Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Skin Appendage Disord ; 10(3): 199-206, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835716

ABSTRACT

Background: Squamous cell carcinoma (SCC) and SCC in situ (Bowen's disease) are the most common malignancies of the nail unit. They are frequently seen in men over 50 and most commonly affect the fingers. The role of high-risk human papillomavirus (HPV) infection has been identified as a key contributor to the development of nail unit SCC. Summary: In this review, we aimed to summarize the current state of our understanding of how HPV contributes to nail unit SCC, the role of genitodigital transmission of HPV, and the clinical features of HPV-associated nail unit SCC. We also review current advances in the treatment of nail unit SCC, with a focus on the potential role of HPV vaccination in the treatment and prevention of nail unit SCC. Key Messages: Nail unit SCC should be recognized as an HPV-associated disease. HPV vaccination may represent a non-surgical modality for the management of these challenging malignancies in the appropriate clinical setting.

2.
Arch Dermatol Res ; 316(6): 233, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795205

ABSTRACT

Immune checkpoint inhibitor (ICI) therapies carry the risk of major immune-related adverse events (irAEs). Among the most severe irAEs is epidermal necrosis that may clinically mimic Stevens-Johnson syndrome (SJS) and toxic epidermal necrosis (TEN). The aim of this study was to provide a summary of the clinical and histological features of ICI-associated epidermal necrosis, with a special focus on factors associated with fatal outcomes in cases of extensive disease. A total of 98 cases, 2 new cases and 96 reported on PubMed and in the literature, of ICI-associated epidermal necrosis were assessed. Development of epidermal necrosis occurred between 1 day and 3 years after starting ICI therapy, with an average onset of 13.8 weeks for patients with limited (< 30% BSA) and 11.3 weeks for those with extensive (≥ 30% BSA) involvement, and a median onset of 5.8 weeks and 4 weeks respectively. A preceding rash was seen in 52 cases and was more common in extensive cases. Mucosal involvement was only reported in 65% of extensive cases but was significantly associated with fatal reactions. Co-administration of cytotoxic chemotherapy was associated with more extensive disease. Recovery was observed in 96% and 65% of those with limited and extensive involvement respectively and no specific therapy was associated with improved survival. Young age was significantly associated with poor outcomes in extensive disease, the average age of surviving patients was 64.5 years old versus 55.1 years old for deceased patients, p < 0.01. Both superficial perivascular and interface/lichenoid inflammatory infiltrates were commonly seen. These findings suggest that ICI-associated epidermal necrosis should be considered a distinct clinical entity from drug-induced SJS/TEN.


Subject(s)
Immune Checkpoint Inhibitors , Necrosis , Stevens-Johnson Syndrome , Humans , Immune Checkpoint Inhibitors/adverse effects , Stevens-Johnson Syndrome/pathology , Stevens-Johnson Syndrome/etiology , Stevens-Johnson Syndrome/immunology , Stevens-Johnson Syndrome/diagnosis , Necrosis/chemically induced , Epidermis/pathology , Epidermis/drug effects , Epidermis/immunology , Middle Aged , Female , Male , Aged , Adult
4.
Article in English | MEDLINE | ID: mdl-35817513

ABSTRACT

Chronic wounds are a significant challenge for patients, healthcare providers, and healthcare systems. Chronic wounds develop due to a complex interplay between chronic inflammation, tissue hypoxia, and oxidative stress, often occurring in the setting of advancing age. Ideally, new therapeutics should address all the components of chronic wound pathophysiology. Mesenchymal stem cell (MSC) therapies show significant promise to promote healing of chronic wounds. Extracellular vesicles (EVs) secreted by MSCs mediate many of their beneficial effects. We review the evidence demonstrating that MSC-EVs target the processes leading to chronic wounds. Additionally, we discuss how MSCs can be influenced to generate more potent wound healing EVs. Finally, we highlight the current state of EV clinical trials for wound healing and important preclinical studies that will lead to optimal use of MSC-EVs for patient care.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Extracellular Vesicles/metabolism , Humans , Inflammation/metabolism , Oxidative Stress , Wound Healing
5.
Pharmaceutics ; 13(10)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34683836

ABSTRACT

Chronic wounds develop when the orderly process of cutaneous wound healing is delayed or disrupted. Development of a chronic wound is associated with significant morbidity and financial burden to the individual and health-care system. Therefore, new therapeutic modalities are needed to address this serious condition. Mesenchymal stem cells (MSCs) promote skin repair, but their clinical use has been limited due to technical challenges. Extracellular vesicles (EVs) are particles released by cells that carry bioactive molecules (lipids, proteins, and nucleic acids) and regulate intercellular communication. EVs (exosomes, microvesicles, and apoptotic bodies) mediate key therapeutic effects of MSCs. In this review we examine the experimental data establishing a role for EVs in wound healing. Then, we explore techniques for designing EVs to function as a targeted drug delivery system and how EVs can be incorporated into biomaterials to produce a personalized wound dressing. Finally, we discuss the status of clinically deploying EVs as a therapeutic agent in wound care.

8.
Exp Dermatol ; 29(1): 93-101, 2020 01.
Article in English | MEDLINE | ID: mdl-31733169

ABSTRACT

Schwann cells (SCs) have long been recognized for their ability to support repair and promote axon regeneration following injury to the peripheral nervous system. In response to nerve injury, they rapidly dedifferentiate into a precursor-like state, secrete an array of inflammatory mediators and growth factors, proliferate, undergo epithelial-to-mesenchymal-like transformation to facilitate migration, phagocytose cellular debris and remodel the extracellular environment to promote regeneration of axons through the site of injury. However, even though a cutaneous role for SCs is becoming increasingly recognized, we argue in this Viewpoint essay that the likely complex functions of SCs in skin physiology and pathology beyond skin sensation and nerve repair deserve more attention and systemic research than they have received so far. For example, SCs promote wound healing, disseminate infection in leprosy, support the growth of neurofibromas/schwannomas and facilitate/accelerate the growth and invasion of melanoma. Despite representing a major dermal cell population, comparatively little is still known about the role of SCs in other dermatoses. To quintessentially illustrate the opportunities that promise to arise from a new skin research focus on SCs, we focus on two dermatoses that are not traditionally associated with SCs, that is, psoriasis and atopic dermatitis (AD), since both show distinct SC changes along with continuous nerve fibre degeneration and regeneration, and an impact of denervation on skin lesions. Specifically, we critically discuss the hypothesis that repeated activation of the SC repair programme occurs in and contributes to psoriasis and AD and delineate experimental approaches how to probe this clinically relevant hypothesis.


Subject(s)
Dermatitis, Atopic/physiopathology , Psoriasis/physiopathology , Schwann Cells/pathology , Schwann Cells/physiology , Skin Physiological Phenomena , Skin/pathology , Animals , Dermatitis, Atopic/pathology , Homeostasis , Humans , Psoriasis/pathology , Skin/cytology , Wound Healing
9.
Neuron ; 103(4): 642-657.e7, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31255486

ABSTRACT

Neuronal subtypes show diverse injury responses, but the molecular underpinnings remain elusive. Using transgenic mice that allow reliable visualization of axonal fate, we demonstrate that intrinsically photosensitive retinal ganglion cells (ipRGCs) are both resilient to cell death and highly regenerative. Using RNA sequencing (RNA-seq), we show genes that are differentially expressed in ipRGCs and that associate with their survival and axon regeneration. Strikingly, thrombospondin-1 (Thbs1) ranked as the most differentially expressed gene, along with the well-documented injury-response genes Atf3 and Jun. THBS1 knockdown in RGCs eliminated axon regeneration. Conversely, RGC overexpression of THBS1 enhanced regeneration in both ipRGCs and non-ipRGCs, an effect that was dependent on syndecan-1, a known THBS1-binding protein. All structural domains of the THBS1 were not equally effective; the trimerization and C-terminal domains promoted regeneration, while the THBS type-1 repeats were dispensable. Our results identify cell-type-specific induction of Thbs1 as a novel gene conferring high regenerative capacity.


Subject(s)
Nerve Regeneration/physiology , Retinal Ganglion Cells/physiology , Thrombospondin 1/physiology , Animals , Apoptosis , Axons/metabolism , Cell Line , Female , Gene Expression Profiling , Genes, Reporter , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Crush , Optic Nerve Injuries/genetics , Optic Nerve Injuries/physiopathology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Rod Opsins/deficiency , Rod Opsins/physiology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/physiology , Thrombospondin 1/biosynthesis , Thrombospondin 1/genetics , Transcription, Genetic
10.
Mol Syst Biol ; 14(8): e8202, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108134

ABSTRACT

Identifying critical pathways governing disease progression is essential for accurate prognosis and effective therapy. We developed a broadly applicable and novel systems-level gene discovery strategy. This approach focused on constitutively active androgen receptor (AR) splice variant-driven pathways as representative of an intractable mechanism of prostate cancer (PC) therapeutic resistance. We performed a meta-analysis of human prostate samples using weighted gene co-expression network analysis combined with experimental AR variant transcriptome analyses. An AR variant-driven gene module that is upregulated during human PC progression was identified. We filtered this module by identifying genes that functionally interacted with AR variants using a high-throughput synthetic genetic array screen in Schizosaccharomyces pombe This strategy identified seven AR variant-regulated genes that also enhance AR activity and drive cancer progression. Expression of the seven genes predicted poor disease-free survival in large independent PC patient cohorts. Pharmacologic inhibition of interacting members of the gene set potently and synergistically decreased PC cell proliferation. This unbiased and novel gene discovery strategy identified a clinically relevant, oncogenic, interacting gene hub with strong prognostic and therapeutic potential in PC.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Cell Line, Tumor , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Prostatic Neoplasms/pathology , RNA Splicing/genetics , Receptors, Androgen/chemistry , Schizosaccharomyces/genetics , Signal Transduction/genetics
11.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28856242

ABSTRACT

Retinal ganglion cells (RGCs), the sole output cells of the retina, are a heterogeneous population of neurons that project axons to visual targets in the brain. Like most CNS neurons, RGCs are considered incapable of mounting long distance axon regeneration. Using immunolabeling-enabled 3D imaging of solvent-cleared organs (iDISCO) in transgenic mice, we tracked the entire paths of individual RGC axons and show that adult RGCs are highly capable of spontaneous long-distance regeneration, even without any treatment. Our results show that the Thy1-H-YFP mouse sparsely labels RGCs, consisting predominantly of regeneration-competent α-type RGCs (αRGCs). Following optic nerve crush, many of the YFP-labeled RGC axons extend considerable distances proximal to the injury site with only a few penetrating through the lesion. This tortuous axon growth proximal to the lesion site is even more striking with intravitreal ciliary neurotrophic factor (CNTF) treatment. We further demonstrate that despite traveling more than 5 mm (i.e., a distance equal to the length of mouse optic nerve), many of these circuitous axons are confined to the injury area and fail to reach the brain. Our results re-evaluate the view that RGCs are naturally incapable of re-extending long axons, and shift the focus from promoting axon elongation, to understanding factors that prevent direct growth of axons through the lesion and the injured nerve.


Subject(s)
Nerve Regeneration , Retinal Ganglion Cells/pathology , Animals , Axons/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Enlargement , Ciliary Neurotrophic Factor/administration & dosage , Ciliary Neurotrophic Factor/metabolism , Female , Imaging, Three-Dimensional , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Transgenic , Nerve Regeneration/physiology , Optic Nerve/pathology , Optic Nerve/physiopathology , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Retinal Ganglion Cells/physiology
12.
Cutis ; 99(5): 339-341, 2017 May.
Article in English | MEDLINE | ID: mdl-28632795

ABSTRACT

Children who are not adequately protected from the sun have an increased risk for developing skin cancers later in life. The primary objective of this study was to determine the sun protection behaviors that black and Hispanic parents in Miami, Florida, employ in infants younger than 6 months. Secondary objectives included determining if this patient population is at risk for infant sunburns and tanning, beliefs among parents regarding sunscreen's efficacy in the prevention of skin cancers, and limitations of sunscreen use. An institutional review board-approved survey was administered to parents presenting to the University of Miami general pediatrics clinic. The main outcome measure was the self-reported consistency with which parents employed each of 6 sun protection strategies in infants. The results of this study highlight some potential shortcomings in current practices in sun protection for black and Hispanic infants.


Subject(s)
Health Knowledge, Attitudes, Practice , Parents , Sunburn/prevention & control , Sunscreening Agents/therapeutic use , Adult , Child, Preschool , Ethnicity , Florida , Humans , Infant , Infant, Newborn , Sunburn/ethnology , Sunscreening Agents/administration & dosage
13.
Cell Rep ; 15(2): 398-410, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050520

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is a transcription factor central to axon regrowth with an enigmatic ability to act in different subcellular regions independently of its transcriptional roles. However, its roles in mature CNS neurons remain unclear. Here, we show that along with nuclear translocation, STAT3 translocates to mitochondria in mature CNS neurons upon cytokine stimulation. Loss- and gain-of-function studies using knockout mice and viral expression of various STAT3 mutants demonstrate that STAT3's transcriptional function is indispensable for CNS axon regrowth, whereas mitochondrial STAT3 enhances bioenergetics and further potentiates regrowth. STAT3's localization, functions, and growth-promoting effects are regulated by mitogen-activated protein kinase kinase (MEK), an effect further enhanced by Pten deletion, leading to extensive axon regrowth in the mouse optic pathway and spinal cord. These results highlight CNS neuronal dependence on STAT3 transcriptional activity, with mitochondrial STAT3 providing ancillary roles, and illustrate a critical contribution for MEK in enhancing diverse STAT3 functions and axon regrowth.


Subject(s)
Aging/metabolism , Axons/metabolism , Central Nervous System/metabolism , Mitochondria/metabolism , STAT3 Transcription Factor/metabolism , Transcription, Genetic , Adenosine Triphosphate/metabolism , Animals , Ciliary Neurotrophic Factor/pharmacology , Electron Transport/drug effects , Female , Gene Deletion , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Regeneration/drug effects , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Domains , Protein Transport , Pyramidal Tracts/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , STAT3 Transcription Factor/chemistry , Structure-Activity Relationship , Subcellular Fractions/metabolism
14.
J Neurosci ; 34(46): 15347-55, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25392502

ABSTRACT

Mammalian target of rapamycin (mTOR) functions as a master sensor of nutrients and energy, and controls protein translation and cell growth. Deletion of phosphatase and tensin homolog (PTEN) in adult CNS neurons promotes regeneration of injured axons in an mTOR-dependent manner. However, others have demonstrated mTOR-independent axon regeneration in different cell types, raising the question of how broadly mTOR regulates axonal regrowth across different systems. Here we define the role of mTOR in promoting collateral sprouting of spared axons, a key axonal remodeling mechanism by which functions are recovered after CNS injury. Using pharmacological inhibition, we demonstrate that mTOR is dispensable for the robust spontaneous sprouting of corticospinal tract axons seen after pyramidotomy in postnatal mice. In contrast, moderate spontaneous axonal sprouting and induced-sprouting seen under different conditions in young adult mice (i.e., PTEN deletion or degradation of chondroitin proteoglycans; CSPGs) are both reduced upon mTOR inhibition. In addition, to further determine the potency of mTOR in promoting sprouting responses, we coinactivate PTEN and CSPGs, and demonstrate that this combination leads to an additive increase in axonal sprouting compared with single treatments. Our findings reveal a developmental switch in mTOR dependency for inducing axonal sprouting, and indicate that PTEN deletion in adult neurons neither recapitulates the regrowth program of postnatal animals, nor is sufficient to completely overcome an inhibitory environment. Accordingly, exploiting mTOR levels by targeting PTEN combined with CSPG degradation represents a promising strategy to promote extensive axonal plasticity in adult mammals.


Subject(s)
Axons/physiology , Brain Injuries/physiopathology , Nerve Regeneration/physiology , TOR Serine-Threonine Kinases/physiology , Aging/genetics , Aging/physiology , Animals , Brain Injuries/pathology , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/antagonists & inhibitors , Chondroitin Sulfate Proteoglycans/physiology , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Regeneration/drug effects , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Pyramidal Tracts/drug effects , Pyramidal Tracts/injuries , Pyramidal Tracts/physiology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
15.
World Neurosurg ; 81(1): 202.e9-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23838365

ABSTRACT

BACKGROUND: Intracranial carcinoid tumors belong to the class of neuroendocrine tumors and their incidence is extremely rare. The pathogenesis and clinical manifestations of carcinoid tumors of the skull base are outlined in this case report. CASE DESCRIPTION: A 61-year-old multimorbid woman presented with transient memory loss. Computed tomographic and magnetic resonance imaging scan of the brain demonstrated a left cavernous sinus mass extending into the infratemporal fossa. The lesion was biopsied using the Caldwell-Luc approach, and histology showed a low-grade neuroendocrine tumor. The tumor was subtotally resected with a neurosurgery/head and neck combined preauricular infratemporal and subtemporal extradural approaches to the cavernous sinus. Further histologic evaluation revealed that the tumor was of carcinoid differentiation with no other primary or metastatic sites detectable. CONCLUSION: Primary intracranial carcinoid tumors, though rare, should be included in the differential diagnosis of extradural and dural-based lesions.


Subject(s)
Carcinoid Tumor/pathology , Cavernous Sinus/pathology , Carcinoid Tumor/surgery , Cavernous Sinus/surgery , Cranial Fossa, Anterior/pathology , Craniotomy , Female , Follow-Up Studies , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Magnetic Resonance Imaging , Memory Disorders/etiology , Middle Aged , Neurosurgical Procedures , Tomography, X-Ray Computed , Treatment Outcome
16.
J Neurosci ; 33(34): 13882-7, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23966707

ABSTRACT

Injury to the CNS leads to formation of scar tissue, which is important in sealing the lesion and inhibiting axon regeneration. The fibrotic scar that comprises a dense extracellular matrix is thought to originate from meningeal cells surrounding the CNS. However, using transgenic mice, we demonstrate that perivascular collagen1α1 cells are the main source of the cellular composition of the fibrotic scar after contusive spinal cord injury in which the dura remains intact. Using genetic lineage tracing, light sheet fluorescent microscopy, and antigenic profiling, we identify collagen1α1 cells as perivascular fibroblasts that are distinct from pericytes. Our results identify collagen1α1 cells as a novel source of the fibrotic scar after spinal cord injury and shift the focus from the meninges to the vasculature during scar formation.


Subject(s)
Cicatrix/etiology , Fibroblasts/pathology , Pericytes/pathology , Spinal Cord Injuries/complications , Analysis of Variance , Animals , Antigens/genetics , Blood Vessels/metabolism , Blood Vessels/pathology , CD13 Antigens/metabolism , Cell Count , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Lectins , Leukocyte Common Antigens , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pericytes/metabolism , Piperidines/metabolism , Proteoglycans/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Spinal Cord Injuries/pathology , Time Factors , Uracil/analogs & derivatives , Uracil/metabolism
17.
Source Code Biol Med ; 8(1): 12, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23692932

ABSTRACT

BACKGROUND: Identifying and quantifying pathological changes in brain electrical activity is important for investigations of brain injury and neurological disease. An example is the development of epilepsy, a secondary consequence of traumatic brain injury. While certain epileptiform events can be identified visually from electroencephalographic (EEG) or electrocorticographic (ECoG) records, quantification of these pathological events has proved to be more difficult. In this study we developed MATLAB-based software that would assist detection of pathological brain electrical activity following traumatic brain injury (TBI) and present our MATLAB code used for the analysis of the ECoG. METHODS: Software was developed using MATLAB(™) and features of the open access EEGLAB. EEGgui is a graphical user interface in the MATLAB programming platform that allows scientists who are not proficient in computer programming to perform a number of elaborate analyses on ECoG signals. The different analyses include Power Spectral Density (PSD), Short Time Fourier analysis and Spectral Entropy (SE). ECoG records used for demonstration of this software were derived from rats that had undergone traumatic brain injury one year earlier. RESULTS: The software provided in this report provides a graphical user interface for displaying ECoG activity and calculating normalized power density using fast fourier transform of the major brain wave frequencies (Delta, Theta, Alpha, Beta1, Beta2 and Gamma). The software further detects events in which power density for these frequency bands exceeds normal ECoG by more than 4 standard deviations. We found that epileptic events could be identified and distinguished from a variety of ECoG phenomena associated with normal changes in behavior. We further found that analysis of spectral entropy was less effective in distinguishing epileptic from normal changes in ECoG activity. CONCLUSION: The software presented here was a successful modification of EEGLAB in the Matlab environment that allows detection of epileptiform ECoG signals in animals after TBI. The code allows import of large EEG or ECoG data records as standard text files and uses fast fourier transform as a basis for detection of abnormal events. The software can also be used to monitor injury-induced changes in spectral entropy if required. We hope that the software will be useful for other investigators in the field of traumatic brain injury and will stimulate future advances of quantitative analysis of brain electrical activity after neurological injury or disease.

18.
J Neurotrauma ; 29(5): 843-50, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-21815786

ABSTRACT

Previous studies on the therapeutic potential of agents affecting the dopamine system in traumatic brain injury (TBI) suggest that dopamine dysregulation may have a major role in behavioral deficit after TBI. We have previously identified that TBI reduces striatal dopamine synthesis and release at 7 days post-injury. In order to reverse deficits in the activity of tyrosine hydroxylase and dopamine release following TBI, we administered nicotine by intraperitoneal injection into rats for 7 days. Tyrosine hydroxylase activity assay demonstrated recovery of activity with nicotine treatment in injured animals. Microdialysis experiments using potassium stimulation to induce dopamine release showed recovery of dopamine release in injured animals receiving nicotine treatment. There was no effect of nicotine injection on extracellular dopamine metabolite levels, indicating the specificity of nicotine's effect on dopamine synthesis and release. Also, the activation of downstream postsynaptic molecule dopamine and cAMP regulated phosphoprotein 32 (DARPP-32) was assessed by Western blots for DARPP-32 phosphorylated at threonine 34 (pDARPP-32-T34). Injury reduced pDARPP-32-T34 levels, but nicotine treatment of injured animals did not alter pDARPP-32-T34 levels, indicating that postsynaptic dopamine signaling is complex, and the recovery of dopamine release may not be sufficient for the recovery of DARPP-32 activity.


Subject(s)
Brain Injuries/metabolism , Corpus Striatum/drug effects , Dopamine/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Microdialysis , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
19.
Brain Res ; 1369: 208-15, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21047500

ABSTRACT

There is increasing evidence that traumatic brain injury (TBI) induces hypofunction of the striatal dopaminergic system, the mechanisms of which are unknown. In this study, we analyzed the activity of striatal tyrosine hydroxylase (TH) in rats at 1 day, 1 week, and 4 weeks after TBI using the controlled cortical impact model. There were no changes in the level of TH phosphorylated at serine 40 site (pser40TH) at 1 day or 4 weeks. At 1 week, injured animals showed decreased pser40TH to 73.9±7.3% (p≤0.05) of sham injured rats. The in vivo TH activity assay showed no significant difference between injured and sham rats at 1 day. However, there was a decreased activity in injured rats to 62.1±8.2% (p≤0.05) and 68.8±6.2% (p≤0.05) of sham injured rats at 1 and 4 weeks, respectively. Also, the activity of protein kinase A, which activates TH, decreased at 1 week (injured: 87.8±2.8%, sham: 100.0±4.2%, p≤0.05). To study the release activity of dopamine after injury, potassium (80 mM)-evoked dopamine release was measured by microdialysis in awake, freely moving rats. Dialysates were collected and analyzed by high-performance liquid chromatography. There were no significant differences in dopamine release at 1 day and 4 weeks between sham and injured groups. At 1 week, there was a significant decrease (injured: 0.067±0.015 µM, sham: 0.127±0.027 µM, p≤0.05). These results suggest that TBI-induced dopamine neurotransmission deficits are, at least in part, attributable to deficits in TH activity.


Subject(s)
Brain Injuries/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Corpus Striatum/injuries , Male , Microdialysis , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...