Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 710608, 2021.
Article in English | MEDLINE | ID: mdl-34504493

ABSTRACT

Aging adversely affects inflammatory processes in the brain, which has important implications in the progression of neurodegenerative disease. Following traumatic brain injury (TBI), aged animals exhibit worsened neurological function and exacerbated microglial-associated neuroinflammation. Type I Interferons (IFN-I) contribute to the development of TBI neuropathology. Further, the Cyclic GMP-AMP Synthase (cGAS) and Stimulator of Interferon Genes (STING) pathway, a key inducer of IFN-I responses, has been implicated in neuroinflammatory activity in several age-related neurodegenerative diseases. Here, we set out to investigate the effects of TBI on cGAS/STING activation, IFN-I signaling and neuroinflammation in young and aged C57Bl/6 male mice. Using a controlled cortical impact model, we evaluated transcriptomic changes in the injured cortex at 24 hours post-injury, and confirmed activation of key neuroinflammatory pathways in biochemical studies. TBI induced changes were highly enriched for transcripts that were involved in inflammatory responses to stress and host defense. Deeper analysis revealed that TBI increased expression of IFN-I related genes (e.g. Ifnb1, Irf7, Ifi204, Isg15) and IFN-I signaling in the injured cortex of aged compared to young mice. There was also a significant age-related increase in the activation of the DNA-recognition pathway, cGAS, which is a key mechanism to propagate IFN-I responses. Finally, enhanced IFN-I signaling in the aged TBI brain was confirmed by increased phosphorylation of STAT1, an important IFN-I effector molecule. This age-related activation of cGAS and IFN-I signaling may prove to be a mechanistic link between microglial-associated neuroinflammation and neurodegeneration in the aged TBI brain.


Subject(s)
Aging/immunology , Brain Injuries, Traumatic/immunology , Interferon Type I/physiology , Nucleotidyltransferases/metabolism , Aging/metabolism , Animals , Enzyme Activation , Interferon Type I/genetics , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Microglia/physiology , Neurodegenerative Diseases/etiology , Neuroinflammatory Diseases/etiology , Signal Transduction/physiology
2.
Genome Res ; 30(6): 885-897, 2020 06.
Article in English | MEDLINE | ID: mdl-32660935

ABSTRACT

RNA-seq is widely used for studying gene expression, but commonly used sequencing platforms produce short reads that only span up to two exon junctions per read. This makes it difficult to accurately determine the composition and phasing of exons within transcripts. Although long-read sequencing improves this issue, it is not amenable to precise quantitation, which limits its utility for differential expression studies. We used long-read isoform sequencing combined with a novel analysis approach to compare alternative splicing of large, repetitive structural genes in muscles. Analysis of muscle structural genes that produce medium (Nrap: 5 kb), large (Neb: 22 kb), and very large (Ttn: 106 kb) transcripts in cardiac muscle, and fast and slow skeletal muscles identified unannotated exons for each of these ubiquitous muscle genes. This also identified differential exon usage and phasing for these genes between the different muscle types. By mapping the in-phase transcript structures to known annotations, we also identified and quantified previously unannotated transcripts. Results were confirmed by endpoint PCR and Sanger sequencing, which revealed muscle-type-specific differential expression of these novel transcripts. The improved transcript identification and quantification shown by our approach removes previous impediments to studies aimed at quantitative differential expression of ultralong transcripts.


Subject(s)
Gene Expression Profiling , High-Throughput Nucleotide Sequencing , RNA, Messenger , Sequence Analysis, RNA , Transcriptome , Alternative Splicing , Computational Biology/methods , Exons , Gene Expression Profiling/methods , Humans , Molecular Sequence Annotation , Organ Specificity/genetics , Repetitive Sequences, Nucleic Acid
3.
Clin Cancer Res ; 26(5): 1152-1161, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31615935

ABSTRACT

PURPOSE: Immunotherapy promises unprecedented benefits to patients with cancer. However, the majority of cancer types, including high-risk neuroblastoma, remain immunologically unresponsive. High-intensity focused ultrasound (HIFU) is a noninvasive technique that can mechanically fractionate tumors, transforming immunologically "cold" tumors into responsive "hot" tumors. EXPERIMENTAL DESIGN: We treated <2% of tumor volume in previously unresponsive, large, refractory murine neuroblastoma tumors with mechanical HIFU and assessed systemic immune response using flow cytometry, ELISA, and gene sequencing. In addition, we combined this treatment with αCTLA-4 and αPD-L1 to study its effect on the immune response and long-term survival. RESULTS: Combining HIFU with αCTLA-4 and αPD-L1 significantly enhances antitumor response, improving survival from 0% to 62.5%. HIFU alone causes upregulation of splenic and lymph node NK cells and circulating IL2, IFNγ, and DAMPs, whereas immune regulators like CD4+Foxp3+, IL10, and VEGF-A are significantly reduced. HIFU combined with checkpoint inhibitors induced significant increases in intratumoral CD4+, CD8α+, and CD8α+CD11c+ cells, CD11c+ in regional lymph nodes, and decrease in circulating IL10 compared with untreated group. We also report significant abscopal effect following unilateral treatment of mice with large, established bilateral tumors using HIFU and checkpoint inhibitors compared with tumors treated with HIFU or checkpoint inhibitors alone (61.1% survival, P < 0.0001). This combination treatment significantly also induces CD4+CD44+hiCD62L+low and CD8α+CD44+hiCD62L+low population and is adoptively transferable, imparting immunity, slowing subsequent de novo tumor engraftment. CONCLUSIONS: Mechanical fractionation of tumors using HIFU can effectively induce immune sensitization in a previously unresponsive murine neuroblastoma model and promises a novel yet efficacious immunoadjuvant modality to overcome therapeutic resistance.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm , High-Intensity Focused Ultrasound Ablation/methods , Immunity, Cellular , Neuroblastoma/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Combined Modality Therapy , Dendritic Cells/immunology , Disease Models, Animal , Lymph Nodes/immunology , Mice , Mice, Inbred A , Neuroblastoma/immunology
4.
Inflamm Res ; 68(11): 969-980, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31446438

ABSTRACT

OBJECTIVE AND DESIGN: The objective of this study was to assess the effect of vamorolone, a first-in-class dissociative steroidal compound, to inhibit inflammation when administered after disease onset in the murine collagen antibody-induced arthritis model of arthritis. ANIMALS: 84 DBA1/J mice were used in this study (n = 12 per treatment group). TREATMENT: Vamorolone or prednisolone was administered orally after disease onset for a duration of 7 days. METHODS: Disease score and bone erosion were assessed using previously described scoring systems. Cytokines were measured in joints via immunoassay, and joint cathepsin B activity (marker of inflammation) was assessed using optical imaging of joints on live mice. RESULTS: We found that vamorolone treatment led to a reduction of several disease parameters including disease score, joint inflammation, and the presence of pro-inflammatory mediators to a degree similar of that observed with prednisolone treatment. More importantly, histopathological analysis of affected joints showed that vamorolone treatment significantly reduced the degree of bone erosion while this bone-sparing property was not observed with prednisolone treatment at any of the tested doses. CONCLUSIONS: While many intervention regimens in other studies are administered prior to disease onset in animal models, the current study involves delivery of the potential therapeutic after disease onset. Based on the findings, vamorolone may offer an efficacious, yet safer alternative to conventional steroidal compounds in the treatment of rheumatoid arthritis and other inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Pregnadienediols/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Collagen Type II/immunology , Cytokines/immunology , Joints/drug effects , Joints/immunology , Joints/pathology , Lipopolysaccharides , Male , Mice, Inbred DBA
5.
Cell Mol Neurobiol ; 35(3): 377-387, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25392236

ABSTRACT

Multiple sclerosis is a chronic disease of the central nervous system characterized by an autoimmune inflammatory reaction that leads to axonal demyelination and tissue damage. Glucocorticoids, such as prednisolone, are effective in the treatment of multiple sclerosis in large part due to their ability to inhibit pro-inflammatory pathways (e.g., NFκB). However, despite their effectiveness, long-term treatment is limited by adverse side effects. VBP15 is a recently described compound synthesized based on the lazeroid steroidal backbone that shows activity in acute and chronic inflammatory conditions, yet displays a much-reduced side effect profile compared to traditional glucocorticoids. The purpose of this study was to determine the effectiveness of VBP15 in inhibiting inflammation and disease progression in experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of multiple sclerosis. Our data show that VBP15 is effective at reducing both disease onset and severity. In parallel studies, we observed that VBP15 was able to inhibit the production of NFκB-regulated pro-inflammatory transcripts in human macrophages. Furthermore, treatment with prednisolone-but not VBP15-increased expression of genes associated with bone loss and muscle atrophy, suggesting lack of side effects of VBP15. These findings suggest that VBP15 may represent a potentially safer alternative to traditional glucocorticoids in the treatment of multiple sclerosis and other inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Pregnadienediols/therapeutic use , Severity of Illness Index , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/pathology , Pregnadienediols/pharmacology , Pregnancy , Treatment Outcome
6.
Top Spinal Cord Inj Rehabil ; 20(2): 147-57, 2014.
Article in English | MEDLINE | ID: mdl-25477737

ABSTRACT

BACKGROUND: Activity-based therapy (ABT) focuses on regaining motor and sensory function below the level of the lesion in persons with a spinal cord injury (SCI). This is accomplished through repetitive training of specific motor tasks. Research has shown that ABT may increase neuroplasticity in the rat and human spinal cord. OBJECTIVE: The primary aim of this study was to examine acute alterations in neuroplasticity-related proteins during ABT in persons with SCI. METHODS: Volunteers were current participants in an ABT program and consisted of 12 men and 3 women (age, 31.8 ± 10.9 years) with chronic SCI (injury duration, 63.9 ± 54.4 months). A single 2-hour bout of ABT consisted of standing load bearing, body weight-supported treadmill training, whole body vibration, and functional electrical stimulation. Blood samples were obtained at baseline and immediately after completion of each modality to determine serum levels of brain-derived neurotrophic factor (BDNF), prolactin, and cortisol. RESULTS: One-way analysis of variance (ANOVA) with repeated measures was used to examine differences in proteins over time. Results revealed baseline levels of BDNF (2.37 ± 1.41 ng/mL) that were lower than previous research has demonstrated in persons with SCI. No change in BDNF or cortisol was found, although prolactin was significantly reduced in response to ABT. CONCLUSION: Despite the length of the bout, acute changes in BDNF were not observed. Whether different intensities or modalities of ABT may promote acute increases in serum BDNF in individuals with SCI remains to be determined and further study is merited.

7.
Brain Behav ; 2(5): 563-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23139902

ABSTRACT

Galectins are pleiotropic carbohydrate-binding lectins involved in inflammation, growth/differentiation, and tissue remodeling. The functional role of galectins in amyotrophic lateral sclerosis (ALS) is unknown. Expression studies revealed increases in galectin-1 mRNA and protein in spinal cords from SOD1(G93A) mice, and in galectin-3 and -9 mRNAs and proteins in spinal cords of both SOD1(G93A) mice and sporadic ALS patients. As the increase in galectin-3 appeared in early presymptomatic stages and increased progressively through to end stage of disease in the mouse, it was selected for additional study, where it was found to be mainly expressed by microglia. Galectin-3 antagonists are not selective and do not readily cross the blood-brain barrier; therefore, we generated SOD1(G93A)/Gal-3(-/-) transgenic mice to evaluate galectin-3 deletion in a widely used mouse model of ALS. Disease progression, neurological symptoms, survival, and inflammation were assessed to determine the effect of galectin-3 deletion on the SOD1(G93A) disease phenotype. Galectin-3 deletion did not change disease onset, but resulted in more rapid progression through functionally defined disease stages, more severely impaired neurological symptoms at all stages of disease, and expiration, on average, 25 days earlier than SOD1(G93A)/Gal-3(+/+) cohorts. In addition, microglial staining, as well as TNF-α, and oxidative injury were increased in SOD1(G93A)/Gal-3(-/-) mice compared with SOD1(G93A)/Gal-3(+/+) cohorts. These data support an important functional role for microglial galectin-3 in neuroinflammation during chronic neurodegenerative disease. We suggest that elevations in galectin-3 by microglia as disease progresses may represent a protective, anti-inflammatory innate immune response to chronic motor neuron degeneration.

8.
Brain Res ; 1475: 96-105, 2012 Sep 26.
Article in English | MEDLINE | ID: mdl-22884909

ABSTRACT

Inflammation has long been implicated in secondary tissue damage after spinal cord injury (SCI). Our previous studies of inflammatory gene expression in rats after SCI revealed two temporally correlated clusters: the first was expressed early after injury and the second was up-regulated later, with peak expression at 1-2 weeks and persistent up-regulation through 6 months. To further address the role of inflammation after SCI, we examined inflammatory genes in a second species, mice, through 28 days after SCI. Using anchor gene clustering analysis, we found similar expression patterns for both the acute and chronic gene clusters previously identified after rat SCI. The acute group returned to normal expression levels by 7 days post injury. The chronic group, which included C1qB, p22(phox) and galectin-3, showed peak expression at 7 days and remained up-regulated through 28 days. Immunohistochemistry and western blot analysis showed that the protein expression of these genes was consistent with the mRNA expression. Further exploration of the role of one of these genes, galectin-3, suggests that galectin-3 may contribute to secondary injury. In summary, our findings extend our prior gene profiling data by demonstrating the chronic expression of a cluster of microglial associated inflammatory genes after SCI in mice. Moreover, by demonstrating that inhibition of one such factor improves recovery, the findings suggest that such chronic up-regulation of inflammatory processes may contribute to secondary tissue damage after SCI, and that there may be a broader therapeutic window for neuroprotection than generally accepted.


Subject(s)
Galectin 3/physiology , Gene Expression Regulation , Inflammation Mediators/metabolism , Spinal Cord Injuries/metabolism , Animals , Animals, Newborn , Cells, Cultured , Galectin 3/biosynthesis , Galectin 3/genetics , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology
9.
J Neuroinflammation ; 8: 130, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21975064

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) induces secondary tissue damage that is associated with inflammation. We have previously demonstrated that inflammation-related gene expression after SCI occurs in two waves - an initial cluster that is acutely and transiently up-regulated within 24 hours, and a more delayed cluster that peaks between 72 hours and 7 days. Here we extend the microarray analysis of these gene clusters up to 6 months post-SCI. METHODS: Adult male rats were subjected to mild, moderate or severe spinal cord contusion injury at T9 using a well-characterized weight-drop model. Tissue from the lesion epicenter was obtained 4 hours, 24 hours, 7 days, 28 days, 3 months or 6 months post-injury and processed for microarray analysis and protein expression. RESULTS: Anchor gene analysis using C1qB revealed a cluster of genes that showed elevated expression through 6 months post-injury, including galectin-3, p22PHOX, gp91PHOX, CD53 and progranulin. The expression of these genes occurred primarily in microglia/macrophage cells and was confirmed at the protein level using both immunohistochemistry and western blotting. As p22PHOX and gp91PHOX are components of the NADPH oxidase enzyme, enzymatic activity and its role in SCI were assessed and NADPH oxidase activity was found to be significantly up-regulated through 6 months post-injury. Further, treating rats with the nonspecific, irreversible NADPH oxidase inhibitor diphenylene iodinium (DPI) reduced both lesion volume and expression of chronic gene cluster proteins one month after trauma. CONCLUSIONS: These data demonstrate that inflammation-related genes are chronically up-regulated after SCI and may contribute to further tissue loss.


Subject(s)
Gene Expression , Inflammation/genetics , Inflammation/immunology , RNA, Messenger/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Animals , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Macrophages/physiology , Magnetic Resonance Imaging , Male , Microarray Analysis , Microglia/physiology , Multigene Family , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
10.
PLoS One ; 5(9): e12981, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20886045

ABSTRACT

The dysferlin deficient SJL/J mouse strain is commonly used to study dysferlin deficient myopathies. Therefore, we systematically evaluated behavior in relatively young (9-25 weeks) SJL/J mice and compared them to C57BL6 mice to determine which functional end points may be the most effective to use for preclinical studies in the SJL/J strain. SJL/J mice had reduced body weight, lower open field scores, higher creatine kinase levels, and less muscle force than did C57BL6 mice. Power calculations for expected effect sizes indicated that grip strength normalized to body weight and open field activity were the most sensitive indicators of functional status in SJL/J mice. Weight and open field scores of SJL/J mice deteriorated over the course of the study, indicating that progressive myopathy was ongoing even in relatively young (<6 months old) SJL/J mice. To further characterize SJL/J mice within the context of treatment, we assessed the effect of fasudil, a rho-kinase inhibitor, on disease phenotype. Fasudil was evaluated based on previous observations that Rho signaling may be overly activated as part of the inflammatory cascade in SJL/J mice. Fasudil treated SJL/J mice showed increased body weight, but decreased grip strength, horizontal activity, and soleus muscle force, compared to untreated SJL/J controls. Fasudil either improved or had no effect on these outcomes in C57BL6 mice. Fasudil also reduced the number of infiltrating macrophages/monocytes in SJL/J muscle tissue, but had no effect on muscle fiber degeneration/regeneration. These studies provide a basis for standardization of preclinical drug testing trials in the dysferlin deficient SJL/J mice, and identify measures of functional status that are potentially translatable to clinical trial outcomes. In addition, the data provide pharmacological evidence suggesting that activation of rho-kinase, at least in part, may represent a beneficial compensatory response in dysferlin deficient myopathies.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Membrane Proteins/deficiency , Muscular Dystrophies, Limb-Girdle/drug therapy , Muscular Dystrophies, Limb-Girdle/metabolism , Protein Kinase Inhibitors/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Dysferlin , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/physiopathology
11.
Glia ; 53(4): 420-33, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16345062

ABSTRACT

The dual role of microglia in cytotoxicity and neuroprotection is believed to depend on the specific, temporal expression of microglial-related genes. To better clarify this issue, we used high-density oligonucleotide microarrays to examine microglial gene expression after spinal cord injury (SCI) in rats. We compared expression changes at the lesion site, as well as in rostral and caudal regions after mild, moderate, or severe SCI. Using microglial-associated anchor genes, we identified two clusters with different temporal profiles. The first, induced by 4 h postinjury to peak between 4 and 24 h, included interleukin-1beta, interleukin-6, osteopontin, and calgranulin, among others. The second was induced 24 h after SCI, and peaked between 72 h and 7 days; it included C1qB, Galectin-3, and p22(phox). These two clusters showed similar expression profiles regardless of injury severity, albeit with slight decreases in expression in mild or severe injury vs. moderate injury. Expression was also decreased rostral and caudal to the lesion site. We validated the expression of selected cluster members at the mRNA and protein levels. In addition, we demonstrated that stimulation of purified microglia in culture induces expression of C1qB, Galectin-3, and p22(phox). Finally, inhibition of p22(phox) activity within microglial cultures significantly suppressed proliferation in response to stimulation, confirming that this gene is involved in microglial activation. Because microglial-related factors have been implicated both in secondary injury and recovery, identification of temporally distinct clusters of genes related to microglial activation may suggest distinct roles for these groups of factors.


Subject(s)
Microglia/physiology , Multigene Family/genetics , Spinal Cord Injuries/pathology , Acetophenones/pharmacology , Animals , Biomarkers , Blotting, Western , Cells, Cultured , Data Interpretation, Statistical , Galectin 3/genetics , Gene Expression/physiology , Gene Expression Profiling , Hyaluronan Receptors/genetics , Immunohistochemistry , Inflammation Mediators/physiology , Male , Membrane Transport Proteins/genetics , Mitochondrial Proteins , NADPH Oxidases/genetics , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/genetics
12.
Ann N Y Acad Sci ; 1053: 472-81, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16179555

ABSTRACT

It has long been recognized that thyrotropin-releasing hormone (TRH) and certain TRH analogues are neuroprotective in a variety of animal models of CNS trauma. In addition to these neuroprotective actions, TRH and most TRH analogues have other physiological actions that may not be desirable for treatment of acute injury, such as analeptic, autonomic, and endocrine effects. We have developed a series of dual-substituted TRH analogues that have strong neuroprotective actions, but are largely devoid of these other physiological actions. In addition, we have developed a family of cyclized dipeptides (diketopiperazines), structurally somewhat related to a metabolic product of TRH, that appear even more effective as neuroprotective agents in vitro and in vivo, and may have nootropic properties. Here, we review these novel tripeptide and dipeptide compounds.


Subject(s)
Neuroprotective Agents , Oligopeptides/pharmacology , Animals , Cyclization , Humans , Oligopeptides/chemistry , Thyrotropin-Releasing Hormone/analogs & derivatives
13.
Physiol Genomics ; 22(3): 368-81, 2005 Aug 11.
Article in English | MEDLINE | ID: mdl-15942019

ABSTRACT

Changes in gene expression contribute to pathophysiological alterations following spinal cord injury (SCI). We examined gene expression over time (4 h, 24 h, 7 days) at the impact site, as well as rostral and caudal regions, following mild, moderate, or severe contusion SCI in rats. High-density oligonucleotide microarrays were used that included approximately 27,000 genes/ESTs (Affymetrix RG-U34; A, B and C arrays), together with multiple analyses (MAS 5.0, dChip). Alterations after mild injury were relatively rapid (4 and 24 h), whereas they were delayed and prolonged after severe injury (24 h and 7 days). The number and magnitude of gene expression changes were greatest at the injury site after moderate injury and increased in rostral and caudal regions as a function of injury severity. Sham surgery resulted in expression changes that were similar to mild injury, suggesting the importance of using time-linked surgical controls as well as naive animals for these kinds of studies. Expression of many genes and ESTs was altered; these were classified functionally based on ontology. Overall representation of these functional classes varied with distance from the site of injury and injury severity, as did the individual genes that contributed to each functional class. Different clustering approaches were used to identify changes in neuronal-specific genes and several transcription factors that have not previously been associated with SCI. This study represents the most comprehensive evaluation of gene expression changes after SCI to date. The results underscore the power of microarray approaches to reveal global genomic responses as well as changes in particular gene clusters and/or families that may be important in the secondary injury cascade.


Subject(s)
Gene Expression Profiling , Spinal Cord Injuries/metabolism , Algorithms , Animals , Base Sequence , Central Nervous System/pathology , Cluster Analysis , Expressed Sequence Tags , Gene Expression , Gene Expression Regulation , Molecular Sequence Data , Multigene Family , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Potassium Channels/chemistry , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic
14.
Neuropharmacology ; 49(3): 410-24, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15907950

ABSTRACT

Thyrotropin-releasing hormone (TRH) and TRH analogues have been reported to be neuroprotective in experimental models of spinal cord injury and head injury. We have previously shown that a diketopiperazine structurally related to the TRH metabolite cyclo-his-pro reduces neuronal cell death in vitro and in vivo. Here we report the neuroprotective activity of other cyclic dipeptides in multiple in vitro models of neuronal injury and after controlled cortical impact (CCI) in mice. Using primary neuronal cultures, three novel dipeptides were compared to the previously reported diketopiperazine as well as to vehicle controls; each of the compounds reduced cell death after direct physical trauma or trophic withdrawal. Two of these peptides also protected against glutamate toxicity and beta-amyloid-induced injury; the latter also strongly inhibited glutamate-induced increases in intracellular calcium. Treatment with each of the test compounds resulted in highly significant improvement of motor and cognitive recovery after CCI, as well as markedly reducing lesion volumes as shown by high field magnetic resonance imaging. DNA microarray studies following fluid percussion induced traumatic brain injury (TBI) in rats showed that treatment with one of these dipeptides after injury significantly down-regulated expression of mRNAs for cell cycle proteins, aquaporins, cathepsins and calpain in ipsilateral cortex and/or hippocampus, while up-regulating expression of brain-derived neurotrophic factor, hypoxia-inducible factor and several heat-shock proteins. Many of these mRNA expression changes were paralleled at the protein level. The fact that these small peptides modulate multiple mechanisms favoring neuronal cell survival, as well as their ability to improve functional outcome and reduce posttraumatic lesion size, suggests that they may have potential utility in clinical head injury.


Subject(s)
Brain Injuries/drug therapy , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Brain Injuries/pathology , Cell Survival/drug effects , Cells, Cultured , Female , Image Interpretation, Computer-Assisted , Immunoblotting , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Magnetic Resonance Imaging , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Oligonucleotide Array Sequence Analysis , Peptides/therapeutic use , Pregnancy , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
15.
Mol Cell Neurosci ; 29(3): 355-71, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15905098

ABSTRACT

Ceramide accumulates in neurons during various disorders associated with acute or chronic neurodegeneration. In these studies, we investigated the mechanisms of ceramide-induced apoptosis in primary cortical neurons using exogenous C(2) ceramide as well as inducing endogenous ceramide accumulation using inhibitors of glucosylceramide synthetase. Ceramide induced the translocation of certain, but not all, pro-apoptotic mitochondrial proteins: cytochrome c, Omi, SMAC, and AIF were released from the mitochondria, whereas Endonuclease G was not. Ceramide also selectively altered the phosphorylation state of members of the MAPK superfamily, causing dephosphorylation of ERK1/2 and hyperphosphorylation of p38 MAP kinases, but not affecting the phosphorylation of JNK or ERK5. Inhibitors of the p38 MAP kinase pathway (SB-202190 or SB-203580) and an inhibitor of the ERK1/2 pathway (U0126) reduced ceramide-induced neuronal death. These p38 and ERK1/2 inhibitors appear to block ceramide-activated apoptotic signaling upstream of the mitochondria, as they attenuated mitochondrial release of cytochrome c, Omi, AIF, and SMAC, as well as reducing ceramide-induced caspase-3 activation.


Subject(s)
Apoptosis/physiology , Ceramides/metabolism , MAP Kinase Signaling System/physiology , Mitochondria/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cells, Cultured , Ceramides/toxicity , Cytochromes c/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Flavoproteins/metabolism , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , High-Temperature Requirement A Serine Peptidase 2 , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neurons/pathology , Rats , Serine Endopeptidases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Cereb Blood Flow Metab ; 24(10): 1119-32, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15529012

ABSTRACT

In studies designed to evaluate the therapeutic window for treatment of traumatic brain injury, the caspase 3 inhibitor z-DEVD-fmk improved neurologic function and reduced lesion volumes when administered at 1 but not at 4, 8, or 24 hours after injury. Moreover, neither caspase 3 nor PARP, a caspase 3 substrate, were cleaved in injured, untreated cortex from 1 to 72 hours after injury. Few cortical neurons expressed active caspase 3 or were TUNEL positive from 6 to 24 hours after injury, and TUNEL staining was primarily Type I (necrotic). Nissl staining revealed extensive neuronal necrosis in the injured cortex from 6 to 24 hours after impact. Considered together, these data suggested that z-DEVD-fmk may reduce neuronal necrosis, so we used an in vitro model of necrotic cell death induced by maitotoxin to test this further and explore the potential mechanism(s) involved. Z-DEVD-fmk (1 nM-100 microM) significantly attenuated maitotoxin induced neuronal cell death and markedly reduced expression of the 145 kD calpain-mediated alpha-spectrin breakdown product after maitotoxin injury. Neither the 120 kD caspase-mediated alpha-spectrin cleavage product nor cathepsin B were expressed after maitotoxin injury. In a cell free assay, z-DEVD-fmk reduced hydrolysis of casein by purified calpain I. Finally, z-DEVD-fmk reduced expression of the 145 kD calpain-mediated alpha-spectrin cleavage fragment after traumatic brain injury in vivo. These data suggest that neuroprotection by z-DEVD-fmk may, in part, reflect inhibition of calpain-related necrotic cell death.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/metabolism , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Oligopeptides/pharmacology , Animals , Brain Injuries/pathology , Calpain/metabolism , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell-Free System , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Necrosis , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/pathology
17.
J Cereb Blood Flow Metab ; 24(5): 564-78, 2004 May.
Article in English | MEDLINE | ID: mdl-15129189

ABSTRACT

Endocannabinoids, including 2-arachidonoylglycerol and anandamide (N-arachidonoylethanolamine; AEA), have neuroprotective effects in the brain through actions at CB1 receptors. However, AEA also binds to vanilloid (VR1) receptors and induces cell death in several cell lines. Here we show that anandamide causes neuronal cell death in vitro and exacerbates cell loss caused by stretch-induced axonal injury or trophic withdrawal in rat primary neuronal cultures. Administered intracerebroventricularly, AEA causes sustained cerebral edema, as reflected by diffusion-weighted magnetic resonance imaging, regional cell loss, and impairment in long-term cognitive function. These effects are mediated, in part, through VR1 as well as through calpain-dependent mechanisms, but not through CB1 receptors or caspases. Central administration of AEA also significantly upregulates genes involved in pro-inflammatory/microglial-related responses. Thus, anandamide produces neurotoxic effects both in vitro and in vivo through multiple mechanisms independent of the CB1 receptor.


Subject(s)
Arachidonic Acids/metabolism , Cannabinoid Receptor Modulators/metabolism , Cell Death/physiology , Endocannabinoids , Neurons/metabolism , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/toxicity , Calpain/metabolism , Cannabinoid Receptor Modulators/administration & dosage , Cannabinoid Receptor Modulators/toxicity , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/anatomy & histology , Cerebral Cortex/metabolism , Embryo, Mammalian/anatomy & histology , Enzyme Activation , Gene Expression Profiling , Hippocampus/anatomy & histology , Hippocampus/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Maze Learning , Memory Disorders , Mice , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Polyunsaturated Alkamides , Rats
18.
J Alzheimers Dis ; 6(6 Suppl): S93-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15665420

ABSTRACT

The tripeptide thyrotropin-releasing hormone (TRH) and/or related analogues have shown neuroprotective activity across multiple animal trauma models as well as in a small clinical trial of spinal cord injury. The metabolic product of TRH (cyclo-his-pro) retains physiological activity. We have developed a number of novel cyclic dipeptides that are structurally similar to cyclo-his-pro, and have examined their neuroprotective activity across multiple in vitro models of neuronal injury and after traumatic brain injury (TBI) in rodents. Four such compounds were found to reduce cell death after trophic withdrawal or traumatic injury in primary neuronal cultures; two also protected against glutamate or beta-amyloid neurotoxicity. All compounds significantly improved motor and cognitive recovery after controlled cortical impact injury in mice, and markedly reduced lesion volumes as shown by high field magnetic resonance imaging. Further, compound 35b, which is being developed for clinical trials, also showed considerable neuroprotection after fluid percussion induced TBI in rats, and improved cognitive function after daily administration in chronically brain injured rats. At a mechanistic level, the drugs attenuate both apoptotic and necrotic cell death in primary neuronal cultures, markedly reduce intracellular calcium accumulation after injury, and limit changes in mitochondrial membrane potential and associated cytochrome c release. In addition, microarray studies show that 35b reduces transcriptional changes after injury for a number of genes (and proteins) that may be associated with secondary injury, including cell cycle genes, aquaporins and cathepsins. It also upregulates brain-derived neurotrophic factor (BDNF), heat shock proteins (HSP) and hypoxia inducible factor (HIF). Thus, these novel dipeptides have multipotential actions that make them candidates for the treatment of both acute and chronic neurodegeneration.


Subject(s)
Dipeptides/pharmacology , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Peptides, Cyclic/pharmacology , Animals , Apoptosis/drug effects , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries/pathology , Brain-Derived Neurotrophic Factor/metabolism , Calcium/metabolism , Cell Culture Techniques , Cognition/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression/genetics , Genes, bcl-2/drug effects , Genes, bcl-2/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Magnetic Resonance Imaging , Maze Learning/drug effects , Necrosis , Neurons/drug effects , Neurons/pathology , Rats , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Up-Regulation/drug effects , Up-Regulation/genetics
19.
Ann Neurol ; 53(4): 454-68, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12666113

ABSTRACT

Spinal cord injury causes secondary biochemical changes leading to neuronal cell death. To clarify the molecular basis of this delayed injury, we subjected rats to spinal cord injury and identified gene expression patterns by high-density oligonucleotide arrays (8,800 genes studied) at 30 minutes, 4 hours, 24 hours, or 7 days after injury (total of 26 U34A profiles). Detailed analyses were limited to 4,300 genes consistently expressed above background. Temporal clustering showed rapid expression of immediate early genes (30 minutes), followed by genes associated with inflammation, oxidative stress, DNA damage, and cell cycle (4 and 24 hours). Functional clustering showed a novel pattern of cell cycle mRNAs at 4 and 24 hours after trauma. Quantitative reverse transcription polymerase chain reaction verified mRNA changes in this group, which included gadd45a, c-myc, cyclin D1 and cdk4, pcna, cyclin G, Rb, and E2F5. Changes in their protein products were quantified by Western blot, and cell-specific expression was determined by immunocytochemistry. Cell cycle proteins showed an increased expression 24 hours after injury and were, in part, colocalized in neurons showing morphological evidence of apoptosis. These findings suggest that cell cycle-related genes, induced after spinal cord injury, are involved in neuronal damage and subsequent cell death.


Subject(s)
Cell Death/genetics , Gene Expression Profiling , Neurons/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Animals , Cell Cycle/genetics , Genes, Immediate-Early/genetics , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Male , Myelitis/genetics , Myelitis/pathology , Myelitis/physiopathology , Neurons/physiology , Oxidative Stress/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord Injuries/physiopathology , Up-Regulation/genetics
20.
J Cereb Blood Flow Metab ; 23(3): 342-54, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12621309

ABSTRACT

The authors developed a novel diketopiperazine that shows neuroprotective activity in a variety of in vitro models, as well as in a clinically relevant experimental model of traumatic brain injury (TBI) in rats. Treatment with 1-ARA-35b (35b), a cyclized dipeptide derived from a modified thyrotropin-releasing hormone (TRH) analog, significantly reduced cell death associated with necrosis (maitotoxin), apoptosis (staurosporine), or mechanical injury in neuronal-glial cocultures. Rats subjected to lateral fluid percussion-induced TBI and then treated with 1 mg/kg intravenous 35b thirty minutes after trauma showed significantly improved motor recovery and spatial learning compared with vehicle-treated controls. Treatment also significantly reduced lesion volumes as shown by magnetic resonance imaging, and decreased the number of TUNEL-positive neurons observed in ipsilateral hippocampus. Unlike TRH or traditional TRH analogs, 35b treatment did not change mean arterial pressure, body temperature, or thyroid-stimulating hormone release, and did not have analeptic activity. Moreover, in contrast to TRH or typical TRH analogs, 35b administration after TBI did not alter free-magnesium concentration or cellular bioenergetic state. Receptor-binding studies showed that 35b did not act with high affinity at 50 classical receptors, channels, or transporters. Thus, 35b shows none of the typical physiologic actions associated with TRH, but possesses neuroprotective actions in vivo and in vitro, and appears to attenuate both necrotic and apoptotic cell death.


Subject(s)
Brain Injuries/physiopathology , Brain Injuries/psychology , Cognition/drug effects , Dipeptides/pharmacology , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Peptides, Cyclic/pharmacology , Piperazines/pharmacology , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiopathology , Brain Injuries/diagnosis , Cells, Cultured , Central Nervous System Stimulants/pharmacology , Coculture Techniques , Dipeptides/metabolism , In Situ Nick-End Labeling , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Peptides, Cyclic/metabolism , Piperazines/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Recovery of Function , Wounds, Nonpenetrating/diagnosis , Wounds, Nonpenetrating/physiopathology , Wounds, Nonpenetrating/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...