Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 339(2): 519-29, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21807883

ABSTRACT

LINGO-1 (leucine-rich repeat and Ig domain containing NOGO receptor interacting protein-1) is a negative regulator of myelination and repair of damaged axons in the central nervous system (CNS). Blocking LINGO-1 function leads to robust remyelination. The anti-LINGO-1 Li81 antibody is currently being evaluated in clinical trials for multiple sclerosis (MS) and is the first MS therapy that directly targets myelin repair. LINGO-1 is selectively expressed in brain and spinal cord but not in peripheral tissues. Perhaps the greatest concern for Li81 therapy is the limited access of the drug to the CNS. Here, we measured Li81 concentrations in brain, spinal cord, and cerebral spinal fluid in rats after systemic administration and correlated them with dose-efficacy responses in rat lysolecithin and experimental autoimmune encephalomyelitis spinal cord models of remyelination. Remyelination was dose-dependent, and levels of Li81 in spinal cord that promoted myelination correlated well with affinity measurements for the binding of Li81 to LINGO-1. Observed Li81 concentrations in the CNS of 0.1 to 0.4% of blood levels are consistent with values reported for other antibodies. To understand the features of the antibody that affect CNS penetration, we also evaluated the pharmacokinetics of Li81 Fab2, Fab, and poly(ethylene glycol)-modified Fab. The reagents all showed similar CNS exposure despite large differences in their sizes, serum half-lives, and volumes of distribution, and area under the curve (AUC) measurements in the CNS directly correlated with AUC measurements in serum. These studies demonstrate that exposure levels achieved by passive diffusion of the Li81 monoclonal antibody into the CNS are sufficient and lead to robust remyelination.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/immunology , Spinal Cord/drug effects , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Cerebrospinal Fluid/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Lysophosphatidylcholines , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Regeneration , Spinal Cord/metabolism , Spinal Cord/pathology
2.
Mol Cell Neurosci ; 48(1): 72-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21740973

ABSTRACT

The sphingosine 1-phosphate (S1P) receptor modulators have emerged as a new therapeutic opportunity paradigm for the treatment of immune-mediated demyelinating diseases such as multiple sclerosis (MS). The S1P analog fingolimod (FTY720) has been shown to alleviate disease burden in immune-mediated animal models of MS, and has been approved for treatment in clinical trials in patients with MS in the United States. While the immunological effects of FTY720 are well established, there is controversy in the literature regarding the contribution of FTY720 on myelin repair. Here, we directly assessed the impact of FTY720 on myelin repair in cuprizone and lysolecithin (LPC) demyelination models that have a minimal immunological component. FTY720 failed to promote remyelination in either animal model. These studies suggest that while FTY720 may be effective at modulating the immunological attack in MS, it may benefit from an add-on therapy to enhance the myelin repair required for long-term functional restoration in MS.


Subject(s)
Immunosuppressive Agents/pharmacology , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Regeneration/drug effects , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Chelating Agents/pharmacology , Cuprizone/pharmacology , Demyelinating Diseases/drug therapy , Disease Models, Animal , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred C57BL , Propylene Glycols/therapeutic use , Rats , Rats, Sprague-Dawley , Sphingosine/pharmacology , Sphingosine/therapeutic use
3.
Nat Med ; 17(7): 816-21, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725297

ABSTRACT

Survival and differentiation of oligodendrocytes are important for the myelination of central nervous system (CNS) axons during development and crucial for myelin repair in CNS demyelinating diseases such as multiple sclerosis. Here we show that death receptor 6 (DR6) is a negative regulator of oligodendrocyte maturation. DR6 is expressed strongly in immature oligodendrocytes and weakly in mature myelin basic protein (MBP)-positive oligodendrocytes. Overexpression of DR6 in oligodendrocytes leads to caspase 3 (casp3) activation and cell death. Attenuation of DR6 function leads to enhanced oligodendrocyte maturation, myelination and downregulation of casp3. Treatment with a DR6 antagonist antibody promotes remyelination in both lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE) models. Consistent with the DR6 antagoinst antibody studies, DR6-null mice show enhanced remyelination in both demyelination models. These studies reveal a pivotal role for DR6 signaling in immature oligodendrocyte maturation and myelination that may provide new therapeutic avenues for the treatment of demyelination disorders such as multiple sclerosis.


Subject(s)
Myelin Sheath/metabolism , Oligodendroglia/metabolism , Receptors, Tumor Necrosis Factor/physiology , Animals , Blotting, Western , Caspase 3/metabolism , Caspase 3/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Survival/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Enzyme Activation , Gene Expression Regulation , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/therapy , Myelin Sheath/physiology , Oligodendroglia/physiology , Rats , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction
4.
Bioconjug Chem ; 22(2): 200-10, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21254764

ABSTRACT

The use of LINGO-1 antagonists to promote repair of damaged myelin is an emerging therapeutic opportunity for treatment of CNS diseases caused by demyelination such as multiple sclerosis. The Li33 anti-LINGO-1 antibody is a potent inducer of myelination in vitro and in vivo, but aggregation issues prevented the engineering of an optimal development candidate. PEGylated Li33 Fab' is one of several versions of the Li33 antibody that is being investigated in an attempt to identify the most favorable anti-LINGO-1 antibody design. For targeted PEGylation, a Li33 Fab' construct was engineered with a single unpaired cysteine in the heavy-chain hinge sequence. The Fab' was expressed in CHO cells, purified, and PEGylated with 20 kDa methoxy-poly(ethylene glycol) maleimide using a reaction strategy optimized to improve the yield of the PEG-Fab'. Biochemical analysis of the Li33 PEG-Fab' verified the selectivity of the PEGylation reaction. The in vitro and in vivo attributes of the PEG-Fab' were benchmarked against a Li33 full antibody. Both the Li33 PEG-Fab' and intact antibody bound LINGO-1 with nanomolar affinity, promoted myelination in an in vitro signaling assay, and promoted the repair of damaged myelin in the rat lysolecithin model. These studies extend our understanding of the biological activity of the Li33 mAb and validate the use of an anti-LINGO-1 PEG-Fab' for treatment of CNS diseases caused by demyelination.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin Fab Fragments/chemistry , Polyethylene Glycols/chemistry , Animals , Antibodies, Monoclonal/immunology , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/isolation & purification , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Models, Animal , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Rats
5.
Ann Neurol ; 65(3): 304-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19334062

ABSTRACT

OBJECTIVE: Repair of demyelinated axons in diseases such as multiple sclerosis requires activation of the myelination program in existing or newly recruited oligodendrocyte precursor cells (OPCs). The control of OPC differentiation and initiation of myelination during repair is poorly understood. In this study, we test the ability of anti-LINGO-1 reagents to promote myelination in vitro and remyelination in the rodent adult central nervous system in vivo. METHODS: The effects of LINGO-1 antagonists on the differentiation of OPCs and the promotion of myelination has been assayed using a combination of coculture and slice culture preparations. Using three different animal models of demyelination and remyelination, we morphologically and functionally assessed the effects of LINGO-1 antagonists on OPC differentiation and myelin repair. RESULTS: The data indicate that in vitro treatment with antagonists of LINGO-1 promote OPC differentiation and myelination, whereas in vivo remyelination is accelerated in lysophosphatidylcholine- or cuprizone-induced demyelination. This remyelination is associated with enhanced OPC differentiation and functional recovery of conduction velocities in demyelinated axons. INTERPRETATION: Our studies demonstrate that LINGO-1 antagonism promotes OPC differentiation and remyelination, and suggest LINGO-1 functions as an inhibitor of OPC differentiation to retard central nervous system remyelination.


Subject(s)
Cell Differentiation/physiology , Demyelinating Autoimmune Diseases, CNS/physiopathology , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Animals, Newborn , Antibodies/pharmacology , Antibodies/therapeutic use , Cell Differentiation/drug effects , Cells, Cultured , Cuprizone/toxicity , Demyelinating Autoimmune Diseases, CNS/chemically induced , Demyelinating Autoimmune Diseases, CNS/drug therapy , Demyelinating Autoimmune Diseases, CNS/pathology , Disease Models, Animal , Ganglia, Spinal/cytology , Lysophosphatidylcholines/toxicity , Membrane Proteins/immunology , Membrane Proteins/physiology , Mice , Myelin Proteins/metabolism , Myelin Sheath/drug effects , Myelin Sheath/physiology , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/physiology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects
6.
Mol Cell Neurosci ; 39(2): 258-67, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18692574

ABSTRACT

Oligodendrocyte-myelin glycoprotein (OMgp) is a myelin component that has been shown in vitro to inhibit neurite outgrowth by binding to the Nogo-66 receptor (NgR1)/Lingo-1/Taj (TROY)/p75 receptor complex to activate the RhoA pathway. To investigate the effects of OMgp on axon regeneration in vivo, OMgp(-/-) mice on a mixed 129/Sv/C57BL/6 (129BL6) or a C57BL/6 (BL6) genetic background were tested in two spinal cord injury (SCI) models - a severe complete transection or a milder dorsal hemisection. OMgp(-/-) mice on the mixed 129BL6 genetic background showed greater functional improvement compared to OMgp(+/+) littermates, with increased numbers of cholera toxin B-labeled ascending sensory axons and 5-HT(+) descending axons and less RhoA activation after spinal cord injury. Myelin isolated from OMgp(-/-) mice (129BL6) was significantly less inhibitory to neurite outgrowth than wild-type (wt) myelin in vitro. However, OMgp(-/-) mice on a BL/6 genetic background showed neither statistically significant functional recovery nor axonal sprouting following dorsal hemisection.


Subject(s)
Axons/physiology , Myelin-Associated Glycoprotein/deficiency , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Analysis of Variance , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Cholera Toxin/metabolism , Dextrans/metabolism , Exploratory Behavior/physiology , Female , Functional Laterality/genetics , GPI-Linked Proteins , Ganglia, Spinal/pathology , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Neurites/physiology , Neurons/pathology , Recovery of Function/genetics , Serotonin/metabolism , Time Factors , rhoA GTP-Binding Protein/metabolism
7.
Mol Cell Neurosci ; 33(3): 311-20, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17011208

ABSTRACT

LINGO-1 is a CNS-specific protein and a functional component of the NgR1/p75/LINGO-1 and NgR1/TAJ(TROY)/LINGO-1 signaling complexes that mediate inhibition of axonal outgrowth. These receptor complexes mediate the axonal growth inhibitory effects of Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte-myelin glycoprotein (OMgp) via RhoA activation. Soluble LINGO-1 (LINGO-1-Fc), which acts as an antagonist of these pathways by blocking LINGO-1 binding to NgR1, was administered to rats after dorsal or lateral hemisection of the spinal cord. LINGO-1-Fc treatment significantly improved functional recovery, promoted axonal sprouting and decreased RhoA activation and increased oligodendrocyte and neuronal survival after either rubrospinal or corticospinal tract transection. These experiments demonstrate an important role for LINGO-1 in modulating axonal outgrowth in vivo and that treatment with LINGO-1-Fc can significantly enhance recovery after spinal cord injury.


Subject(s)
Axons/drug effects , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Analysis of Variance , Animals , Apoptosis/drug effects , Axons/physiology , Caspase 3/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Forelimb/drug effects , Humans , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , MAP Kinase Kinase 4/metabolism , Membrane Proteins/chemistry , Membrane Proteins/physiology , Nerve Regeneration/drug effects , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Organogenesis/drug effects , Protein Binding/drug effects , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Time Factors , Tubulin/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Eur J Neurosci ; 22(3): 587-94, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16101740

ABSTRACT

Methylprednisolone (MP) is a synthetic glucocorticoid used for the treatment of spinal cord injury (SCI). Soluble Nogo-66 receptor (NgR) ectodomain is a novel experimental therapy for SCI that promotes axonal regeneration by blocking the growth inhibitory effects of myelin constituents in the adult central nervous system. To evaluate the potential complementarity of these mechanistically distinct pharmacological reagents we compared their effects alone and in combination after thoracic (T7) dorsal hemisection in the rat. Treatment with an ecto-domain of the rat NgR (27-310) fused to a rat IgG [NgR(310)ecto-Fc] (50 microm intrathecal, 0.25 microL/h for 28 days) or MP alone (30 mg/kg i.v., 0, 4 and 8 h postinjury) improved the rate and extent of functional recovery measured using Basso, Beattie, Bresnahan (BBB) scoring and footprint analysis. The effect of MP treatment on BBB score was apparent the day after SCI whereas the effect of NgR(310)ecto-Fc was not apparent until 2 weeks after SCI. NgR(310)ecto-Fc or MP treatment resulted in increased axonal sprouting and/or regeneration, quantified by counting biotin dextran amine-labeled corticospinal tract axons, and increased the number of axons contacting motor neurons in the ventral horn gray matter caudal to the lesion. Combined treatment with NgR(310)ecto-Fc and MP had a more pronounced effect on recovery of function and axonal growth compared with either treatment alone. The data demonstrate that NgR(310)ecto-Fc and MP act in a temporally and mechanistically distinct manner and suggest that they may have complementary effects.


Subject(s)
Methylprednisolone/therapeutic use , Receptors, Peptide/therapeutic use , Spinal Cord Injuries/drug therapy , Analysis of Variance , Animals , Axons/drug effects , Axons/physiology , Behavior, Animal , Biotin/analogs & derivatives , Biotin/metabolism , Cells, Cultured , Chick Embryo , Dextrans/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Drug Therapy, Combination , Exploratory Behavior/drug effects , Female , GPI-Linked Proteins , Ganglia, Spinal/cytology , Immunoglobulin G/therapeutic use , Laminectomy/methods , Myelin Proteins , Myelin Sheath/metabolism , Nerve Regeneration/drug effects , Neurons/drug effects , Neurons/physiology , Nogo Receptor 1 , Pyramidal Tracts/drug effects , Pyramidal Tracts/metabolism , Rats , Rats, Long-Evans , Receptors, Cell Surface , Receptors, Peptide/biosynthesis , Receptors, Peptide/chemistry , Receptors, Peptide/immunology , Recombinant Proteins/therapeutic use , Recovery of Function/drug effects , Spinal Cord Injuries/physiopathology
9.
J Neurosci ; 24(46): 10511-20, 2004 Nov 17.
Article in English | MEDLINE | ID: mdl-15548666

ABSTRACT

The growth of injured axons in the adult mammalian CNS is limited after injury. Three myelin proteins, Nogo, MAG (myelin-associated glycoprotein), and OMgp (oligodendrocyte myelin glycoprotein), bind to the Nogo-66 receptor (NgR) and inhibit axonal growth in vitro. Transgenic or viral blockade of NgR function allows axonal sprouting in vivo. Here, we administered the soluble function-blocking NgR ectodomain [aa 27-310; NgR(310)ecto] to spinal-injured rats. Purified NgR(310)ecto-Fc protein was delivered intrathecally after midthoracic dorsal over-hemisection. Axonal sprouting of corticospinal and raphespinal fibers in NgR(310)ecto-Fc-treated animals correlates with improved spinal cord electrical conduction and improved locomotion. The ability of soluble NgR(310)ecto to promote axon growth and locomotor recovery demonstrates a therapeutic potential for NgR antagonism in traumatic spinal cord injury.


Subject(s)
Axons/physiology , Myelin Proteins/antagonists & inhibitors , Myelin-Associated Glycoprotein/antagonists & inhibitors , Myelin-Associated Glycoprotein/metabolism , Receptors, Peptide/physiology , Spinal Cord Injuries/pathology , Animals , Axons/metabolism , Evoked Potentials, Motor , Female , GPI-Linked Proteins , Injections, Spinal , Motor Activity , Myelin-Oligodendrocyte Glycoprotein , Nogo Proteins , Nogo Receptor 1 , Oligodendroglia/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Serotonin/metabolism , Solubility , Spinal Cord/physiopathology , Spinal Cord/ultrastructure , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
10.
Nat Neurosci ; 7(3): 221-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14966521

ABSTRACT

Axon regeneration in the adult CNS is prevented by inhibitors in myelin. These inhibitors seem to modulate RhoA activity by binding to a receptor complex comprising a ligand-binding subunit (the Nogo-66 receptor NgR1) and a signal transducing subunit (the neurotrophin receptor p75). However, in reconstituted non-neuronal systems, NgR1 and p75 together are unable to activate RhoA, suggesting that additional components of the receptor may exist. Here we describe LINGO-1, a nervous system-specific transmembrane protein that binds NgR1 and p75 and that is an additional functional component of the NgR1/p75 signaling complex. In non-neuronal cells, coexpression of human NgR1, p75 and LINGO-1 conferred responsiveness to oligodendrocyte myelin glycoprotein, as measured by RhoA activation. A dominant-negative human LINGO-1 construct attenuated myelin inhibition in transfected primary neuronal cultures. This effect on neurons was mimicked using an exogenously added human LINGO-1-Fc fusion protein. Together these observations suggest that LINGO-1 has an important role in CNS biology.


Subject(s)
Membrane Proteins/metabolism , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Regeneration/physiology , Receptors, Cell Surface/metabolism , Receptors, Nerve Growth Factor/metabolism , Amino Acid Sequence/genetics , Animals , Animals, Newborn , Astrocytes/metabolism , Axons/metabolism , Base Sequence/genetics , Cells, Cultured , DNA, Complementary/analysis , DNA, Complementary/genetics , Fetus , GPI-Linked Proteins , Humans , Macromolecular Substances , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Molecular Sequence Data , Mutation/genetics , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/isolation & purification , Myelin-Oligodendrocyte Glycoprotein , Nerve Tissue Proteins , Nogo Receptor 1 , Protein Structure, Tertiary/genetics , Rats , Receptor, Nerve Growth Factor , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...