Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(4): e0231194, 2020.
Article in English | MEDLINE | ID: mdl-32271817

ABSTRACT

Various injuries to the neural tissues can cause irreversible damage to multiple functions of the nervous system ranging from motor control to cognitive function. The limited treatment options available for patients have led to extensive interest in studying the mechanisms of neuronal regeneration and recovery from injury. Since many neurons are terminally differentiated, by increasing cell survival following injury it may be possible to minimize the impact of these injuries and provide translational potential for treatment of neuronal diseases. While several cell types are known to survive injury through plasma membrane repair mechanisms, there has been little investigation of membrane repair in neurons and even fewer efforts to target membrane repair as a therapy in neurons. Studies from our laboratory group and others demonstrated that mitsugumin 53 (MG53), a muscle-enriched tripartite motif (TRIM) family protein also known as TRIM72, is an essential component of the cell membrane repair machinery in skeletal muscle. Interestingly, recombinant human MG53 (rhMG53) can be applied exogenously to increase membrane repair capacity both in vitro and in vivo. Increasing the membrane repair capacity of neurons could potentially minimize the death of these cells and affect the progression of various neuronal diseases. In this study we assess the therapeutic potential of rhMG53 to increase membrane repair in cultured neurons and in an in vivo mouse model of neurotrauma. We found that a robust repair response exists in various neuronal cells and that rhMG53 can increase neuronal membrane repair both in vitro and in vivo. These findings provide direct evidence of conserved membrane repair responses in neurons and that these repair mechanisms can be targeted as a potential therapeutic approach for neuronal injury.


Subject(s)
Nerve Regeneration , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Wound Healing , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Crush Injuries/pathology , Crush Injuries/physiopathology , Disease Models, Animal , Humans , Membrane Proteins/metabolism , Membranes , Mice, Inbred C57BL , Nerve Regeneration/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Recombinant Proteins/pharmacology , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Tripartite Motif Proteins/pharmacology , Wound Healing/drug effects
2.
J Neurosci ; 39(7): 1139-1149, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30559153

ABSTRACT

Clinical studies indicate that psychosocial stress contributes to adverse chronic pain outcomes in patients, but it is unclear how this is initiated or amplified by stress. Repeated social defeat (RSD) is a mouse model of psychosocial stress that activates microglia, increases neuroinflammatory signaling, and augments pain and anxiety-like behaviors. We hypothesized that activated microglia within the spinal cord facilitate increased pain sensitivity following RSD. Here we show that mechanical allodynia in male mice was increased with exposure to RSD. This stress-induced behavior corresponded with increased mRNA expression of several inflammatory genes, including IL-1ß, TNF-α, CCL2, and TLR4 in the lumbar spinal cord. While there were several adhesion and chemokine-related genes increased in the lumbar spinal cord after RSD, there was no accumulation of monocytes or neutrophils. Notably, there was evidence of microglial activation selectively within the nociceptive neurocircuitry of the dorsal horn of the lumbar cord. Elimination of microglia using the colony stimulating factor 1 receptor antagonist PLX5622 from the brain and spinal cord prevented the development of mechanical allodynia in RSD-exposed mice. Microglial elimination also attenuated RSD-induced IL-1ß, CCR2, and TLR4 mRNA expression in the lumbar spinal cord. Together, RSD-induced allodynia was associated with microglia-mediated inflammation within the dorsal horn of the lumbar spinal cord.SIGNIFICANCE STATEMENT Mounting evidence indicates that psychological stress contributes to the onset and progression of adverse nociceptive conditions. We show here that repeated social defeat stress causes increased pain sensitivity due to inflammatory signaling within the nociceptive circuits of the spinal cord. Studies here mechanistically tested the role of microglia in the development of pain by stress. Pharmacological ablation of microglia prevented stress-induced pain sensitivity. These findings demonstrate that microglia are critical mediators in the induction of pain conditions by stress. Moreover, these studies provide a proof of principle that microglia can be targeted as a therapeutic strategy to mitigate adverse pain conditions.


Subject(s)
Chronic Pain/physiopathology , Chronic Pain/psychology , Inflammation/psychology , Microglia , Social Environment , Spinal Cord Diseases/psychology , Stress, Psychological/psychology , Animals , Anxiety/psychology , Behavior, Animal , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Chronic Pain/genetics , Gene Expression Regulation/genetics , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Inflammation/genetics , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Organic Chemicals/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Spinal Cord , Spinal Cord Diseases/genetics , Spinal Cord Diseases/physiopathology , Spinal Cord Injuries , Stress, Psychological/genetics
3.
J Neurotrauma ; 35(18): 2167-2182, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29385890

ABSTRACT

Sensorimotor recovery after spinal cord injury (SCI) is of utmost importance to injured individuals and will rely on improved understanding of SCI pathology and recovery. Novel transgenic mouse lines facilitate discovery, but must be understood to be effective. The purpose of this study was to characterize the sensory and motor behavior of a common transgenic mouse line (Thy1-GFP-M) before and after SCI. Thy1-GFP-M positive (TG+) mice and their transgene negative littermates (TG-) were acquired from two sources (in-house colony, n = 32, Jackson Laboratories, n = 4). C57BL/6J wild-type (WT) mice (Jackson Laboratories, n = 10) were strain controls. Moderate-severe T9 contusion (SCI) or transection (TX) occurred in TG+ (SCI, n = 25, TX, n = 5), TG- (SCI, n = 5), and WT (SCI, n = 10) mice. To determine responsiveness to rehabilitation, a cohort of TG+ mice with SCI (n = 4) had flat treadmill (TM) training 42-49 days post-injury (dpi). To characterize recovery, we performed Basso Mouse Scale, Grid Walk, von Frey Hair, and Plantar Heat Testing before and out to day 42 post-SCI. Open field locomotion was significantly better in the Thy1 SCI groups (TG+ and TG-) compared with WT by 7 dpi (p < 0.01) and was maintained through 42 dpi (p < 0.01). These unexpected locomotor gains were not apparent during grid walking, indicating severe impairment of precise motor control. Thy1 derived mice were hypersensitive to mechanical stimuli at baseline (p < 0.05). After SCI, mechanical hyposensitivity emerged in Thy1 derived groups (p < 0.001), while thermal hyperalgesia occurred in all groups (p < 0.001). Importantly, consistent findings across TG+ and TG- groups suggest that the effects are mediated by the genetic background rather than transgene manipulation itself. Surprisingly, TM training restored mechanical and thermal sensation to baseline levels in TG+ mice with SCI. This behavioral profile and responsiveness to chronic training will be important to consider when choosing models to study the mechanisms underlying sensorimotor recovery after SCI.


Subject(s)
Behavior, Animal/physiology , Disease Models, Animal , Spinal Cord Injuries/physiopathology , Thy-1 Antigens/genetics , Animals , Locomotion/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Sci Rep ; 8(1): 723, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335509

ABSTRACT

Interleukin 1 is a pleiotropic cytokine that mediates diverse functions through its receptor, type I interleukin 1 receptor (IL-1R1). Most previous studies have focused on the expression and function of IL-1R1 in immune cells. Here we performed a comprehensive mapping of IL-1R1 distribution in multiple peripheral tissues using our IL-1R1 reporter (IL-1R1GR/GR) mice. This method yielded the highest sensitivity of in situ detection of IL-1R1 mRNA and protein. Besides validating previously reported IL-1R1 expression in the endocrine tissues including pituitary and pancreas, our results refuted previously reported exclusive IL-1R1 expression in neurons of the spinal cord dorsal horn and dorsal root ganglia (DRG). Instead, IL-1R1 expression was detected in endothelial cells within DRG, spinal cord, pancreas, colon, muscles and many immune organs. In addition, gp38+ fibroblastic reticular cells (FRCs), rather than tissue macrophages or other immune cells, were found to express high levels of IL-1R1 in colon and many immune organs. A functional test of spleen FRCs showed that they responded rapidly to systemic IL-1ß stimulation in vivo. Taken together, this study provides a rigorous re-examination of IL-1R1 expression in peripheral tissues and reveals tissue FRCs as a previously unappreciated novel high IL-1R1-expressing cell type in peripheral IL-1 signaling.


Subject(s)
Animal Structures/chemistry , Animal Structures/physiology , Gene Expression Profiling , Receptors, Interleukin-1/biosynthesis , Animals , Mice , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1/genetics
5.
Neural Plast ; 2017: 8640970, 2017.
Article in English | MEDLINE | ID: mdl-28928988

ABSTRACT

Stress, injury, and disease trigger glucocorticoid (GC) elevation. Elevated GCs bind to the ubiquitously expressed glucocorticoid receptor (GR). While GRs are in every cell in the nervous system, the expression level varies, suggesting that diverse cell types react differently to GR activation. Stress/GCs induce structural plasticity in neurons, Schwann cells, microglia, oligodendrocytes, and astrocytes as well as affect neurotransmission by changing the release and reuptake of glutamate. While general nervous system plasticity is essential for adaptation and learning and memory, stress-induced plasticity is often maladaptive and contributes to neuropsychiatric disorders and neuropathic pain. In this brief review, we describe the evidence that stress/GCs activate GR to promote cell type-specific changes in cellular plasticity throughout the nervous system.


Subject(s)
Brain/metabolism , Cell Plasticity/physiology , Glucocorticoids/metabolism , Neuroglia/metabolism , Receptors, Glucocorticoid/metabolism , Spinal Cord/metabolism , Animals , Brain/cytology , Neuroglia/cytology , Spinal Cord/cytology
6.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28828403

ABSTRACT

Stress and glucocorticoid (GC) release are common behavioral and hormonal responses to injury or disease. In the brain, stress/GCs can alter neuron structure and function leading to cognitive impairment. Stress and GCs also exacerbate pain, but whether a corresponding change occurs in structural plasticity of sensory neurons is unknown. Here, we show that in female mice (Mus musculus) basal GC receptor (Nr3c1, also known as GR) expression in dorsal root ganglion (DRG) sensory neurons is 15-fold higher than in neurons in canonical stress-responsive brain regions (M. musculus). In response to stress or GCs, adult DRG neurite growth increases through mechanisms involving GR-dependent gene transcription. In vivo, prior exposure to an acute systemic stress increases peripheral nerve regeneration. These data have broad clinical implications and highlight the importance of stress and GCs as novel behavioral and circulating modifiers of neuronal plasticity.


Subject(s)
Axons/physiology , Nerve Regeneration/physiology , Receptors, Glucocorticoid/metabolism , Stress, Psychological/complications , Stress, Psychological/pathology , Transcriptional Activation/physiology , Activating Transcription Factor 3/metabolism , Animals , Calcium-Binding Proteins , Disease Models, Animal , Female , Ganglia, Spinal/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hormone Antagonists/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mifepristone/pharmacology , Nerve Tissue Proteins/metabolism , Neurites/pathology , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/physiology , Stathmin
7.
FEBS Lett ; 591(14): 2091-2105, 2017 07.
Article in English | MEDLINE | ID: mdl-28626869

ABSTRACT

Peripheral neurons regenerate their axons after injury. Transcriptional regulation by microRNAs (miRNAs) is one possible mechanism controlling regeneration. We profiled miRNA expression in mouse dorsal root ganglion neurons after a sciatic nerve crush, and identified 49 differentially expressed miRNAs. We evaluated the functional role of each miRNA using a phenotypic analysis approach. To predict the targets of the miRNAs we employed RNA-Sequencing and examined transcription at the isoform level. We identify thousands of differentially expressed isoforms and bioinformatically associate the miRNAs that modulate neurite growth with their putative target isoforms to outline a network of regulatory events underlying peripheral nerve regeneration. MiR-298, let-7a, and let-7f enhance neurite growth and target the majority of isoforms in the differentially expressed network.


Subject(s)
Ganglia, Spinal/cytology , MicroRNAs/genetics , Neuronal Outgrowth/genetics , Animals , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Phenotype , RNA Isoforms/genetics , Sequence Analysis, RNA , Transcription, Genetic
9.
Glia ; 65(6): 883-899, 2017 06.
Article in English | MEDLINE | ID: mdl-28251686

ABSTRACT

Oligodendrocyte progenitor cells (OPCs) are present throughout the adult brain and spinal cord and can replace oligodendrocytes lost to injury, aging, or disease. Their differentiation, however, is inhibited by myelin debris, making clearance of this debris an important step for cellular repair following demyelination. In models of peripheral nerve injury, TLR4 activation by lipopolysaccharide (LPS) promotes macrophage phagocytosis of debris. Here we tested whether the novel synthetic TLR4 agonist E6020, a Lipid A mimetic, promotes myelin debris clearance and remyelination in spinal cord white matter following lysolecithin-induced demyelination. In vitro, E6020 induced TLR4-dependent cytokine expression (TNFα, IL1ß, IL-6) and NF-κB signaling, albeit at ∼10-fold reduced potency compared to LPS. Microinjection of E6020 into the intact rat spinal cord gray/white matter border induced macrophage activation, OPC proliferation, and robust oligodendrogenesis, similar to what we described previously using an intraspinal LPS microinjection model. Finally, a single co-injection of E6020 with lysolecithin into spinal cord white matter increased axon sparing, accelerated myelin debris clearance, enhanced Schwann cell infiltration into demyelinated lesions, and increased the number of remyelinated axons. In vitro assays confirmed that direct stimulation of macrophages by E6020 stimulates myelin phagocytosis. These data implicate TLR4 signaling in promoting repair after CNS demyelination, likely by stimulating phagocytic activity of macrophages, sparing axons, recruiting myelinating cells, and promoting remyelination. This work furthers our understanding of immune-myelin interactions and identifies a novel synthetic TLR4 agonist as a potential therapeutic avenue for white matter demyelinating conditions such as spinal cord injury and multiple sclerosis.


Subject(s)
Demyelinating Diseases/drug therapy , Myelin Sheath/drug effects , Neuroprotective Agents/pharmacology , Phospholipids/pharmacology , Spinal Cord/drug effects , Animals , Axons/drug effects , Axons/pathology , Axons/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Disease Models, Animal , Female , Lysophosphatidylcholines , Macrophages/drug effects , Macrophages/physiology , Mice, Inbred C3H , Mice, Knockout , Myelin Sheath/pathology , Myelin Sheath/physiology , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Neural Stem Cells/physiology , Phagocytosis/drug effects , Phagocytosis/physiology , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/physiopathology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
10.
J Neurosci ; 36(23): 6352-64, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27277810

ABSTRACT

UNLABELLED: Acute oligodendrocyte (OL) death after traumatic spinal cord injury (SCI) is followed by robust neuron-glial antigen 2 (NG2)-positive OL progenitor proliferation and differentiation into new OLs. Inflammatory mediators are prevalent during both phases and can influence the fate of NG2 cells and OLs. Specifically, toll-like receptor (TLR) 4 signaling induces OL genesis in the naive spinal cord, and lack of TLR4 signaling impairs white matter sparing and functional recovery after SCI. Therefore, we hypothesized that TLR4 signaling may regulate oligodendrogenesis after SCI. C3H/HeJ (TLR4-deficient) and control (C3H/HeOuJ) mice received a moderate midthoracic spinal contusion. TLR4-deficient mice showed worse functional recovery and reduced OL numbers compared with controls at 24 h after injury through chronic time points. Acute OL loss was accompanied by reduced ferritin expression, which is regulated by TLR4 and needed for effective iron storage. TLR4-deficient injured spinal cords also displayed features consistent with reduced OL genesis, including reduced NG2 expression, fewer BrdU-positive OLs, altered BMP4 signaling and inhibitor of differentiation 4 (ID4) expression, and delayed myelin phagocytosis. Expression of several factors, including IGF-1, FGF2, IL-1ß, and PDGF-A, was altered in TLR4-deficient injured spinal cords compared with wild types. Together, these data show that TLR4 signaling after SCI is important for OL lineage cell sparing and replacement, as well as in regulating cytokine and growth factor expression. These results highlight new roles for TLR4 in endogenous SCI repair and emphasize that altering the function of a single immune-related receptor can dramatically change the reparative responses of multiple cellular constituents in the injured CNS milieu. SIGNIFICANCE STATEMENT: Myelinating cells of the CNS [oligodendrocytes (OLs)] are killed for several weeks after traumatic spinal cord injury (SCI), but they are replaced by resident progenitor cells. How the concurrent inflammatory signaling affects this endogenous reparative response is unclear. Here, we provide evidence that immune receptor toll-like receptor 4 (TLR4) supports OL lineage cell sparing, long-term OL and OL progenitor replacement, and chronic functional recovery. We show that TLR4 signaling is essential for acute iron storage, regulating cytokine and growth factor expression, and efficient myelin debris clearance, all of which influence OL replacement. Importantly, the current study reveals that a single immune receptor is essential for repair responses after SCI, and the potential mechanisms of this beneficial effect likely change over time after injury.


Subject(s)
Gene Expression Regulation/genetics , Nerve Regeneration/genetics , Oligodendroglia/physiology , Spinal Cord Injuries/pathology , Toll-Like Receptor 4/deficiency , Animals , Axons/pathology , Cell Differentiation/physiology , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Exploratory Behavior/physiology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred C3H , Mice, Transgenic , Nerve Regeneration/physiology , Phagocytosis/genetics , Recovery of Function/genetics , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Toll-Like Receptor 4/genetics
11.
World J Psychiatry ; 6(1): 18-30, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-27014595

ABSTRACT

The brain is a complex network system that has the capacity to support emotion, thought, action, learning and memory, and is characterized by constant activity, constant structural remodeling, and constant attempt to compensate for this remodeling. The basic insight that emerges from complex network organization is that substantively different networks can share common key organizational principles. Moreover, the interdependence of network organization and behavior has been successfully demonstrated for several specific tasks. From this viewpoint, increasing experimental/clinical observations suggest that mental disorders are neural network disorders. On one hand, single psychiatric disorders arise from multiple, multifactorial molecular and cellular structural/functional alterations spreading throughout local/global circuits leading to multifaceted and heterogeneous clinical symptoms. On the other hand, various mental diseases may share functional deficits across the same neural circuit as reflected in the overlap of symptoms throughout clinical diagnoses. An integrated framework including experimental measures and clinical observations will be necessary to formulate a coherent and comprehensive understanding of how neural connectivity mediates and constraints the phenotypic expression of psychiatric disorders.

12.
Neural Regen Res ; 11(1): 37-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26981074

ABSTRACT

Glucocorticoid and glucocorticoid receptor (GC/GR) interactions alter numerous aspects of neuronal function. These consequences (e.g., anti-inflammatory vs. pro-inflammatory) can vary depending on the duration of GC exposure or central nervous system (CNS) injury model. In this review we discuss how GC/GR interactions impact neuronal recovery after a central or peripheral nerve injury and discuss how GC exposure duration can produce divergent CNS neuronal growth responses. Finally we consider how new findings on gender specific immune cell responses after a nerve injury could intersect with GC/GR interactions to impact pain processing.

13.
Nat Commun ; 7: 10794, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26908229

ABSTRACT

Treatment for heart disease, the leading cause of death in the world, has progressed little for several decades. Here we develop a protein engineering approach to directly tune in vivo cardiac contractility by tailoring the ability of the heart to respond to the Ca(2+) signal. Promisingly, our smartly formulated Ca(2+)-sensitizing TnC (L48Q) enhances heart function without any adverse effects that are commonly observed with positive inotropes. In a myocardial infarction (MI) model of heart failure, expression of TnC L48Q before the MI preserves cardiac function and performance. Moreover, expression of TnC L48Q after the MI therapeutically enhances cardiac function and performance, without compromising survival. We demonstrate engineering TnC can specifically and precisely modulate cardiac contractility that when combined with gene therapy can be employed as a therapeutic strategy for heart disease.


Subject(s)
Calcium/metabolism , Heart Ventricles/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Protein Engineering , Troponin C/genetics , Ventricular Function , Animals , Calcium Signaling , Electrocardiography , Exercise Test , Exercise Tolerance , Genetic Therapy , Genetic Vectors , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Myocardial Contraction , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Optical Imaging , Rabbits , Troponin C/metabolism , Ultrasonography
14.
J Neurosci ; 35(41): 13860-7, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468186

ABSTRACT

Understanding why adult CNS neurons fail to regenerate their axons following injury remains a central challenge of neuroscience research. A more complete appreciation of the biological mechanisms shaping the injured nervous system is a crucial prerequisite for the development of robust therapies to promote neural repair. Historically, the identification of regeneration associated signaling pathways has been impeded by the limitations of available genetic and molecular tools. As we progress into an era in which the high-throughput interrogation of gene expression is commonplace and our knowledge base of interactome data is rapidly expanding, we can now begin to assemble a more comprehensive view of the complex biology governing axon regeneration. Here, we highlight current and ongoing work featuring transcriptomic approaches toward the discovery of novel molecular mechanisms that can be manipulated to promote neural repair. SIGNIFICANCE STATEMENT: Transcriptional profiling is a powerful technique with broad applications in the field of neuroscience. Recent advances such as single-cell transcriptomics, CNS cell type-specific and developmental stage-specific expression libraries are rapidly enhancing the power of transcriptomics for neuroscience applications. However, extracting biologically meaningful information from large transcriptomic datasets remains a formidable challenge. This mini-symposium will highlight current work using transcriptomic approaches to identify regulatory networks in the injured nervous system. We will discuss analytical strategies for transcriptomics data, the significance of noncoding RNA networks, and the utility of multiomic data integration. Though the studies featured here specifically focus on neural repair, the approaches highlighted in this mini-symposium will be of broad interest and utility to neuroscientists working in diverse areas of the field.


Subject(s)
Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Gene Expression Profiling/methods , Nerve Regeneration/physiology , Transcriptome/physiology , Animals , Humans
15.
Mol Cell Neurosci ; 68: 272-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26306672

ABSTRACT

Neurons in the embryonic and peripheral nervous system respond to injury by activating transcriptional programs supportive of axon growth, ultimately resulting in functional recovery. In contrast, neurons in the adult central nervous system (CNS) possess a limited capacity to regenerate axons after injury, fundamentally constraining repair. Activating pro-regenerative gene expression in CNS neurons is a promising therapeutic approach, but progress is hampered by incomplete knowledge of the relevant transcription factors. An emerging hypothesis is that factors implicated in cellular growth and motility outside the nervous system may also control axon growth in neurons. We therefore tested sixty-nine transcription factors, previously identified as possessing tumor suppressive or oncogenic properties in non-neuronal cells, in assays of neurite outgrowth. This screen identified YAP1 and E2F1 as enhancers of neurite outgrowth, and PITX1, RBM14, ZBTB16, and HHEX as inhibitors. Follow-up experiments are focused on the tumor suppressor HHEX, one of the strongest growth inhibitors. HHEX is widely expressed in adult CNS neurons, including corticospinal tract neurons after spinal injury, but is present only in trace amounts in immature cortical neurons and adult peripheral neurons. HHEX overexpression in early postnatal cortical neurons reduced both initial axonogenesis and the rate of axon elongation, and domain deletion analysis strongly implicated transcriptional repression as the underlying mechanism. These findings suggest a role for HHEX in restricting axon growth in the developing CNS, and substantiate the hypothesis that previously identified oncogenes and tumor suppressors can play conserved roles in axon extension.


Subject(s)
Axons/physiology , Central Nervous System/cytology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Neurons/cytology , Animals , Animals, Newborn , Fluoresceins/metabolism , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Rats , Rats, Sprague-Dawley , Transfection
16.
Front Genet ; 5: 131, 2014.
Article in English | MEDLINE | ID: mdl-24904634

ABSTRACT

Neuropathic pain (NP) is caused by damage to the nervous system, resulting in dysfunction and aberrant pain. The cellular functions (e.g., peripheral neuron spinal cord innervation, neuronal excitability) associated with NP often develop over time and are likely associated with gene expression changes. Gene expression studies on the cells involved in NP (e.g., sensory dorsal root ganglion neurons) are publically available; the mining of these studies may enable the identification of novel targets and the subsequent development of therapies that are essential for improving quality of life for the millions of individuals suffering with NP. Here we analyzed a publically available microarray dataset (GSE30165) in order to identify new RNAs (e.g., messenger RNA (mRNA) isoforms and non-coding RNAs) underlying NP. GSE30165 profiled gene expression in dorsal root ganglion neurons (DRG) and in sciatic nerve (SN) after resection, a NP model. Gene ontological analysis shows enrichment for sensory and neuronal processes. Protein network analysis demonstrates DRG upregulated genes typical to an injury and NP response. Of the top changing genes, 34 and 36% are associated with more than one protein coding isoform in the DRG and SN, respectively. The majority of genes are receptor and enzymes. We identified 15 long non-coding RNAs (lncRNAs) targeting these genes in LNCipedia.org, an online comprehensive lncRNA database. These RNAs represent new therapeutic targets for preventing NP development and this approach demonstrates the feasibility of data reanalysis for their identification.

17.
Semin Immunol ; 26(5): 409-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24768088

ABSTRACT

Traumatic spinal cord injury (SCI) activates the hypothalamic-pituitary-adrenal (HPA) axis, a potent neuroendocrine regulator of stress and inflammation. SCI also elicits a profound and sustained intraspinal and systemic inflammatory response. Together, stress hormones and inflammatory mediators will affect the growth and survival of neural and non-neural cells and ultimately neurologic recovery after SCI. Glucocorticoids (GCs) are endogenous anti-inflammatory steroids that are synthesized in response to stress or injury, in part to regulate inflammation. Exogenous synthetic GCs are often used for similar purposes in various diseases; however, their safety and efficacy in pre-clinical and clinical SCI is controversial. The relatively recent discovery that macrophage migration inhibitory factor (MIF) is produced throughout the body and can override the anti-inflammatory effects of GCs may provide unique insight to the importance of endogenous and exogenous GCs after SCI. Here, we review both GCs and MIF and discuss the potential relevance of their interactions after SCI, especially their role in regulating maladaptive mechanisms of plasticity and repair that may contribute to the onset and maintenance of neuropathic pain.


Subject(s)
Glucocorticoids/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Neuralgia/metabolism , Receptors, Glucocorticoid/metabolism , Spinal Cord Injuries/metabolism , Animals , Gene Expression Regulation , Glucocorticoids/genetics , Glucocorticoids/immunology , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Neuralgia/genetics , Neuralgia/immunology , Neuralgia/pathology , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/metabolism , Protein Interaction Mapping , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/immunology , Signal Transduction , Spinal Cord Injuries/genetics , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
18.
Mol Cell Neurosci ; 59: 97-105, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24521823

ABSTRACT

A number of genes regulate regeneration of peripheral axons, but their ability to drive axon growth and regeneration in the central nervous system (CNS) remains largely untested. To address this question we overexpressed eight transcription factors and one small GTPase alone and in pairwise combinations to test whether combinatorial overexpression would have a synergistic impact on CNS neuron neurite growth. The Jun oncogene/signal transducer and activator of transcription 6 (JUN/STAT6) combination increased neurite growth in dissociated cortical neurons and in injured cortical slices. In injured cortical slices, JUN overexpression increased axon growth to a similar extent as JUN and STAT6 together. Interestingly, JUN overexpression was not associated with increased growth associated protein 43 (GAP43) or integrin alpha 7 (ITGA7) expression, though these are predicted transcriptional targets. This study demonstrates that JUN overexpression in cortical neurons stimulates axon growth, but does so independently of changes in expression of genes thought to be critical for JUNs effects on axon growth. We conclude that JUN activity underlies this CNS axonal growth response, and that it is mechanistically distinct from peripheral regeneration responses, in which increases in JUN expression coincide with increases in GAP43 expression.


Subject(s)
Axons/metabolism , Cerebral Cortex/growth & development , Oncogene Protein p65(gag-jun)/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Axons/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Nerve Regeneration , Neurogenesis , Oncogene Protein p65(gag-jun)/genetics , Rats , Rats, Sprague-Dawley , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
19.
Proc Natl Acad Sci U S A ; 109(19): 7517-22, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22529377

ABSTRACT

Axon regeneration in the central nervous system normally fails, in part because of a developmental decline in the intrinsic ability of CNS projection neurons to extend axons. Members of the KLF family of transcription factors regulate regenerative potential in developing CNS neurons. Expression of one family member, KLF7, is down-regulated developmentally, and overexpression of KLF7 in cortical neurons in vitro promotes axonal growth. To circumvent difficulties in achieving high neuronal expression of exogenous KLF7, we created a chimera with the VP16 transactivation domain, which displayed enhanced neuronal expression compared with the native protein while maintaining transcriptional activation and growth promotion in vitro. Overexpression of VP16-KLF7 overcame the developmental loss of regenerative ability in cortical slice cultures. Adult corticospinal tract (CST) neurons failed to up-regulate KLF7 in response to axon injury, and overexpression of VP16-KLF7 in vivo promoted both sprouting and regenerative axon growth in the CST of adult mice. These findings identify a unique means of promoting CST axon regeneration in vivo by reengineering a developmentally down-regulated, growth-promoting transcription factor.


Subject(s)
Axons/physiology , Kruppel-Like Transcription Factors/metabolism , Nerve Regeneration/physiology , Pyramidal Tracts/physiology , Animals , Axons/metabolism , Cells, Cultured , Etoposide , Female , Gene Expression , Genetic Engineering , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Herpes Simplex Virus Protein Vmw65/genetics , Humans , Immunohistochemistry , Kruppel-Like Transcription Factors/genetics , Luminescent Measurements/methods , Mice , Mice, Inbred C57BL , Mutation , Nerve Regeneration/genetics , Neurites/metabolism , Neurites/physiology , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Pyramidal Tracts/cytology , Pyramidal Tracts/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Transcriptional Activation
20.
PLoS One ; 7(1): e30417, 2012.
Article in English | MEDLINE | ID: mdl-22272348

ABSTRACT

To fully understand cell type identity and function in the nervous system there is a need to understand neuronal gene expression at the level of isoform diversity. Here we applied Next Generation Sequencing of the transcriptome (RNA-Seq) to purified sensory neurons and cerebellar granular neurons (CGNs) grown on an axonal growth permissive substrate. The goal of the analysis was to uncover neuronal type specific isoforms as a prelude to understanding patterns of gene expression underlying their intrinsic growth abilities. Global gene expression patterns were comparable to those found for other cell types, in that a vast majority of genes were expressed at low abundance. Nearly 18% of gene loci produced more than one transcript. More than 8000 isoforms were differentially expressed, either to different degrees in different neuronal types or uniquely expressed in one or the other. Sensory neurons expressed a larger number of genes and gene isoforms than did CGNs. To begin to understand the mechanisms responsible for the differential gene/isoform expression we identified transcription factor binding sites present specifically in the upstream genomic sequences of differentially expressed isoforms, and analyzed the 3' untranslated regions (3' UTRs) for microRNA (miRNA) target sites. Our analysis defines isoform diversity for two neuronal types with diverse axon growth capabilities and begins to elucidate the complex transcriptional landscape in two neuronal populations.


Subject(s)
Gene Expression Regulation , High-Throughput Nucleotide Sequencing/methods , Neurons/metabolism , Sensory Receptor Cells/metabolism , Transcriptome , 5' Untranslated Regions/genetics , Animals , Binding Sites/genetics , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Cluster Analysis , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Logistic Models , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Open Reading Frames/genetics , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Protein Isoforms/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...