Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Brain Res ; 1823: 148682, 2024 01 15.
Article in English | MEDLINE | ID: mdl-37989436

ABSTRACT

Gulf War Illness (GWI) is a chronic multi-symptom disorder affecting approximately 30 % of Veterans deployed to the Persian Gulf from 1990 to 91. GWI encompasses a wide spectrum of symptoms which frequently include neurological problems such as learning and memory impairments, mood disorders, and an increased incidence of neurodegenerative disorders. Combined exposure to both reversible and irreversible acetylcholinesterase (AChE) inhibitors has been identified as a likely risk factor for GWI. It is possible that the exposures affected connectivity in the brain, and it was also unknown whether this could benefit from treatment. We assessed chronic changes in dendritic architecture in granule cells of the dentate gyrus following exposure to pyridostigmine bromide (PB, 0.7 mg/kg), chlorpyrifos (CPF, 12.5 mg/kg), and N,N-diethyl-m-toluamide (DEET, 7.5 mg/kg) in male C57Bl/6J mice. We also evaluated the therapeutic effects of dietary administration for eight weeks of 1 % tert-butylhydroquinone (tBHQ), a Nrf2 activator, on long-term neuronal morphology. We found that Gulf War toxicant exposure resulted in reduced dendritic length and branching as well as overall spine density in dentate granule cells at 14 weeks post-exposure and that these effects were ameliorated by treatment with tBHQ. These findings indicate that Gulf War toxicant exposure results in chronic changes to dentate granule cell morphology and that modulation of neuroprotective transcription factors such as Nrf2 may improve long-term neuronal health in the hippocampus.


Subject(s)
NF-E2-Related Factor 2 , Persian Gulf Syndrome , Mice , Animals , Male , Acetylcholinesterase , Gulf War , Persian Gulf Syndrome/drug therapy , Persian Gulf Syndrome/chemically induced , Cholinesterase Inhibitors/pharmacology , Brain , Disease Models, Animal
2.
Cell Mol Neurobiol ; 42(5): 1453-1463, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33417143

ABSTRACT

Mild traumatic brain injuries can have long-term consequences that interfere with the life of the patient and impose a burden on our health care system. Oxidative stress has been identified as a contributing factor for the progression of neurodegeneration following TBI. A major source of oxidative stress for many veterans is cigarette smoking and second-hand smoke, which has been shown to have an effect on TBI recovery. To examine the potential influences of second-hand smoke during recovery from TBI, we utilized a mouse model of closed head injury, followed by repeated exposure to cigarette smoke and treatment with a neuroprotective antioxidant. We found that neither the mild injuries nor the smoke exposure produced axonal damage detectable with amino cupric silver staining. However, complexity in the dendritic arbors was significantly reduced after mild TBI plus smoke exposure. In the hippocampus, there were astrocytic responses, including Cyp2e1 upregulation, after the injury and tobacco smoke insult. This study provides useful context for the importance of lifestyle changes, such as reducing or eliminating cigarette smoking, during recovery from TBI.


Subject(s)
Brain Concussion , Brain Injuries, Traumatic , Tobacco Smoke Pollution , Animals , Astrocytes , Hippocampus , Humans , Mice
3.
Sci Rep ; 11(1): 23559, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876621

ABSTRACT

Traumatic brain injury (TBI) is a brain dysfunction without present treatment. Previous studies have shown that animals fed ketogenic diet (KD) perform better in learning tasks than those fed standard diet (SD) following brain injury. The goal of this study was to examine whether KD is a neuroprotective in TBI mouse model. We utilized a closed head injury model to induce TBI in mice, followed by up to 30 days of KD/SD. Elevated levels of ketone bodies were confirmed in the blood following KD. Cognitive and behavioral performance was assessed post injury and molecular and cellular changes were assessed within the temporal cortex and hippocampus. Y-maze and Novel Object Recognition tasks indicated that mTBI mice maintained on KD displayed better cognitive abilities than mTBI mice maintained on SD. Mice maintained on SD post-injury demonstrated SIRT1 reduction when compared with uninjured and KD groups. In addition, KD management attenuated mTBI-induced astrocyte reactivity in the dentate gyrus and decreased degeneration of neurons in the dentate gyrus and in the cortex. These results support accumulating evidence that KD may be an effective approach to increase the brain's resistance to damage and suggest a potential new therapeutic strategy for treating TBI.


Subject(s)
Brain Injuries, Traumatic/diet therapy , Diet, Ketogenic , Animals , Anxiety , Astrocytes/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/psychology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognitive Dysfunction/diet therapy , Cognitive Dysfunction/psychology , Disease Models, Animal , Head Injuries, Closed/blood , Head Injuries, Closed/diet therapy , Head Injuries, Closed/psychology , Hippocampus/metabolism , Hippocampus/pathology , Ketone Bodies/blood , Male , Maze Learning , Mice , Mice, Inbred ICR , Neurons/pathology , Recognition, Psychology , Sirtuin 1/metabolism
4.
J Biochem Mol Toxicol ; 35(12): e22913, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34528356

ABSTRACT

Gulf War Illness (GWI) is defined by the Centers for Disease Control and Prevention (CDC) as a multi-symptom illness having at least one symptom from two of three factors, which include: fatigue, mood-cognition problems, and musculoskeletal disorders. The cluster of long-term symptoms is unique to military personnel from coalition countries including United States, Australia, and the United Kingdom that served in Operation Desert Storm from 1990 to 1991. Reporting of these symptoms is much lower among soldiers deployed in other parts of the world like Bosnia during the same time period. The exact cause of GWI is unknown, but combined exposure to N,N-diethyl-m-toluamide (DEET), organophosphates like chlorpyrifos (CPF), and pyridostigmine bromide (PB), has been hypothesized as a potential mechanism. Mitochondrial dysfunction is known to occur in most neurodegenerative diseases that share symptoms with GWI and has therefore been implicated in GWI. Although exposure to these and other toxicants continues to be investigated as potential causes of GWI, their combined impact on mitochondrial physiology remains unknown. In this study, the effects of combined GWI toxicant exposure on mitochondrial function were determined in a commonly used and readily available immortalized cell line (N2a), whose higher rate of oxygen consumption resembles that of highly metabolic neurons in vivo. We report that combined exposure containing pesticide CPF 71 µM, insect repellants DEET 78 µM, and antitoxins PB 19 µM, causes profound mitochondrial dysfunction after a 4-h incubation resulting in decreased mitochondrial respiratory states in the absence of proapoptotic signaling, proton leak, or significant increase in reactive oxygen species production.


Subject(s)
Chlorpyrifos/toxicity , DEET/toxicity , Mitochondria/drug effects , Neuroblastoma/pathology , Persian Gulf Syndrome , Pyridostigmine Bromide/toxicity , War Exposure , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis/drug effects , Cell Line, Tumor , Humans , Mice , Mitochondria/metabolism , Oxygen Consumption/drug effects , Protein Kinases/metabolism , Signal Transduction/drug effects
5.
Life Sci ; 284: 119845, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34293396

ABSTRACT

AIMS: Approximately 30% of the nearly 700,000 Veterans who were deployed to the Gulf War from 1990 to 1991 have reported experiencing a variety of symptoms including difficulties with learning and memory, depression and anxiety, and increased incidence of neurodegenerative diseases. Combined toxicant exposure to acetylcholinesterase (AChE) inhibitors has been studied extensively as a likely risk factor. In this study, we modeled Gulf War exposure in male C57Bl/6J mice with simultaneous administration of three chemicals implicated as exposure hazards for Gulf War Veterans: pyridostigmine bromide, the anti-sarin prophylactic; chlorpyrifos, an organophosphate insecticide; and the repellant N,N-diethyl-m-toluamide (DEET). MAIN METHODS: Following two weeks of daily exposure, we examined changes in gene expression by whole transcriptome sequencing (RNA-Seq) with hippocampal isolates. Hippocampal-associated spatial memory was assessed with a Y-maze task. We hypothesized that genes important for neuronal health become dysregulated by toxicant-induced damage and that these detrimental inflammatory gene expression profiles could lead to chronic neurodegeneration. KEY FINDINGS: We found dysregulation of genes indicating a pro-inflammatory response and downregulation of genes associated with neuronal health and several important immediate early genes (IEGs), including Arc and Egr1, which were both reduced approximately 1.5-fold. Mice exposed to PB + CPF + DEET displayed a 1.6-fold reduction in preference for the novel arm, indicating impaired spatial memory. SIGNIFICANCE: Differentially expressed genes observed at an acute timepoint may provide insight into the pathophysiology of Gulf War Illness and further explanations for chronic neurodegeneration after toxicant exposure.


Subject(s)
Gene Expression Regulation , Gulf War , Hippocampus/metabolism , Animals , Down-Regulation/drug effects , Down-Regulation/genetics , Environmental Pollutants/toxicity , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Ontology , Hippocampus/drug effects , Male , Maze Learning , Mice, Inbred C57BL , Spatial Memory/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
6.
J Alzheimers Dis ; 79(4): 1443-1449, 2021.
Article in English | MEDLINE | ID: mdl-33459652

ABSTRACT

An estimated 5 million Americans are living with Alzheimer's disease (AD), and there is also a significant impact on caregivers, with an additional 16 million Americans providing unpaid care for individuals with AD and other dementias. These numbers are projected to increase in the coming years. While AD is still without a cure, continued research efforts have led to better understanding of pathology and potential risk factors that could be exploited to slow disease progression. A bidirectional relationship between sleep deprivation and AD has been suggested and is well supported by both human and animal studies. Even brief episodes of inadequate sleep have been shown to cause an increase in amyloidß and tau proteins, both well-established contributors toAD pathology. Sleep deprivation is also the most common consequence of post-traumatic stress disorder (PTSD). Patients with PTSD frequently present with sleep disturbances and also develop dementia at twice the rate of the general population accounting for a disproportionate representation of AD among U.S. Veterans. The goal of this review is to highlight the relationship triad between sleep deprivation, AD, and PTSD as well as their impact on molecular mechanisms driving AD pathology.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/pathology , Sleep Deprivation/etiology , Stress Disorders, Post-Traumatic/complications , Animals , Humans
7.
Brain Res ; 1746: 147019, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32681835

ABSTRACT

Traumatic brain injury has been described as the signature affliction of recent military conflicts and repetitive TBIs, particularly associated with military and athletic activities, typically result in more severe clinical effects. The majority of TBIs are mild, but they can result in long term cognitive deficits for which there is no effective treatment. One of the most significant deficits observed in TBI patients is memory loss, which suggests that TBI can induce pathological changes within the hippocampus. tert-butylhydroquinone (tBHQ) and pioglitazone activate the Nrf2 and PPAR-γ transcription factors, respectively, and both have been shown to be neuroprotective in model systems. We examined the morphological changes within the hippocampus following repetitive mild TBI and simultaneous treatment with both factors. We utilized a closed head injury mouse model with five injuries over 5 weeks. Our results showed marked morphological changes among the dendrites and dendritic spines of the neurons of the dentate gyrus of the hippocampus. We observed decreases in overall dendritic length, as well as in the quantity and density of dendritic spines. Our treatment partially ameliorated these effects, suggesting that the Nrf2 and PPAR-γ transcription factors may be important targets for future drug development in the treatment of TBI in humans.


Subject(s)
Brain Concussion/pathology , Dendritic Spines/pathology , Hippocampus/pathology , Animals , Brain Concussion/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydroquinones/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/agonists , Neuroprotective Agents/pharmacology , PPAR gamma/agonists , Pioglitazone/pharmacology
8.
J Neurotrauma ; 37(17): 1910-1917, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32292111

ABSTRACT

The worldwide incidence of traumatic brain injury (TBI) is ∼0.5% per year and the frequency is significantly higher among military personnel and athletes. Repetitive TBIs are associated with military and athletic activities, and typically involve more severe consequences. The majority of TBIs are mild; however, these still can result in long-term cognitive deficits, and there is currently no effective treatment. tert-Butylhydroquinone (tBHQ) and pioglitazone can activate the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and peroxisome proliferator-activated receptor-gamma (PPAR-γ) transcription factors, respectively, and each has been shown to be neuroprotective in various model systems. We examined behavioral and gene expression changes after repetitive mild TBI followed by simultaneous treatment with both factors. We used a repetitive closed head injury of mice involving five injuries with a 1-week interval between each TBI. We found that memory performance was significantly reduced by the injuries, unless the TBIs were followed by the tBHQ and pioglitazone administrations. Certain genes; for example, growth hormone and osteopontin, were downregulated by the injury, and this was reversed by the treatment, whereas other genes; for example, a tumor necrosis factor receptor, were upregulated by the injury and restored if the post-injury treatment was administered. Analysis of gene expression levels affected by the injury and/or the treatment point to potential mechanisms that could be exploited therapeutically.


Subject(s)
Brain Concussion/genetics , Brain Concussion/metabolism , Maze Learning/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR
9.
Sci Rep ; 10(1): 2206, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32042033

ABSTRACT

Traumatic brain injury (TBI) has been designated as a signature injury of modern military conflicts. Blast trauma, in particular, has come to make up a significant portion of the TBIs which are sustained in warzones. Though most TBIs are mild, even mild TBI can induce long term effects, including cognitive and memory deficits. In our study, we utilized a mouse model of mild blast-related TBI (bTBI) to investigate TBI-induced changes within the cortex and hippocampus. We performed rapid Golgi staining on the layer IV and V pyramidal neurons of the parietal cortex and the CA1 basilar tree of the hippocampus and quantified dendritic branching and distribution. We found decreased dendritic branching within both the cortex and hippocampus in injured mice. Within parietal cortex, this decreased branching was most evident within the middle region, while outer and inner regions resembled that of control mice. This study provides important knowledge in the study of how the shockwave associated with a blast explosion impacts different brain regions.


Subject(s)
Blast Injuries/pathology , Brain Concussion/pathology , CA1 Region, Hippocampal/pathology , Dendrites/pathology , Parietal Lobe/pathology , Animals , Armed Conflicts , Blast Injuries/etiology , Brain Concussion/etiology , CA1 Region, Hippocampal/cytology , Disease Models, Animal , Explosions , Golgi Apparatus/pathology , Humans , Male , Mice , Parietal Lobe/cytology , Pyramidal Cells/cytology , Pyramidal Cells/pathology
10.
BMC Neurosci ; 20(1): 44, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31438853

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a widespread public health problem and a signature injury of our military in modern conflicts. Despite the long-term effects of even mild brain injuries, an effective treatment remains elusive. Coffee and several of its compounds, including caffeine, have been identified as having neuroprotective effects in studies of neurodegenerative disease. Given the molecular similarities between TBI and neurodegenerative disease, we have devised a study to test a nanocoffee extract in the treatment of a mouse model of mild TBI. RESULTS: After a single injury and two subsequent injections of nanocoffee, we identified treatment as being associated with improved behavioral outcomes, favorable molecular signaling changes, and dendritic changes suggestive of improved neuronal health. CONCLUSIONS: We have identified coffee extracts as a potential viable multifaceted treatment approach to target the secondary injury associated with TBI.


Subject(s)
Brain Concussion/prevention & control , Coffea/chemistry , Maze Learning/drug effects , Plant Extracts/pharmacology , Proteins/metabolism , Recognition, Psychology/drug effects , Animals , Brain Concussion/pathology , Brain Concussion/psychology , Cerebral Cortex/metabolism , Dendrites/pathology , Hippocampus/metabolism , Male , Mice , Nanoparticles/chemistry , Neuroprotective Agents/pharmacology , Plant Extracts/chemistry , Sonication , Water/chemistry
11.
Exp Neurol ; 315: 9-14, 2019 05.
Article in English | MEDLINE | ID: mdl-30711646

ABSTRACT

Traumatic brain injury (TBI) continues to be a signature injury of our modern conflicts. Due in part to increased use of improvised explosive devices (IEDs), we have seen blast trauma make up a significant portion of TBIs sustained by deployed troops and civilians. In addition to the physical injury, TBI is also a common comorbidity with post-traumatic stress disorder (PTSD). Previous research suggests that PTSD is often associated with increased signaling within the amygdala, leading to feelings of fear and hyperarousal. In our study, we utilized a mouse model of mild blast-related TBI (bTBI) to investigate how TBI induces changes within the amygdala, which may provide favorable conditions for the development of PTSD. To do this, we performed Golgi staining on the stellate neurons of the basolateral amygdala and quantified dendritic amount, distribution, and complexity. We found increases in dendritic branching and in the density of dendritic spines in injured mice. Increases in spine density appears to be primarily due to increases in memory associated mushroom type dendritic spines. These changes observed in our bTBI model that are consistent with chronic stress models, suggesting an important connection between the physical changes induced by TBI and the neurological symptoms of PTSD.


Subject(s)
Amygdala/pathology , Blast Injuries/pathology , Brain Concussion/pathology , Nerve Net/pathology , Animals , Blast Injuries/psychology , Brain Concussion/psychology , Cell Size , Dendrites/pathology , Dendrites/ultrastructure , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , Disease Models, Animal , Male , Mice , Mice, Inbred ICR , Stress Disorders, Post-Traumatic/etiology , Stress Disorders, Post-Traumatic/psychology
12.
J Mol Neurosci ; 66(1): 114-120, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30105628

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most common adult-onset neuromuscular disease for which there is currently no effective treatment. The progression of ALS includes loss of motor neurons controlling the voluntary muscles, with much of this loss occurring at the neuromuscular junction. In an effort to better understand changes at the neuromuscular junction, we utilized the wobbler mouse model of motor neuron loss. We examined biceps and end plate morphologies and monitored selected factors involved in end plate function. Structural volumes were determined from 3D reconstructions that were generated for the end plates. Wobbler mice exhibited size reductions of both the muscle fibers and the end plates within the biceps, and we found that the end plate volumes were the most sensitive indicator of the degeneration. Concurrently, we found increases in calcitonin gene-related peptide (CGRP) and its receptor in wobbler biceps and spinal cord. We also found increases in gene expression of two acetylcholine receptors within the wobbler biceps, which may be a result of altered CGRP/CALCRL (calcitonin receptor-like receptor) expression.


Subject(s)
Motor Endplate/pathology , Neurodegenerative Diseases/pathology , Vesicular Transport Proteins/genetics , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Mice , Motor Endplate/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism
13.
J Neurotrauma ; 35(1): 139-148, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28701108

ABSTRACT

Worldwide head injuries are a growing problem. In the United States alone, 1.7 million people suffer a head injury each year. While most of these injuries are mild, head injury sufferers still sustain symptoms that can have major medical and economical impacts. Moreover, repetitive mild head injuries, like those observed in active military personnel and athletes, have demonstrated a more severe and long-term set of consequences. In an effort to better understand the delayed pathological changes following multiple mild head injuries, we used a mouse model of mild closed head injury (with no motor deficits observed by rotarod testing) and measured dendritic complexity at 30 days after injury and potentially related factors up to 60 days post-injury. We found an increase in TDP-43 protein at 60 days post-injury in the hippocampus and a decrease in autophagy factors three days post-injury. Alterations in dendritic complexity were neuronal subtype and location specific. Measurements of neurotropic factors suggest that an increase in complexity in the cortex may be a consequence of neuronal loss of the less connected neurons.


Subject(s)
Brain Concussion/metabolism , Brain Concussion/pathology , DNA-Binding Proteins/biosynthesis , Dendrites/pathology , Animals , Autophagy/physiology , Disease Models, Animal , Male , Mice , Protein Biosynthesis/physiology
14.
Sci Rep ; 7(1): 3735, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623327

ABSTRACT

Mild blast traumatic brain injury (B-TBI) induced lasting cognitive impairments in novel object recognition and less severe deficits in Y-maze behaviors. B-TBI significantly reduced the levels of synaptophysin (SYP) protein staining in cortical (CTX) and hippocampal (HIPP) tissues. Treatment with exendin-4 (Ex-4) delivered by subcutaneous micro-osmotic pumps 48 hours prior to or 2 hours immediately after B-TBI prevented the induction of both cognitive deficits and B-TBI induced changes in SYP staining. The effects of a series of biaxial stretch injuries (BSI) on a neuronal derived cell line, HT22 cells, were assessed in an in vitro model of TBI. Biaxial stretch damage induced shrunken neurites and cell death. Treatment of HT22 cultures with Ex-4 (25 to 100 nM), prior to injury, attenuated the cytotoxic effects of BSI and preserved neurite length similar to sham treated cells. These data imply that treatment with Ex-4 may represent a viable option for the management of secondary events triggered by blast-induced, mild traumatic brain injury that is commonly observed in militarized zones.


Subject(s)
Blast Injuries/metabolism , Brain Injuries, Traumatic/prevention & control , Cognitive Dysfunction/prevention & control , Exenatide/pharmacology , Hippocampus/metabolism , Synaptophysin/metabolism , Animals , Blast Injuries/pathology , Blast Injuries/prevention & control , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Line , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Hippocampus/pathology , Male , Mice
15.
Mol Cell Endocrinol ; 436: 41-9, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27394190

ABSTRACT

PANcreatic-DERived factor (PANDER) is a member of a superfamily of FAM3 proteins modulating glycemic levels by metabolic regulation of the liver and pancreas. The precise PANDER-induced hepatic signaling mechanism is still being elucidated and has been very complex due to the pleiotropic nature of this novel hormone. Our PANDER transgenic (PANTG) mouse displays a selective hepatic insulin resistant (SHIR) phenotype whereby insulin signaling is blunted yet lipogenesis is increased, a phenomena observed in type 2 diabetes. To examine the complex PANDER-induced mechanism of SHIR, we utilized quantitative mass spectrometry-based proteomic analysis using Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to reveal the global hepatic proteome differences within the PANTG under the metabolic states of fasting, fed and insulin-stimulated conditions. Proteomic analysis identified lipid metabolism as one of the top cellular functions differentially altered in all metabolic states. Differentially expressed proteins within the PANTG having a lipid metabolic role included ACC, ACLY, CD36, CYP7A1, FASN and SCD1. Central to the differentially expressed proteins involved in lipid metabolism was the predicted activation of the liver X receptor (LXR) pathway. Western analysis validated the increased hepatic expression of LXRα along with LXR-directed targets such as FASN and CYP7A1 within the PANTG liver. Furthermore, recombinant PANDER was capable of inducing LXR promoter activity in-vitro as determined by luciferase reporter assays. Taken together, PANDER strongly impacts hepatic lipid metabolism across metabolic states and may induce a SHIR phenotype via the LXR pathway.


Subject(s)
Cytokines/genetics , Lipogenesis , Liver X Receptors/metabolism , Liver/metabolism , Proteomics/methods , Animals , Blotting, Western , Fatty Acid Synthases/metabolism , Female , Gene Expression Profiling , Gene Regulatory Networks , Isotope Labeling , Lipogenesis/genetics , Liver X Receptors/genetics , Male , Mice, Inbred BALB C , Mice, Transgenic , Reproducibility of Results , Transcription, Genetic
16.
Mol Cell Endocrinol ; 413: 101-12, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26123584

ABSTRACT

PANcreatic-DERived factor (PANDER, FAM3B) has been shown to regulate glycemic levels via interactions with both pancreatic islets and the liver. Although PANDER is predominantly expressed from the endocrine pancreas, recent work has provided sufficient evidence that the liver may also be an additional tissue source of PANDER production. At physiological levels, PANDER is capable of disrupting insulin signaling and promoting increased hepatic glucose production. As shown in some animal models, strong expression of PANDER, induced by viral delivery within the liver, induces hepatic steatosis. However, no studies to date have explicitly characterized the transcriptional regulation of PANDER from the liver. Therefore, our investigation elucidated the nutrient and hormonal regulation of the hepatic PANDER promoter. Initial RNA-ligated rapid amplification of cDNA ends identified a novel transcription start site (TSS) approximately 26 bp upstream of the PANDER translational start codon not previously revealed in pancreatic ß-cell lines. Western evaluation of various murine tissues demonstrated robust expression in the liver and brain. Promoter analysis identified strong tissue-specific activity of the PANDER promoter in both human and murine liver-derived cell lines. The minimal element responsible for maximal promoter activity within hepatic cell lines was located between -293 and -3 of the identified TSS. PANDER promoter activity was inhibited by both insulin and palmitate, whereas glucose strongly increased expression. The minimal element was responsible for maximal glucose-responsive and basal activity. Co-transfection reporter assays, chromatin-immunoprecipitation (ChIP) and site-directed mutagenesis revealed that the carbohydrate-responsive element binding protein (ChREBP) increased PANDER promoter activity and interacted with the PANDER promoter. E-box 3 was shown to be critical for basal and glucose responsive expression. In summary, in-vitro and in-vivo glucose is a potent stimulator of the PANDER promoter within the liver and this response may be facilitated by ChREBP.


Subject(s)
Cytokines/biosynthesis , Glucose/metabolism , Insulin/metabolism , Liver/metabolism , Neoplasm Proteins/biosynthesis , Response Elements/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cytokines/genetics , Hep G2 Cells , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Liver/cytology , Mice , NIH 3T3 Cells , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...