - Scott R. Barnum
- Department of Microbiology, University of Alabama at Birmingham, 845 19 St. S., BBRB/842, Birmingham, AL, 35294 USA
- Department of Neurology, University of Alabama at Birmingham, 845 19 St. S., BBRB/842, Birmingham, AL, 35294 USA
| |
Collapse
|
16
|
Wacheck V. [Oligonucleotide therapeutics - an emerging novel class of compounds]. Wien Med Wochenschr 2007; 156:481-7. [PMID: 17041803 DOI: 10.1007/s10354-006-0331-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Accepted: 05/02/2006] [Indexed: 12/14/2022]
Abstract
Oligonucleotide therapeutics are short, single- or double-stranded DNA or RNA molecules consisting of strands of 10-50 nucleotides. By targeted modulation of gene expression oligonucleotides provide the chance of targeting diseases at their molecular level. Within this novel emerging class of compounds oligonucleotide therapeutics are discriminated by their structure, function and mode of action. While antisense oligonucleotides, ribozymes and siRNAs suppress the expression of a protein by complementary hybridizing with their target mRNA, aptamers bind like antibodies to their target protein and thereby inhibit its function. Immunostimulatory oligonucleotides are due to sequence motifs within their nucleotide sequence able to trigger a therapeutic exploitable immune response. Currently, there are only two oligonucleotide therapeutics approved by the FDA, namely the antisense oligonucleotide Fomivirsen and the aptamer Macugen. In this review the mode of action of the diverse oligonucleotide therapeutics and their current status in clinical development will be discussed.
Collapse
Affiliation(s)
- Volker Wacheck
- Abteilung für Experimentelle Onkologie/Molekulare Pharmakologie, Universitätsklinik für Klinische Pharmakologie, Medizinische Universität Wien, Wien, Austria.
| |
Collapse
|
17
|
Engelhardt B. Immune cell migration across the blood–brain barrier: molecular mechanisms and therapeutic targeting. FUTURE NEUROLOGY 2006. [DOI: 10.2217/14796708.1.1.47] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The endothelial blood–brain barrier (BBB) and the epithelial blood–cerebrospinal fluid barrier protect the CNS from the constantly changing milieu within the bloodstream. The BBB strictly controls immune cell entry into the CNS, which is rare under physiological conditions. During a variety of pathological conditions of the CNS, such as viral or bacterial infections, or during inflammatory diseases, such as multiple sclerosis, immunocompetent cells readily traverse the BBB and subsequently enter the CNS parenchyma. Most of the available information on immune cell entry into the CNS is derived from studying experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. Consequently, our current knowledge on traffic signals mediating immune cell entry across the BBB during immunosurveillance and disease results mainly from experimental data in the EAE model. Therefore, a large part of this review summarizes these findings. Similarly, the potential benefits and risks associated with therapeutic targeting of immune cell trafficking across the BBB will be discussed in the context of multiple sclerosis, since elucidation of the molecular mechanisms relevant to this disease have largely relied on the use of its in vivo model, EAE.
Collapse
Affiliation(s)
- Britta Engelhardt
- Theodore Kocher Institute, University of Bern, Freiestr. 1, CH-3012 Switzerland
| |
Collapse
|