Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Neurochem ; 159(4): 742-761, 2021 11.
Article in English | MEDLINE | ID: mdl-34216036

ABSTRACT

Protein arginine methyltransferases (PRMTs) are a family of enzymes involved in gene regulation and protein/histone modifications. PRMT8 is primarily expressed in the central nervous system, specifically within the cellular membrane and synaptic vesicles. Recently, PRMT8 has been described to play key roles in neuronal signaling such as a regulator of dendritic arborization, synaptic function and maturation, and neuronal differentiation and plasticity. Here, we examined the role of PRMT8 in response to hypoxia-induced stress in brain metabolism. Our results from liquid chromatography mass spectrometry, mitochondrial oxygen consumption rate, and protein analyses indicate that PRMT8(-/-) knockout mice presented with altered membrane phospholipid composition, decreased mitochondrial stress capacity, and increased neuroinflammatory markers, such as tumor necrosis factor alpha and ionized calcium binding adaptor molecule 1 (Iba1, a specific marker for microglia/macrophage activation) after hypoxic stress. Furthermore, adenovirus-based overexpression of PRMT8 reversed the changes in membrane phospholipid composition, mitochondrial stress capacity, and neuroinflammatory markers. Together, our findings establish PRMT8 as an important regulatory component of membrane phospholipid composition, short-term memory function, mitochondrial function, and neuroinflammation in response to hypoxic stress.


Subject(s)
Energy Metabolism/genetics , Hypoxia/genetics , Mitochondria/genetics , Mitochondria/metabolism , Neuroinflammatory Diseases/genetics , Protein-Arginine N-Methyltransferases/genetics , Animals , Calcium-Binding Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytokines/analysis , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Neural Stem Cells , Oxygen Consumption , Phospholipids/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Am J Physiol Heart Circ Physiol ; 319(5): H1044-H1050, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32946263

ABSTRACT

Cardiopulmonary arrest (CA) is the leading cause of death and disability in the United States. CA-induced brain injury is influenced by multifactorial processes, including reduced cerebral blood flow (hypoperfusion) and neuroinflammation, which can lead to neuronal cell death and functional deficits. We have identified serum and glucocorticoid-regulated kinase-1 (SGK1) as a new target in brain ischemia previously described in the heart, liver, and kidneys (i.e., diabetes and hypertension). Our data suggest brain SGK1 mRNA and protein expression (i.e., hippocampus), presented with hypoperfusion (low cerebral blood flow) and neuroinflammation, leading to further studies of the potential role of SGK1 in CA-induced brain injury. We used a 6-min asphyxia cardiac arrest (ACA) rat model to induce global cerebral ischemia. Modulation of SGK1 was implemented via GSK650394, a SGK1-specific inhibitor (1.2 µg/kg icv). Accordingly, treatment with GSK650394 attenuated cortical hypoperfusion and neuroinflammation (via Iba1 expression) after ACA, whereas neuronal survival was enhanced in the CA1 region of the hippocampus. Learning/memory deficits were observed 3 days after ACA but ameliorated with GSK650394. In conclusion, SGK1 is a major contributor to ACA-induced brain injury and neurological deficits, while inhibition of SGK1 with GSK650394 provided neuroprotection against CA-induced hypoperfusion, neuroinflammation, neuronal cell death, and learning/memory deficits. Our studies could lead to a novel, therapeutic target for alleviating brain injury following cerebral ischemia.NEW & NOTEWORTHY Upregulation of SGK1 exacerbates brain injury during cerebral ischemia. Inhibition of SGK1 affords neuroprotection against cardiac arrest-induced hypoperfusion, neuroinflammation, neuronal cell death, and neurological deficits.


Subject(s)
Brain Injuries/metabolism , Heart Arrest/complications , Immediate-Early Proteins/genetics , Memory , Protein Serine-Threonine Kinases/genetics , Animals , Benzoates/pharmacology , Brain Injuries/drug therapy , Brain Injuries/etiology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cerebrovascular Circulation , Hippocampus/drug effects , Hippocampus/metabolism , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
3.
Am J Physiol Heart Circ Physiol ; 312(1): H182-H188, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27864234

ABSTRACT

Sympathetic nervous system activity is increased after cardiopulmonary arrest, resulting in vasoconstrictor release from the perivascular sympathetic nerves of cerebral arteries. However, the pathophysiological function of the perivascular sympathetic nerves in the ischemic brain remains unclear. A rat model of global cerebral ischemia (asphyxial cardiac arrest, ACA) was used to investigate perivascular sympathetic nerves of cerebral arteries via bilateral decentralization (preganglionic lesion) of the superior cervical ganglion (SCG). Decentralization of the SCG 5 days before ACA alleviated hypoperfusion and afforded hippocampal neuroprotection and improved functional outcomes. These studies can provide further insights into the functional mechanism(s) of the sympathetic nervous system during ischemia. NEW & NOTEWORTHY: Interruption of the perivascular sympathetic nerves can alleviate CA-induced hypoperfusion and neuronal cell death in the CA1 region of the hippocampus to enhance functional learning and memory.


Subject(s)
Brain Ischemia/pathology , CA1 Region, Hippocampal/pathology , Cerebral Arteries/innervation , Neurons/pathology , Neurovascular Coupling , Superior Cervical Ganglion , Sympathectomy , Sympathetic Nervous System , Animals , Asphyxia/etiology , Brain/pathology , Brain/physiopathology , Brain Ischemia/physiopathology , CA1 Region, Hippocampal/physiopathology , Cell Death , Disease Models, Animal , Heart Arrest/complications , Learning/physiology , Male , Memory/physiology , Microscopy, Confocal , Neuroprotection , Rats , Rats, Sprague-Dawley
4.
Neuromolecular Med ; 24(2): 97-112, 2022 06.
Article in English | MEDLINE | ID: mdl-34019239

ABSTRACT

Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.


Subject(s)
Brain Injuries , Brain Ischemia , Heart Arrest , Animals , Brain Injuries/etiology , Brain Ischemia/complications , Heart Arrest/complications , Heart Arrest/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics
5.
Article in English | MEDLINE | ID: mdl-33445063

ABSTRACT

We previously discovered that palmitic acid methyl ester (PAME) is a potent vasodilator released from the sympathetic ganglion with vasoactive properties. Post-treatment with PAME can enhance cortical cerebral blood flow and functional learning and memory, while inhibiting neuronal cell death in the CA1 region of the hippocampus under pathological conditions (i.e. cerebral ischemia). Since mechanisms underlying PAME-mediated neuroprotection remain unclear, we investigated the possible neuroprotective mechanisms of PAME after 6 min of asphyxial cardiac arrest (ACA, an animal model of global cerebral ischemia). Our results from capillary-based immunoassay (for the detection of proteins) and cytokine array suggest that PAME (0.02 mg/kg) can decrease neuroinflammatory markers, such as ionized calcium binding adaptor molecule 1 (Iba1, a specific marker for microglia/macrophage activation) and inflammatory cytokines after cardiopulmonary resuscitation. Additionally, the mitochondrial oxygen consumption rate (OCR) and respiratory function in the hippocampal slices were restored following ACA (via Seahorse XF24 Extracellular Flux Analyzer) suggesting that PAME can ameliorate mitochondrial dysfunction. Finally, hippocampal protein arginine methyltransferase 1 (PRMT1) and PRMT8 are enhanced in the presence of PAME to suggest a possible pathway of methylated fatty acids to modulate arginine-based enzymatic methylation. Altogether, our findings suggest that PAME can provide neuroprotection in the presence of ACA to alleviate neuroinflammation and ameliorate mitochondrial dysfunction.


Subject(s)
Calcium-Binding Proteins/metabolism , Heart Arrest/drug therapy , Hippocampus/metabolism , Microfilament Proteins/metabolism , Mitochondria/metabolism , Neuroprotective Agents/administration & dosage , Palmitates/administration & dosage , Animals , Cardiopulmonary Resuscitation , Cerebrovascular Circulation/drug effects , Cytokines , Disease Models, Animal , Heart Arrest/metabolism , Hippocampus/drug effects , Male , Membrane Proteins/metabolism , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Oxygen Consumption , Palmitates/pharmacology , Protein-Arginine N-Methyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/metabolism
6.
Mol Neurobiol ; 57(3): 1716-1732, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31823198

ABSTRACT

The methylation of arginine residues by protein arginine methyltransferases (PRMTs) is a type of post-translational modification which is important for numerous cellular processes, including mRNA splicing, DNA repair, signal transduction, protein interaction, and transport. PRMTs have been extensively associated with various pathologies, including cancer, inflammation, and immunity response. However, the role of PRMTs has not been well described in vascular and neurological function. Aberrant expression of PRMTs can alter its metabolic products, asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA). Increased ADMA levels are recognized as an independent risk factor for cardiovascular disease and mortality. Recent studies have provided considerable advances in the development of small-molecule inhibitors of PRMTs to study their function under normal and pathological states. In this review, we aim to elucidate the particular roles of PRMTs in vascular and neuronal function as a potential target for cardiovascular and neurological diseases.


Subject(s)
Arginine/analogs & derivatives , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Arginine/metabolism , Humans , Methylation , Protein Processing, Post-Translational/physiology
7.
Article in English | MEDLINE | ID: mdl-32663656

ABSTRACT

Cardiac arrest causes neuronal damage and functional impairments that can result in learning/memory dysfunction after ischemia. We previously identified a saturated fatty acid (stearic acid methyl ester, SAME) that was released from the superior cervical ganglion (sympathetic ganglion). The function of stearic acid methyl ester is currently unknown. Here, we show that SAME can inhibit the detrimental effects of global cerebral ischemia (i.e. cardiac arrest). Treatment with SAME in the presence of asphyxial cardiac arrest (ACA) revived learning and working memory deficits. Similarly, SAME-treated hippocampal slices after oxygen-glucose deprivation inhibited neuronal cell death. Moreover, SAME afforded neuroprotection against ACA in the CA1 region of the hippocampus, reduced ionized calcium-binding adapter molecule 1 expression and inflammatory cytokines/chemokines, with restoration in mitochondria respiration. Altogether, we describe a unique and uncharted role of saturated fatty acids in the brain that may have important implications against cerebral ischemia.


Subject(s)
Asphyxia/drug therapy , CA1 Region, Hippocampal/metabolism , Heart Arrest/drug therapy , Neuroprotection/drug effects , Stearic Acids/pharmacology , Animals , Asphyxia/metabolism , Asphyxia/physiopathology , CA1 Region, Hippocampal/physiopathology , Disease Models, Animal , Heart Arrest/metabolism , Heart Arrest/physiopathology , Male , Rats , Rats, Sprague-Dawley
8.
Article in English | MEDLINE | ID: mdl-30514597

ABSTRACT

We previously discovered that palmitic acid methyl ester (PAME) is a potent vasodilator first identified and released from the superior cervical ganglion and remain understudied. Thus, we investigated PAME's role in modulating cerebral blood flow (CBF) and neuroprotection after 6 min of cardiac arrest (model of global cerebral ischemia). Our results suggest that PAME can enhance CBF under normal physiological conditions, while administration of PAME (0.02 mg/kg) immediately after cardiopulmonary resuscitation can also enhance CBF in vivo. Additionally, functional learning and spatial memory assessments (via T-maze) 3 days after asphyxial cardiac arrest (ACA) suggest that PAME-treated rats have improved learning and memory recovery versus ACA alone. Furthermore, improved neuronal survival in the CA1 region of the hippocampus were observed in PAME-treated, ACA-induced rats. Altogether, our findings suggest that PAME can enhance CBF, alleviate neuronal cell death, and promote functional outcomes in the presence of ACA.


Subject(s)
CA1 Region, Hippocampal/drug effects , Heart Arrest/prevention & control , Neuroprotective Agents/administration & dosage , Palmitates/administration & dosage , Animals , Cardiopulmonary Resuscitation , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Heart Arrest/therapy , Learning/drug effects , Neuroprotective Agents/pharmacology , Palmitates/pharmacology , Rats , Rats, Sprague-Dawley , Spatial Memory/drug effects
9.
J Vis Exp ; (131)2018 01 05.
Article in English | MEDLINE | ID: mdl-29364254

ABSTRACT

BACKGROUND: Evaluating mild to moderate cognitive impairment in a global cerebral ischemia (i.e. cardiac arrest) model can be difficult due to poor locomotion after surgery. For example, rats who undergo surgical procedures and are subjected to the Morris water maze may not be able to swim, thus voiding the experiment. New Method: We established a modified behavioral spontaneous alternation T-maze test. The major advantage of the modified T-maze protocol is its relatively simple design that is powerful enough to assess functional learning/memory after ischemia. Additionally, the data analysis is simple and straightforward. We used the T-maze to determine the rats' learning/memory deficits both in the presence or absence of mild to moderate (6 min) asphyxial cardiac arrest (ACA). Rats have a natural tendency for exploration and will explore the alternate arms in the T-maze, whereas hippocampal-lesioned rats tend to adopt a side-preference resulting in decreased spontaneous alternation ratios, revealing the hippocampal-related functional learning/memory in the presence or absence of ACA. RESULTS: ACA groups have higher side-preference ratios and lower alternations as compared to control. Comparison with Existing Method(s): The Morris water and Barnes maze are more prominent for assessing learning/memory function. However, the Morris water maze is more stressful than other mazes. The Barnes maze is widely used to measure reference (long-term) memory, while ACA-induced neurocognitive deficits are more closely related to working (short-term) memory. CONCLUSIONS: We have developed a simple, yet effective strategy to delineate working (short-term) memory via the T-maze in our global cerebral ischemia model (ACA).


Subject(s)
Heart Arrest/physiopathology , Memory Disorders/diagnosis , Animals , Heart Arrest/diagnosis , Heart Arrest/surgery , Male , Memory Disorders/physiopathology , Rats
10.
Neural Regen Res ; 13(3): 373-385, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29623912

ABSTRACT

Cerebral ischemia is one of the leading causes of morbidity and mortality worldwide. Although stroke (a form of cerebral ischemia)-related costs are expected to reach 240.67 billion dollars by 2030, options for treatment against cerebral ischemia/stroke are limited. All therapies except anti-thrombolytics (i.e., tissue plasminogen activator) and hypothermia have failed to reduce neuronal injury, neurological deficits, and mortality rates following cerebral ischemia, which suggests that development of novel therapies against stroke/cerebral ischemia are urgently needed. Here, we discuss the possible mechanism(s) underlying cerebral ischemia-induced brain injury, as well as current and future novel therapies (i.e., growth factors, nicotinamide adenine dinucleotide, melatonin, resveratrol, protein kinase C isozymes, pifithrin, hypothermia, fatty acids, sympathoplegic drugs, and stem cells) as it relates to cerebral ischemia.

SELECTION OF CITATIONS
SEARCH DETAIL