Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Alzheimers Res Ther ; 13(1): 32, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504364

ABSTRACT

BACKGROUND: Prominent activation of microglial immune/inflammatory processes is a characteristic feature of brains of patients with tauopathies including Alzheimer's disease (AD), suggesting that neuroinflammation may be a critical factor in their pathogenesis. Strategies aimed at developing new therapeutics for tauopathies based on anti-inflammation or immunomodulation are likely to be promising avenues of research. We previously developed JM4-a 19'mer cyclic peptide derived from the first loop of human erythropoietin. This peptide possesses beneficial immune modulatory and tissue protective effects while lacking the undesirable side effects of full-length erythropoietin. In this preclinical study, we investigated the effect of chronic JM4 treatment on the PS19 mouse that carries the P301S mutant human tau gene, linked to a form of frontotemporal dementia. This transgenic mouse has been widely used as a model of tauopathies including AD and related dementias. METHODS: Daily subcutaneous treatment of female PS19 mice with JM4 was initiated before disease onset and continued on for the animals' lifespan. The progression of neurological deficit and the lifespan of these mice were assessed. To evaluate the effect of JM4 treatment on cognition of these animals, the PS19 mice underwent Barnes maze test and elevated plus maze test. In addition, neuronal loss, phosphorylated tau aggregation, and microglial activation were assessed using immunohistochemistry of PS19 mouse brain sections. RESULTS: JM4 treatment of PS19 mice initiated before disease onset reduced neurological deficit, prolonged lifespan, and rescued memory impairment. The beneficial effects of JM4 were accompanied by reductions in neuronal loss, phosphorylated tau aggregation, and microglial activation in the PS19 mouse brain. LIMITATIONS: Use of a single dose of JM4 and female mice only. CONCLUSION: JM4 is a potential novel therapeutic agent for the treatment of tauopathies including AD and related dementias.


Subject(s)
Erythropoietin , Tauopathies , Animals , Brain/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Tauopathies/drug therapy , tau Proteins/genetics , tau Proteins/metabolism
2.
Neurotherapeutics ; 18(1): 401-411, 2021 01.
Article in English | MEDLINE | ID: mdl-32959273

ABSTRACT

Potent beneficial immunomodulatory and anti-inflammatory effects of whole-molecule erythropoietin have been demonstrated in a variety of animal disease models including experimental autoimmune encephalomyelitis (EAE); however, excessive hematopoiesis limits its use in clinical applications. Our group previously generated an Epo-derived small peptide JM4 that is side-effect free and has strong neuroprotective activity without hematologic effects. Here, we investigated the long-term clinical effects of brief treatment with JM4 in chronic relapsing EAE using bioluminescence imaging (BLI) in transgenic mice containing the luciferase gene driven by the murine GFAP promoter. EAE mice treated with JM4 exhibited marked improvement in clinical scores and showed fewer disease flareups than control animals. JM4 therapy concomitantly led to markedly decreased GFAP bioluminescence in the brain and spinal cord in both acute and chronic relapsing EAE mouse models. We found a marker for toxic A1 astrocytes, complement component C3, that is upregulated in the brain and cord of EAE mice and sharply reduced in JM4-treated animals. In addition, an abnormally leaky neurovascular unit permeability was rapidly normalized within 5 days by JM4 therapy. The prolonged therapeutic benefit seen following brief JM4 treatment in EAE mice closely resemble that recently described in humans receiving pulsed immune reconstitution therapy with the disease-modifying compounds, alemtuzumab and cladribine. Our study suggests that JM4 therapy may have widespread clinical applicability for long-term treatment of inflammatory demyelinating diseases and that BLI is a useful noninvasive means of monitoring murine disease activity of the central nervous system.


Subject(s)
Erythropoietin/therapeutic use , Peptide Fragments/therapeutic use , Animals , Brain/drug effects , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Luminescent Measurements , Male , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Spinal Cord/drug effects , Spinal Cord/pathology
3.
Brain Behav ; 11(1): e01920, 2021 01.
Article in English | MEDLINE | ID: mdl-33300690

ABSTRACT

BACKGROUND AND PURPOSE: Canine distemper virus (CDV) is a candidate agent in the etiology of multiple sclerosis (MS). Elevated anti-CDV levels were previously found in the sera from MS patients compared with controls. We now investigated whether there was an age-related association with the presence of antibodies specific to CDV-hemagglutinin (H) protein in MS. METHODS: Sera from patients with MS, other neurological diseases, and inflammatory and/or autoimmune diseases, and healthy individuals were screened for anti-CDV in an ELISA using linear peptides of the CDV-H protein as antigen. Antibody levels to measles and varicella-zoster virus were measured and served as controls. RESULTS: Analysis of the new cohort of MS patients and controls confirmed our initial finding of elevated anti-CDV-H levels in MS patients. An increase in measles but not varicella-zoster virus antibody levels was found in MS patients compared with healthy controls. Data from the new cohort of patients and controls were combined with data from the original study and analyzed with respect to age distribution of anti-CDV IgG. Mean CDV antibody levels were significantly elevated in each decade from 20 to 50 years of age in MS compared with healthy and disease controls. Antibody levels to measles virus were not consistently elevated during this age span. A striking relationship (p < .0001, odds ratio = 5.0) was observed between elevated anti-CDV-H levels and diagnosis of MS. CONCLUSIONS: The finding that anti-CDV levels are elevated in MS patients of all ages provides substantial evidence of a strong association between elevated anti-CDV and MS.


Subject(s)
Distemper Virus, Canine , Distemper , Multiple Sclerosis , Animals , Antibodies, Viral , Dogs , Enzyme-Linked Immunosorbent Assay , Humans , Measles virus
4.
Front Aging Neurosci ; 11: 252, 2019.
Article in English | MEDLINE | ID: mdl-31572168

ABSTRACT

Many studies of tauopathy use transgenic mice that overexpress the P301S mutant form of tau. Neuronal damage in these mice is associated with astrogliosis and induction of glial fibrillary acidic protein (GFAP) expression. GFAP-luc transgenic mice express firefly luciferase under the GFAP promoter, allowing bioluminescence to be measured non-invasively as a surrogate biomarker for astrogliosis. We bred double transgenic mice possessing both P301S and GFAP-luc cassettes and compared them to control mice bearing only the GFAP-luc transgene. We used serial bioluminescent images to define the onset and the time course of astrogliosis in these mice and this was correlated with the development of clinical deficit. Mice containing both GFAP-luc and P301S transgenes showed increased luminescence indicative of astroglial activation in the brain and spinal cord. Starting at 5 months old, the onset of clinical deterioration in these mice corresponded closely to the initial rise in the luminescent signal. Post mortem analysis showed the elevated luminescence was correlated with hyperphosphorylated tau deposition in the hippocampus of double transgenic mice. We used this method to determine the therapeutic effect of JM4 peptide [a small peptide immunomodulatory agent derived from human erythropoietin (EPO)] on double transgenic mice. JM4 treatment significantly decreased the intensity of luminescence, neurological deficit and hyperphosphorylated tau in mice with both the P301S and GFAP-luc transgenes. These findings indicate that bioluminescence imaging (BLI) is a powerful tool for quantifying GFAP expression in living P301S mice and can be used as a noninvasive biomarker of tau-induced neurodegeneration in preclinical therapeutic trials.

5.
Mult Scler Relat Disord ; 34: 164, 2019 09.
Article in English | MEDLINE | ID: mdl-31543259
6.
Neurotherapeutics ; 13(2): 418-27, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26715414

ABSTRACT

There is currently no effective medical treatment for traumatic brain injury (TBI). Beyond the immediate physical damage caused by the initial impact, additional damage evolves due to the inflammatory response that follows brain injury. Here we show that therapy with JM4, a low molecular weight 19-amino acid nonhematopoietic erythropoietin (EPO) peptidyl fragment, containing amino acids 28-46 derived from the first loop of EPO, markedly reduces acute brain injury. Mice underwent controlled cortical injury and received either whole molecule EPO, JM4, or sham-treatment with phosphate-buffered saline. Animals treated with JM4 peptide exhibited a large decrease in number of dead neural cells and a marked reduction in lesion size at both 3 and 8 days postinjury. Therapy with JM4 also led to improved functional recovery and we observed a treatment window for JM4 peptide that remained open for at least 9 h postinjury. The full-length EPO molecule was divided into a series of 6 contiguous peptide segments; the JM4-containing segment and the adjoining downstream region contained the bulk of the death attenuating effects seen with intact EPO molecule following TBI. These findings indicate that the JM4 molecule substantially blocks cell death and brain injury following acute brain trauma and, as such, presents an excellent opportunity to explore the therapeutic potential of a small-peptide EPO derivative in the medical treatment of TBI.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Erythropoietin/therapeutic use , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/pathology , Brain Injuries, Traumatic/pathology , Cell Death/drug effects , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL
7.
Neurotherapeutics ; 12(4): 850-61, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26271954

ABSTRACT

Beneficial effects of short-term whole-molecule erythropoietin (EPO) therapy have been demonstrated on several animal models of diverse central nervous system pathology. However, the increased hematocrit induced by EPO-driven marrow stimulation greatly limits its potential for side effect-free therapy. We created a library of EPO-derived fragments based on the hypothesis that 2 distinct functions, erythropoiesis and tissue protection, reside in different regions of the molecule. Several small EPO-derived peptides within the Aß loop of whole EPO molecule were screened for tissue protection in EAE mice. The 19-mer JM-4 peptide that contains 2 cysteine molecules consistently demonstrated the most potent clinical beneficial effects without producing hematocrit alterations in animal models of EAE. The JM-4-induced tissue protection was associated with modulation of the immunoregulatory process that drives inflammation and provokes subsequent autoimmune damage. Like the whole EPO molecule, JM-4 effectively modulated immune/inflammatory reaction within both the peripheral lymphatic tissue and central nervous system. The major effects induced by JM-4 include blocked expansion of monocyte/dendritic antigen presenting cell and T helper 17 cell populations, decreased proinflammatory cytokine production, and sharply enhanced expansion of the regulatory T-cell population. JM-4 shows promise for treatment of a broad spectrum of neural and non-neural conditions associated with inflammation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Erythropoietin/therapeutic use , Lymphocytes/drug effects , Animals , Apoptosis/drug effects , Cytokines/metabolism , Disease Models, Animal , Erythropoietin/chemistry , Humans , Lymph Nodes/drug effects , Mice, Inbred C57BL , PC12 Cells , Rats , Spinal Cord/drug effects , Spinal Cord/pathology
8.
J Hematol Oncol ; 7: 91, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25523825

ABSTRACT

BACKGROUND: Most patients with small cell lung cancer (SCLC) or neuroblastoma (NB) already show clinically detectable metastases at diagnosis and have an extremely poor prognosis even when treated with combined modalities. The HuD-antigen is a neuronal RNA-binding protein that is expressed in 100% of SCLC tumor cells and over 50% of neuroblastoma cells. The correlation between high titers of circulating anti-HuD antibodies in patients and spontaneous tumor remission suggests that the HuD-antigen might be a potential molecular target for immunotherapy. METHODS: We have constructed a new antibody-toxin compound (called BW-2) by assembling a mouse anti-human-HuD monoclonal antibody onto streptavidin/saporin complexes. RESULTS: We found that the immunotoxin BW-2 specifically killed HuD-positive human SCLC and NB cancer cells at very low concentrations in vitro. Moreover, intratumoral immunotoxin therapy in a nude mouse model of human SCLC (n = 6) significantly reduced local tumor progression without causing toxicity. When the same intratumoral immunotoxin protocol was applied to an immunocompetent A/J mouse model of NB, significant inhibition of local tumor growth was also observed. In neuroblastoma allografted A/J mice (n = 5) treated twice with intratumoral immunotoxin, significant tumor regression occurred in over 80% of the animals and their duration of tumor response was significantly prolonged. CONCLUSIONS: Our study suggests that anti-HuD based immunotoxin therapy may prove to be an effective alternative treatment for patients with SCLC and NB.


Subject(s)
ELAV Proteins/immunology , Immunotoxins/pharmacology , Lung Neoplasms/drug therapy , Neuroblastoma/drug therapy , Small Cell Lung Carcinoma/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , ELAV-Like Protein 4 , Humans , Immunotherapy/methods , Immunotoxins/chemistry , Lung Neoplasms/immunology , Male , Mice , Mice, Nude , Neuroblastoma/immunology , Prognosis , Ribosome Inactivating Proteins, Type 1/chemistry , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Small Cell Lung Carcinoma/immunology , Streptavidin/chemistry , Streptavidin/pharmacology , Xenograft Model Antitumor Assays
9.
Int J MS Care ; 14(3): 105-14, 2012.
Article in English | MEDLINE | ID: mdl-24453741

ABSTRACT

It has recently been suggested that the Lublin-Reingold clinical classification of multiple sclerosis (MS) be modified to include the use of magnetic resonance imaging (MRI). An international consensus conference sponsored by the Consortium of Multiple Sclerosis Centers (CMSC) was held from March 5 to 7, 2010, to review the available evidence on the need for such modification of the Lublin-Reingold criteria and whether the addition of MRI or other biomarkers might lead to a better understanding of MS pathophysiology and disease course over time. The conference participants concluded that evidence of new MRI gadolinium-enhancing (Gd+) T1-weighted lesions and unequivocally new or enlarging T2-weighted lesions (subclinical activity, subclinical relapses) should be added to the clinical classification of MS in distinguishing relapsing inflammatory from progressive forms of the disease. The consensus was that these changes to the classification system would provide more rigorous definitions and categorization of MS course, leading to better insights as to the evolution and treatment of MS.

11.
PLoS One ; 3(4): e1924, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18382691

ABSTRACT

BACKGROUND: Beneficial effects of short-term erythropoietin (EPO) therapy have been demonstrated in several animal models of acute neurologic injury, including experimental autoimmune encephalomyelitis (EAE)--the animal model of multiple sclerosis. We have found that EPO treatment substantially reduces the acute clinical paralysis seen in EAE mice and this improvement is accompanied by a large reduction in the mononuclear cell infiltration and downregulation of glial MHC class II expression within the inflamed CNS. Other reports have recently indicated that peripherally generated anti-inflammatory CD4(+)Foxp3(+) regulatory T cells (Tregs) and the IL17-producing CD4+ T helper cell (Th17) subpopulations play key antagonistic roles in EAE pathogenesis. However, no information regarding the effects of EPO therapy on the behavior of the general mononuclear-lymphocyte population, Tregs or Th17 cells in EAE has emerged. METHODS AND FINDINGS: We first determined in vivo that EPO therapy markedly suppressed MOG specific T cell proliferation and sharply reduced the number of reactive dendritic cells (CD11c positive) in EAE lymph nodes during both inductive and later symptomatic phases of MOG(35-55) induced EAE. We then determined the effect in vivo of EPO on numbers of peripheral Treg cells and Th17 cells. We found that EPO treatment modulated immune balance in both the periphery and the inflamed spinal cord by promoting a large expansion in Treg cells, inhibiting Th17 polarization and abrogating proliferation of the antigen presenting dendritic cell population. Finally we utilized tissue culture assays to show that exposure to EPO in vitro similarly downregulated MOG-specific T cell proliferation and also greatly suppressed T cell production of pro-inflammatory cytokines. CONCLUSIONS: Taken together, our findings reveal an important new locus whereby EPO induces substantial long-term tissue protection in the host through signaling to several critical subsets of immune cells that reside in the peripheral lymphatic system.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Erythropoietin/pharmacology , Animals , Autoimmune Diseases , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Forkhead Transcription Factors/metabolism , Genes, MHC Class II , Inflammation/metabolism , Interleukin-17/biosynthesis , Leukocytes, Mononuclear/cytology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL
12.
Brain Res ; 1203: 126-32, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18316065

ABSTRACT

Sodium valproate (VPA) administered to neonatal mice causes cognitive and motor deficits similar to those observed in humans with autism. In an effort to further evaluate similarities between early VPA exposure and autism, the present study examined treated mice for deficits in social behavior and neuronal damage. BALB/c mice injected on P14 with 400 mg/kg VPA engaged in fewer social interactions (including ano-genital sniffs, allogrooming, and crawl-under/over behaviors) than control mice. Treated mice also exhibited reduced motor activity in a social context but were not significantly different from controls when motor activity was assessed in non-social settings. A second set of BALB/c mice were treated with VPA on P14 and sacrificed at different times thereafter for histopathological analysis. At time-points 12 and 24 h following VPA, treated mice had up to a 30-fold increase in the number of TUNEL-positive cells in the external granule cell layer of the cerebellum and a 10-fold increase in TUNEL-positive cells in the dentate gyrus of the hippocampus. These observations may provide a histopathological correlate for the social deficits observed following post-natal VPA exposure and supports the use of early VPA administration as an animal model for the study of autism.


Subject(s)
Anticonvulsants/toxicity , Apoptosis/drug effects , Behavioral Symptoms/chemically induced , Behavioral Symptoms/pathology , Valproic Acid/toxicity , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cell Count , Female , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Sex Factors , Social Behavior , Time Factors
13.
Ann Neurol ; 56(6): 767-77, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15562412

ABSTRACT

We have known for a long time that erythropoietin signaling plays a key role in bone marrow erythrocyte proliferation. However, recent studies have indicated that erythropoietin also may have protective effects on the nervous system. This unexpected role remains incompletely characterized. To investigate the potential neuroprotective role of erythropoietin in the central nervous system, we assessed its effects on a well-characterized autoimmune demyelinating model of multiple sclerosis-myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE) in the mouse. We found that erythropoietin administered intravenously for 14 days after the onset of symptoms reduced both disease severity and duration of maximum impairment at dose levels as low as 50U/kg (p < 0.001). We assessed the neuropathology of diseased spinal cords and found that erythropoietin-treated EAE animals had reduced axonal damage, inflammatory cell infiltration and demyelination, and diminished blood-brain barrier leakage when compared with saline-treated EAE controls. Moreover, the pronounced upregulation of spinal cord major histocompatibility complex (MHC) class II expression found in saline-treated EAE was significantly reduced in erythropoietin-treated animals, a finding we replicated in vitro, using microglial cultures. The notion that short-term erythropoietin therapy might be of clinical benefit in human autoimmune demyelinating diseases needs investigation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Erythropoietin/therapeutic use , Animals , Cell Count/methods , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Mice, Inbred C57BL
14.
J Neurosci Res ; 69(2): 189-96, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12111800

ABSTRACT

The purpose of this study was to determine how oligodendrocytes die following Fas receptor activation. An immortalized human oligodendrocyte hybrid line (MO3.13) was challenged with Fas ligand (FasL), and cell death was assessed by flow cytometry and DNA gel electrophoresis. Caspase activation was determined by either Western immunoblotting on cell extracts or by whole-cell flow cytometry. FasL challenge clearly induced substantial apoptotic cell death in the oligodendrocyte hybrid cell line, as judged by flow cytometry and by the presence of prominent low molecular weight DNA banding patterns after gel electrophoresis. Western immunoblots showed marked increases in cleaved caspase-1, 8, and 3, indicating that the extrinsic Fas death receptor-induced pathway was activated. The intrinsic mitochondrial pathway was also activated, but only at a minimal level. These findings demonstrate that there are several independent molecular sites within the extrinsic caspase cascade in oligodendrocytes where inhibitory compounds may be capable of blocking cell death in vivo.


Subject(s)
Caspases/metabolism , Membrane Glycoproteins/pharmacology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , fas Receptor/metabolism , Apoptosis , Blotting, Western , Caspase 3 , Caspase 9 , Cell Culture Techniques , Electrophoresis, Agar Gel , Fas Ligand Protein , Flow Cytometry , Humans , Hybrid Cells , Membrane Glycoproteins/administration & dosage , Oligodendroglia/enzymology
15.
J Neurol Sci ; 197(1-2): 9-18, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11997061

ABSTRACT

Caspase-1 is responsible for processing inflammatory cytokines and is associated with the induction of apoptosis. Using RT-PCR, we found that caspase-1 mRNA transcripts from frozen brain extracts were significantly elevated in multiple sclerosis (MS) compared to controls. Immunohistochemical staining using a specific antiserum confirmed the marked up regulation of caspase-1 within acute and chronic MS plaques, while little staining was seen in control brains. In addition to the expected caspase-1 expression in microglia and infiltrating perivascular mononuclear cells, we found that cytoplasmic caspase-1 expression was sharply increased in the resident oligodendrocytes of MS lesions. The TUNEL reaction for fragmented DNA co-localized over an occasional caspase-1-expressing cell and large numbers of caspase-1-positive "corpses" were observed within phagocytic macrophages of an acute evolving MS lesion. Studies using an immortalized human oligodendroglial hybrid cell line exposed to cytokine challenge showed that death induction was blocked by the caspase-1-like inhibitor Z-YVAD-fmk, while the caspase-3-like inhibitor Z-DEVD-fmk was less effective. Cellular levels of procaspase-1 were reduced compared to controls in oligodendroglia induced to die by cytokine challenge, as judged by Western immunoblotting. Our results suggest that caspase-1 may play a role in the inflammatory and apoptotic processes associated with MS pathogenesis.


Subject(s)
Caspase 1/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oligodendroglia/enzymology , Caspase 1/analysis , Cell Death , Cells, Cultured , Gene Expression , Humans , Hybrid Cells , In Situ Nick-End Labeling , Macrophages/pathology , Nerve Fibers/enzymology , Nerve Fibers/pathology , Oligodendroglia/pathology , Phagocytosis , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...