Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Cell Physiol ; 2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36036549

ABSTRACT

Alzheimer's disease (AD) is the leading cause of mortality, disability, and long-term care burden in the United States, with women comprising the majority of AD diagnoses. While AD-related dementia is associated with tau and amyloid beta accumulation, concurrent derangements in cerebral blood flow have been observed alongside these proteinopathies in humans and rodent models. The homeostatic production of nitric oxide synthases (NOS) becomes uncoupled in AD which leads to decreased NO-mediated vasodilation and oxidative stress via the production of peroxynitrite (ONOO-∙) superoxide species. Here, we investigate the role of the novel protein arginine methyltransferase 4 (PRMT4) enzyme function and its downstream product asymmetric dimethyl arginine (ADMA) as it relates to NOS dysregulation and cerebral blood flow in AD. ADMA (type-1 PRMT product) has been shown to bind NOS as a noncanonic ligand causing enzymatic dysfunction. Our results from RT-qPCR and protein analyses suggest that aged (9-12 months) female mice bearing tau- and amyloid beta-producing transgenic mutations (3xTg-AD) express higher levels of PRMT4 in the hippocampus when compared to age- and sex-matched C57BL6/J mice. In addition, we performed studies to quantify the expression and activity of different NOS isoforms. Furthermore, laser speckle contrast imaging analysis was indicative that 3xTg-AD mice have dysfunctional NOS activity, resulting in reduced production of NO metabolites, enhanced production of free-radical ONOO-, and decreased cerebral blood flow. Notably, the aforementioned phenomena can be reversed via pharmacologic PRMT4 inhibition. Together, these findings implicate the potential importance of PRMT4 signaling in the pathogenesis of Alzheimer's-related cerebrovascular derangement.

2.
J Pharmacol Exp Ther ; 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35680377

ABSTRACT

Oxidative damage is believed to play a major role in the etiology of many age-related diseases and the normal aging process. We previously reported that sulindac, a cyclooxygenase (COX) inhibitor and FDA approved anti-inflammatory drug, has chemoprotective activity in cells and intact organs by initiating a pharmacological preconditioning response, similar to ischemic preconditioning (IPC). The mechanism is independent of its COX inhibitory activity as suggested by studies on the protection of the heart against oxidative damage from ischemia/reperfusion and retinal pigmented endothelial (RPE) cells against chemical oxidative and UV damage . Unfortunately, sulindac is not recommended for long-term use due to toxicities resulting from its COX inhibitory activity. To develop a safer and more efficacious derivative of sulindac, we screened a library of indenes and identified a lead compound, MCI-100, that lacked significant COX inhibitory activity but displayed greater potency than sulindac to protect RPE cells against oxidative damage. MCI-100 also protected the intact rat heart against ischemia/reperfusion damage following oral administration. The chemoprotective activity of MCI-100 involves a preconditioning response similar to sulindac, which is supported by RNA sequencing data showing common genes that are induced or repressed by sulindac or MCI-100 treatment. Both sulindac and MCI-100 protection against oxidative damage may involve modulation of Wnt/ß-catenin signaling resulting in proliferation while inhibiting TGFb signaling leading to apoptosis. In summary MCI-100, is more active than sulindac in protecting cells against oxidative damage, but without significant NSAID activity, and could have therapeutic potential in treatment of diseases that involve oxidative damage. Significance Statement In this study, we describe a novel sulindac derivative, MCI-100, that lacks significant COX inhibitory activity, but is appreciably more potent than sulindac in protecting retinal pigmented epithelial (RPE) cells against oxidative damage. Oral administration of MCI-100 markedly protected the rat heart against ischemia/reperfusion damage. MCI-100 has potential therapeutic value as a drug candidate for age-related diseases by protecting cells against oxidative damage and preventing organ failure.

3.
J Biomed Sci ; 27(1): 19, 2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31907023

ABSTRACT

BACKGROUND: The FDA approved drug granulocyte-colony stimulating factor (G-CSF) displays anti-apoptotic and immunomodulatory properties with neurogenesis and angiogenic functions. It is known to demonstrate neuroprotective mechanisms against ischemic global stroke. Autophagy is a method for the degradation of intracellular components and in particular, unrestrained autophagy may lead to uncontrolled digestion of affected neurons as well as neuronal death in cerebral ischemia. Mitochondrial dynamics is vital for the regulation of cell survival and death after cerebral ischemia and an early upstream event in neuronal death is mitochondrial fission. We examined the pro-survival mechanisms of G-CSF against apoptosis resulting from autophagy, mitochondrial stress and endoplasmic reticulum (ER) stress. METHODS: Male Swiss Webster mice (20 weeks of age) were subjected to bilateral common carotid artery occlusion (BCAO) for 30 min. After occlusion, mice were injected with G-CSF (50 µg/kg) subcutaneously for 4 days. Behavioral analysis was carried out using the corner test and locomotor activity test before animals were sacrificed on day 4 or day 7. Key proteins in ER stress, autophagy and mitochondrial stress induced apoptosis were analyzed by immunoblotting. RESULTS: G-CSF improved neurological deficits and improved behavioral performance on corner and locomotor test. G-CSF binds to G-CSF receptors and its activation leads to upregulation of Akt phosphorylation (P-Akt) which in turn decreases levels of the ER stress sensor, GRP 78 and expression of proteins involved in ER stress apoptosis pathway; ATF6, ATF4, eIF2α, XBP1, Caspase 12 and CHOP. G-CSF treatment significantly decreased Beclin-1, an autophagy marker, and decreased mitochondrial stress biomarkers DRP1 and P53. G-CSF also up-regulated the mitochondrial fusion protein, OPA1 and anti-apoptotic protein Bcl-2 while down-regulating the pro-apoptotic proteins Bax, Bak and PUMA. CONCLUSIONS: G-CSF is an endogenous ligand in the CNS that has a dual activity that is beneficial both in reducing acute neuronal degeneration and adding to long-term plasticity after cerebral ischemia. G-CSF treatment exerts neuroprotective effects on damaged neurons through the suppression of the ER stress and mitochondrial stress and maintains cellular homeostasis by decreasing pro-apoptotic proteins and increasing of anti-apoptotic proteins.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Stroke , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Behavior, Animal/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mitochondrial Dynamics/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Stroke/drug therapy , Stroke/metabolism , Stroke/pathology
4.
J Biomed Sci ; 27(1): 99, 2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33126859

ABSTRACT

BACKGROUND: Global ischemia is the resulting effect of a cardiopulmonary arrest (CPA). Presently there is no effective treatment to address neurological deficits in patients who survived a CPA. Granulocyte-colony stimulating factor is a growth factor (G-CSF) with a plethora of beneficial effects, including neuroprotection. Clinical application of human G-CSF (hG-CSF) is limited due to its plasma half-life of 4 h. Therefore, novel approaches need to be investigated that would (1) enable prolonged manifestation of hG-CSF and (2) demonstrate G-CSF efficacy from studying the underlying protective mechanisms of hG-CSF. In our previous work, we used the self-complementary adeno-associated virus (stereotype2: scAAV2) as a vector to transfect the hG-CSF gene into the global ischemic brain of a mouse. As an extension of that work, we now seek to elucidate the protective mechanisms of hG-CSF gene therapy against endoplasmic reticulum induced stress, mitochondrial dynamics and autophagy in global ischemia. METHOD: A single drop of either AAV-CMV-hG-CSF or AAV-CMV-GFP was dropped into the conjunctival sac of the Swiss Webster mouse's left eye, 30-60 min after bilateral common artery occlusion (BCAO). The efficacy of the expressed hG-CSF gene product was analyzed by monitoring the expression levels of endoplasmic reticulum stress (ER), mitochondrial dynamics and autophagic proteins over 4- and 7-days post-BCAO in vulnerable brain regions including the striatum, overlying cortex (frontal brain regions) and the hippocampus (middle brain regions). Statistical analysis was performed using mostly One-Way Analysis of variance (ANOVA), except for behavioral analysis, which used Repeated Measures Two-Way ANOVA, post hoc analysis was performed using the Tukey test. RESULTS: Several biomarkers that facilitated cellular death, including CHOP and GRP78 (ER stress) DRP1 (mitochondrial dynamics) and Beclin 1, p62 and LC3-ll (autophagy) were significantly downregulated by hG-CSF gene transfer. hG-CSF gene therapy also significantly upregulated antiapoptotic Bcl2 while downregulating pro-apoptotic Bax. The beneficial effects of hG-CSF gene therapy resulted in an overall improvement in functional behavior. CONCLUSION: Taken together, this study has substantiated the approach of sustaining the protein expression of hG-CSF by eye drop administration of the hG-CSF gene. In addition, the study has validated the efficacy of using hG-CSF gene therapy against endoplasmic reticulum induced stress, mitochondrial dynamics and autophagy in global ischemia.


Subject(s)
Autophagy , Carotid Stenosis/physiopathology , Endoplasmic Reticulum Stress , Genetic Therapy/methods , Granulocyte Colony-Stimulating Factor/therapeutic use , Mitochondrial Dynamics , Stroke/therapy , Animals , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Male , Mice
5.
Adv Exp Med Biol ; 975 Pt 1: 193-205, 2017.
Article in English | MEDLINE | ID: mdl-28849455

ABSTRACT

Taurine, as a free amino acid, is found at high levels in many tissues including brain, heart and skeletal muscle and is known to demonstrate neuroprotective effects in a range of disease conditions including stroke and neurodegenerative disease. Using in vitro culture systems we have demonstrated that taurine can elicit protection against endoplasmic reticulum stress (ER stress) from glutamate excitotoxicity or from excessive reactive oxygen species in PC12 cells or rat neuronal cultures. In our current investigation we hypothesized that taurine treatment after stroke in the rat middle cerebral artery occlusion (MCAO) model would render protection against ER stress processes as reflected in decreased levels of expression of ER stress pathway components. We demonstrated that taurine elicited high level protection and inhibited both ATF-6 and IRE-1 ER stress pathway components. As ischemic stroke has a complex pathology it is likely that certain combination treatment approaches targeting multiple disease mechanisms may have excellent potential for efficacy. We have previously employed the partial NMDA antagonist DETC-MeSO to render protection against in vivo ischemic stroke using a rat cerebral ischemia model. Here we tested administration of subcutaneous administration of 0.56 mg/kg DETC-MeSO or 40 mg/kg of taurine separately or as combined treatment after a 120 min cerebral ischemia in the rat MCAO model. Neither drug alone demonstrated protection at the low doses employed. Remarkably however the combination of low dose DETC-MeSO plus low dose taurine conferred a diminished infarct size and an enhanced Neuroscore (reflecting decreased neurological deficit). Analysis of ER stress markers pPERK, peIF-2-alpha and cleaved ATF-6 all showed decreased expression demonstrating that all 3 ER stress pathways were inhibited concurrent with a synergistic protective effect by the post-stroke administration of this DETC-MeSO-taurine combination treatment.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Neuroprotective Agents/pharmacology , Stroke/metabolism , Stroke/pathology , Taurine/pharmacology , Animals , Disease Models, Animal , Ditiocarb/analogs & derivatives , Ditiocarb/pharmacology , Drug Synergism , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
6.
Adv Exp Med Biol ; 975 Pt 1: 207-216, 2017.
Article in English | MEDLINE | ID: mdl-28849456

ABSTRACT

Ischemic stroke is one of the greatest contributors to death and long term disability in developed countries. Ischemia induced brain injury arises due to excessive release of glutamate and involves cell death due to apoptosis and endoplasmic reticulum (ER) stress responses. Despite major research efforts there are currently no effective treatments for stroke. Taurine, a free amino acid found in high concentrations in many invertebrate and vertebrate systems can provide protection against a range of neurological disorders. Here we demonstrate that taurine can combat ER stress responses induced by glutamate or by hypoxia/re-oxygenation in neuronal cell lines and primary neuronal cultures. Taurine decreased expression of ER stress markers GRP78, CHOP, Bim and caspase 12 in primary neuronal cultures exposed to hypoxia/re-oxygenation. In analyzing individual ER stress pathways we demonstrated that taurine treatment can result in reduced levels of cleaved ATF6 and decreased p-IRE1 levels. We hypothesized that because of the complex nature of stroke a combination therapy approach may be optimal. For this reason we proceeded to test combination therapies using taurine plus low dose administration of an additional drug: either granulocyte colony stimulating factor (G-CSF) or sulindac a non-steroidal anti-inflammatory drug with potent protective functions through signaling via ischemic preconditioning pathways. When primary neurons were pretreated with 25 mM taurine and 25 ng/mL G-CSF for I hour and then exposed to high levels of glutamate, the taurine/G-CSF combination increased the protective effect against glutamate toxicity to 88% cell survival compared to 75% cell survival from an individual treatment with taurine or G-CSF alone. Pre-exposure of PC12 cells to 5 mM taurine or 25 µM sulindac did not protect the cells from hypoxia/re-oxygenation stress whereas at these concentrations the combination of taurine plus sulindac provided significant protection. In summary we have demonstrated the protective effect of taurine in primary neuronal cultures against hypoxia with re-oxygenation through inhibition of ATF6 or p-IRE-1 pathway but not the PERK pathway of ER stress. Furthermore the combinations of taurine plus an additional drug (either G-CSF or sulindac) can show enhanced potency for protecting PC 12 cells from glutamate toxicity or hypoxia/re-oxygenation through inhibition of ER stress responses.


Subject(s)
Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Taurine/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Glutamic Acid/toxicity , Granulocyte Colony-Stimulating Factor/pharmacology , PC12 Cells , Rats , Reperfusion Injury , Sulindac/pharmacology
7.
Proc Natl Acad Sci U S A ; 111(47): 16754-9, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385631

ABSTRACT

The retinal pigmented epithelial (RPE) layer is one of the major ocular tissues affected by oxidative stress and is known to play an important role in the etiology of age-related macular degeneration (AMD), the major cause of blinding in the elderly. In the present study, sulindac, a nonsteroidal antiinflammatory drug (NSAID), was tested for protection against oxidative stress-induced damage in an established RPE cell line (ARPE-19). Besides its established antiinflammatory activity, sulindac has previously been shown to protect cardiac tissue against ischemia/reperfusion damage, although the exact mechanism was not elucidated. As shown here, sulindac can also protect RPE cells from chemical oxidative damage or UV light by initiating a protective mechanism similar to what is observed in ischemic preconditioning (IPC) response. The mechanism of protection appears to be triggered by reactive oxygen species (ROS) and involves known IPC signaling components such as PKG and PKC epsilon in addition to the mitochondrial ATP-sensitive K(+) channel. Sulindac induced iNOS and Hsp70, late-phase IPC markers in the RPE cells. A unique feature of the sulindac protective response is that it involves activation of the peroxisome proliferator-activated receptor alpha (PPAR-α). We have also used low-passage human fetal RPE and polarized primary fetal RPE cells to validate the basic observation that sulindac can protect retinal cells against oxidative stress. These findings indicate a mechanism for preventing oxidative stress in RPE cells and suggest that sulindac could be used therapeutically for slowing the progression of AMD.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , PPAR alpha/physiology , Retinal Pigment Epithelium/drug effects , Sulindac/pharmacology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress
8.
Exp Physiol ; 101(9): 1185-1190, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-26990582

ABSTRACT

NEW FINDINGS: What is the topic of this review? Although the mammalian brain is exquisitely sensitive to hypoxia, some turtles survive complete anoxia by decreasing metabolic demand to match reduced energy supply. These animal models may help to elucidate neuroprotective mechanisms and reveal novel therapeutic targets for diseases of oxygen deprivation. What advances does it highlight? The mitogen-activated protein kinases (MAPKs) are part of the suite of adaptive responses to anoxia that are modulated by adenosine, a 'retaliatory metabolite' released in early anoxia. In anoxic turtle neurons, upregulation of pro-survival Akt and extracellular signal-regulated kinase 1/2 and suppression of the p38MAPK and JNK pathways promote cell survival, as does the anoxic- and post-anoxic upregulation of the antioxidant methionine sulfoxide reductase. Mammalian neurons undergo rapid degeneration when oxygen supply is curtailed. Neuroprotective pathways are induced during hypoxia/ischaemia, but their analysis is complicated by concurrent pathological events. Survival mechanisms can be investigated in anoxia-tolerant freshwater turtle species, which survive oxygen deprivation and post-anoxic reoxygenation by entrance into a state of reversible hypometabolism. Many energy-demanding processes are suppressed, including ion flux and neurotransmitter release, whereas cellular protective mechanisms, including certain mitogen-activated protein kinases (MAPKs), are upregulated. This superfamily of serine/threonine kinases plays a significant role in vital cellular processes, including cell proliferation, differentiation, stress adaptation and apoptosis in response to external stimuli. Here, we report that neuronal survival relies on robust co-ordination between the major signalling cascades, with upregulation of the pro-survival Akt and extracellular signal-regulated kinase 1/2 and suppression of the p38MAPK and JNK pathways. Other protective responses, including the upregulation of heat shock proteins and antioxidants, allow the turtle brain to abrogate potential oxidative stress upon reoxygenation.

9.
Amino Acids ; 46(1): 31-46, 2014 Jan.
Article in English | MEDLINE | ID: mdl-22903433

ABSTRACT

In the present era, investigators seek to find therapeutic interventions that are multifaceted in their mode of action. Such targets provide the most advantageous routes for addressing the multiplicity of pathophysiological avenues that lead to neuronal dysfunction and death observed in neurological disorders and neurodegenerative diseases. Taurine, an endogenous amino acid, exhibits a plethora of physiological functions in the central nervous system. In this review, we describe the mode of action of taurine and its clinical application in the neurological diseases: Alzheimer's disease, Parkinson's disease and Huntington's disease.


Subject(s)
Central Nervous System/metabolism , Neurodegenerative Diseases/metabolism , Taurine/metabolism , Animals , Central Nervous System/pathology , Humans , Neurodegenerative Diseases/mortality , Neurodegenerative Diseases/pathology
10.
J Neurochem ; 125(5): 774-89, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23286698

ABSTRACT

Many psychotropic substances used either for medications or illicit recreational purposes are able to produce an increase in extracellular serotonin (5HT) in the CNS. 5HT is well known to improve mood; however, only when the levels of its release are in an appropriate range. Excessive 5HT is harmful, and will generally result in serotonin syndrome. To date, clinical diagnosis of serotonin syndrome relies exclusively on observation of symptoms because of a lack of available laboratory tests. The goal of this study was to characterize the onset of the syndrome using laboratory settings to determine excessive 5HT-evoked neurological abnormalities. Experiments were carried out in rats with the syndrome being elicited by three groups of 5HT-promoting drugs: (i) (±)-3,4-methylenedioxymethamphetamine (MDMA); (ii) a combination of the monoamine oxidase inhibitor clorgyline with the 5HT precursor 5-hydroxytryptophan; (iii) clorgyline combined with the serotonin-selective reuptake inhibitor paroxetine. The onset of the syndrome was characterized by electroencephalography (EEG), tremor, and brain/plasma 5HT tests. We found that a mild syndrome was associated with reduced EEG amplitudes while a severe syndrome strongly with seizure-like EEG activity and increased tremor activity. The occurrence of the syndrome was confirmed with microdialysis, showing excessive 5HT efflux in brain dialysate and the increased concentration of unbound 5HT in the plasma. Our findings suggest that the syndrome onset can be revealed with EEG recording, measurements of tremor activity and changes of unbound 5HT concentration in the plasma.


Subject(s)
Brain/metabolism , Electroencephalography/drug effects , Serotonin Agents/toxicity , Serotonin Syndrome/metabolism , Serotonin Syndrome/physiopathology , Serotonin/metabolism , Animals , Brain/drug effects , Brain Chemistry/drug effects , Brain Chemistry/physiology , Electroencephalography/methods , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Male , Rats , Rats, Sprague-Dawley , Serotonin Syndrome/chemically induced
11.
Adv Exp Med Biol ; 775: 19-27, 2013.
Article in English | MEDLINE | ID: mdl-23392921

ABSTRACT

Taurine plays multiple roles in the CNS including acting as a -neuro-modulator, an osmoregulator, a regulator of cytoplasmic calcium levels, a trophic factor in development, and a neuroprotectant. In neurons taurine has been shown to prevent mitochondrial dysfunction and to protect against endoplasmic reticulum (ER) stress associated with neurological disorders. In cortical neurons in culture taurine protects against excitotoxicity through reversing an increase in levels of key ER signaling components including eIF-2-alpha and cleaved ATF6. The role of communication between the ER and mitochondrion is also important and examples are presented of protection by taurine against ER stress together with prevention of subsequent mitochondrial initiated apoptosis.


Subject(s)
Neuroprotective Agents/pharmacology , Taurine/pharmacology , Animals , Endoplasmic Reticulum Stress/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Nervous System Diseases/drug therapy , Nervous System Diseases/pathology , Neuroprotective Agents/therapeutic use , Receptors, Neurotransmitter/metabolism , Taurine/therapeutic use
12.
Adv Exp Med Biol ; 775: 167-75, 2013.
Article in English | MEDLINE | ID: mdl-23392933

ABSTRACT

Stroke is one of the leading causes of mortality and disability worldwide. There is no effective treatment for stroke despite extensive research. Taurine is a free amino acid which is present at high concentrations in a range of organs including the brain, heart, and retina in mammalian systems. It had been shown that taurine can significantly increase cell survival under stroke conditions using both in vivo and in vitro models. Recently, we have found that several agents including granulocyte colony-stimulating factor (G-CSF), a stem cell enhancer and facilitator;S-methyl-N-diethylthiolcarbamate sulfoxide (DETC-MeSO), an NMDA receptor partial antagonist; sulindac, a potent antioxidant; and taurine, a neuroprotectant and calcium regulator, are effective in protecting against stroke-induced neuronal injury when used alone or in combination in both animal and tissue/cell culture models. In this chapter, we demonstrate that taurine can protect human neuroblastoma cells measured by ATP assay under conditions of hypoxia or oxygen/glucose deprivation (OGD). In addition, we found that taurine exerts its protective function by suppressing the OGD-induced upregulation of endoplasmic reticulum (ER) stress markers and proapoptotic proteins. A model depicting the mode of action of taurine in protecting neuroblastoma cells under OGD conditions is presented.


Subject(s)
Cytoprotection/drug effects , Glucose/deficiency , Neuroblastoma/pathology , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Taurine/pharmacology , Activating Transcription Factor 4/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Humans , Membrane Proteins/metabolism , Models, Biological , Neuroblastoma/metabolism , Neuroprotective Agents/therapeutic use , Protein Phosphatase 1 , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Regulatory Factor X Transcription Factors , Stress, Physiological/drug effects , Transcription Factors/metabolism , Tumor Cells, Cultured
13.
Adv Exp Med Biol ; 776: 241-58, 2013.
Article in English | MEDLINE | ID: mdl-23392887

ABSTRACT

Taurine is an inhibitory neurotransmitter and is one of the most abundant amino acids present in the mammalian nervous system. Taurine has been shown to provide protection against neurological diseases, such as Huntington's disease, Alzheimer's disease, and stroke. Ischemic stroke is one of the leading causes of death and disability in the world. It is generally believed that ischemia-induced brain injury is largely due to excessive release of glutamate resulting in excitotoxicity and cell death. Despite extensive research, there are still no effective interventions for stroke. Recently, we have shown that taurine can provide effective protection against endoplasmic reticulum (ER) stress induced by excitotoxicity or oxidative stress in PC12 cell line or primary neuronal cell cultures. In this study, we employed hypoxia/reoxygenation conditions for primary cortical neuronal cell cultures as an in vitro model of stroke as well as the in vivo model of rat focal middle cerebral artery occlusion (MCAO). Our data showed that when primary neuronal cultures were first subjected to hypoxic conditions (0.3%, 24 h) followed by reoxygenation (21%, 24-48 h), the cell viability was greatly reduced. In the animal model of stroke (MCAO), we found that 2 h ischemia followed by 4 days reperfusion resulted in an infarct of 47.42 ± 9.86% in sections 6 mm from the frontal pole. Using taurine greatly increased cell viability in primary neuronal cell culture and decreased the infarct area of sections at 6 mm to 26.76 ± 6.91% in the MCAO model. Furthermore, levels of the ER stress protein markers GRP78, caspase-12, CHOP, and p-IRE-1 which were markedly increased in both the in vitro and in vivo models significantly declined after taurine administration, suggesting that taurine may exert neuroprotection functions in both models. Moreover, taurine could downregulate the ratio of cleaved ATF6 and full-length ATF6 in both models. In the animal model of stroke, taurine induced an upregulation of the Bcl-2/Bax ratio and downregulation of caspase-3 protein activity indicating that it attenuates apoptosis in the core of the ischemic infarct. Our results show not only taurine elicits neuroprotection through the activation of the ATF6 and the IRE1 pathways, but also it can reduce apoptosis in these models.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Infarction, Middle Cerebral Artery/pathology , Neurons/pathology , Neuroprotective Agents/pharmacology , Stroke/etiology , Taurine/pharmacology , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Brain Ischemia/pathology , Caspase 12/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Female , Hypoxia/complications , Hypoxia/pathology , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Male , Membrane Proteins/metabolism , Models, Biological , Neurons/drug effects , Neurons/enzymology , Neuroprotective Agents/therapeutic use , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stroke/drug therapy , Stroke/pathology , Taurine/therapeutic use , eIF-2 Kinase/metabolism
14.
Biomedicines ; 11(7)2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37509524

ABSTRACT

Carbamathione (Carb), an NMDA glutamate receptor partial antagonist, has potent neuroprotective functions against hypoxia- or ischemia-induced neuronal injury in cell- or animal-based stroke models. We used PC-12 cell cultures as a cell-based model and bilateral carotid artery occlusion (BCAO) for stroke. Whole-cell patch clamp recording in the mouse retinal ganglion cells was performed. Key proteins involved in apoptosis, endoplasmic reticulum (ER) stress, and heat shock proteins were analyzed using immunoblotting. Carb is effective in protecting PC12 cells against glutamate- or hypoxia-induced cell injury. Electrophysiological results show that Carb attenuates NMDA-mediated glutamate currents in the retinal ganglion cells, which results in activation of the AKT signaling pathway and increased expression of pro-cell survival biomarkers, e.g., Hsp 27, P-AKT, and Bcl2 and decreased expression of pro-cell death markers, e.g., Beclin 1, Bax, and Cleaved caspase 3, and ER stress markers, e.g., CHOP, IRE1, XBP1, ATF 4, and eIF2α. Using the BCAO animal stroke model, we found that Carb reduced the brain infarct volume and decreased levels of ER stress markers, GRP 78, CHOP, and at the behavioral level, e.g., a decrease in asymmetric turns and an increase in locomotor activity. These findings for Carb provide promising and rational strategies for stroke therapy.

15.
Amino Acids ; 43(2): 845-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22080215

ABSTRACT

Stroke (hypoxia) is one of the leading causes of mortality in the developed countries, and it can induce excessive glutamate release and endoplasmic reticulum (ER) stress. Taurine, as a free amino acid, present in high concentrations in a range of organs in mammals, can provide protection against multiple neurological diseases. Here, we present a study to investigate the potential protective benefits of taurine against ER stress induced by glutamate and hypoxia/reoxygenation in primary cortical neuronal cultures. We found that taurine suppresses the up-regulation of caspase-12 and GADD153/CHOP induced by hypoxia/reoxygenation, suggesting that taurine may exert a protective function against hypoxia/reoxygenation by reducing the ER stress. Moreover, taurine can down-regulate the ratio of cleaved ATF6 and full length ATF6, and p-IRE1 expression, indicating that taurine inhibits the ER stress induced by hypoxia/reoxygenation and glutamate through suppressing ATF6 and IRE1 pathways.


Subject(s)
Endoplasmic Reticulum Stress , Glutamic Acid/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Taurine/pharmacology , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 6/metabolism , Adenosine Triphosphate/metabolism , Animals , Caspase 12/metabolism , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutamic Acid/physiology , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/physiology , Phosphorylation , Primary Cell Culture , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Rats , Signal Transduction , Transcription Factor CHOP/metabolism
16.
Proc Natl Acad Sci U S A ; 106(46): 19611-6, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19884509

ABSTRACT

We have recently shown that sulindac, an anti-inflammatory drug, enhances the killing of cancer cells, but not normal cells, under conditions of oxidative stress, by mechanisms unrelated to its cyclooxygenase (COX) inhibition. To further study the protective effect of sulindac on cells exposed to oxidative stress, we have investigated the effect of sulindac on rat cardiac myocytes subjected to hypoxia/reoxygenation, as well as in a Langendorff model of myocardial ischemia. Low levels of sulindac could protect cardiac myocytes against cell death due to hypoxia/reoxygenation. In the Langendorff model sulindac provided significant protection against cell death, when the drug was fed to the animals before the removal of the heart for the Langendorff procedure. The results indicate that the primary protective effect of sulindac in these experiments does not involve its role as a COX inhibitor. Numerous signaling pathways have been implicated in myocardial protective mechanisms, many of which involve fluctuations in reactive oxygen species (ROS) levels. The results suggest that low levels of sulindac can induce a preconditioning response, triggered by ROS, to protect cardiac tissues against oxidative damage. Blocking of preconditioning pathways by administration of the PKC blocker chelerythrine abrogated the ischemic protection afforded by sulindac. Secondly, after feeding of sulindac, two end-effectors of preconditioning, inducible nitric oxide synthase and heat shock protein 27, were found to be markedly induced in the heart, dependent on PKC. These results suggest that sulindac may have therapeutic potential as a preconditioning agent.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Cytoprotection , Heart/drug effects , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/prevention & control , Sulindac/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Disease Models, Animal , HSP27 Heat-Shock Proteins/metabolism , Myocardium , Nitric Oxide Synthase Type II/metabolism , Protein Kinase C/metabolism , Rats , Reactive Oxygen Species/metabolism , Sulindac/therapeutic use
17.
Biochem Biophys Res Commun ; 402(4): 608-13, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20971073

ABSTRACT

Methionine sulfoxide reductase A (MsrA) is an enzyme that reverses oxidation of methionine in proteins. Using a MsrA gene knockout (MsrA(-/-)) mouse model, we have investigated the role of MsrA in the heart. Our data indicate that cellular contractility and cardiac function are not significantly changed in MsrA(-/-) mice if the hearts are not stressed. However, the cellular contractility, when stressed using a higher stimulation frequency (2Hz), is significantly reduced in MsrA(-/-) cardiac myocytes. MsrA(-/-) cardiac myocytes also show a significant decrease in contractility after oxidative stress using H(2)O(2). Corresponding changes in Ca(2+) transients are observed in MsrA(-/-) cardiomyocytes treated with 2Hz stimulation or with H(2)O(2). Electron microscope analyses reveal a dramatic morphological change of mitochondria in MsrA(-/-) mouse hearts. Further biochemical measurements indicate that protein oxidation levels in MsrA(-/-) mouse hearts are significantly higher than those in wild type controls. Our study demonstrates that the lack of MsrA in cardiac myocytes reduces myocardial cell's capability against stress stimulations resulting in a cellular dysfunction in the heart.


Subject(s)
Methionine Sulfoxide Reductases/deficiency , Mitochondria, Heart/physiology , Myocardial Contraction , Myocytes, Cardiac/physiology , Oxidative Stress , Stress, Mechanical , Animals , Calcium/metabolism , Methionine Sulfoxide Reductases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/enzymology , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure
18.
J Biomed Sci ; 17 Suppl 1: S1, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20804583

ABSTRACT

Taurine demonstrates multiple cellular functions including a central role as a neurotransmitter, as a trophic factor in CNS development, in maintaining the structural integrity of the membrane, in regulating calcium transport and homeostasis, as an osmolyte, as a neuromodulator and as a neuroprotectant. The neurotransmitter properties of taurine are illustrated by its ability to elicit neuronal hyperpolarization, the presence of specific taurine synthesizing enzyme and receptors in the CNS and the presence of a taurine transporter system. Taurine exerts its neuroprotective functions against the glutamate induced excitotoxicity by reducing the glutamate-induced increase of intracellular calcium level, by shifting the ratio of Bcl-2 and Bad ratio in favor of cell survival and by reducing the ER stress. The presence of metabotropic taurine receptors which are negatively coupled to phospholipase C (PLC) signaling pathway through inhibitory G proteins is proposed, and the evidence supporting this notion is also presented.


Subject(s)
Central Nervous System/metabolism , Taurine/metabolism , Animals , Calcium/metabolism , Glutamic Acid/metabolism , Homeostasis/physiology , Humans , Neuroprotective Agents/metabolism , Neurotransmitter Agents/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Neurotransmitter/metabolism , Signal Transduction/physiology , bcl-2-Associated X Protein/metabolism
19.
J Biomed Sci ; 17 Suppl 1: S17, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20804591

ABSTRACT

BACKGROUND: Taurine is a free amino acid present in high concentrations in a variety of organs of mammalians. As an antioxidant, taurine has been found to protect cells against oxidative stress, but the underlying mechanism is still unclear. METHODS: In this report, we present evidence to support the conclusion that taurine exerts a protective function against endoplasmic reticulum (ER) stress induced by H2O2 in PC 12 cells. Oxidative stress was introduced by exposure of PC 12 cells to 250 uM H2O2 for 4 hours. RESULTS: It was found that the cell viability of PC 12 cells decreased with an increase of H2O2 concentration ranging from approximately 76% cell viability at 100 uM H2O2 down to 18% at 500 uM H2O2. At 250 uM H2O2, cell viability was restored to 80% by taurine at 25 mM. Furthermore, H2O2 treatment also caused a marked reduction in the expression of Bcl-2 while no significant change of Bax was observed. Treatment with taurine restored the reduced expression of Bcl-2 close to the control level without any obvious effect on Bax. Furthermore, taurine was also found to suppress up-regulation of GRP78, GADD153/CHOP and Bim induced by H2O2, suggesting that taurine may also exert a protective function against oxidative stress by reducing the ER stress. CONCLUSION: In summary, taurine was shown to protect PC12 cells against oxidative stress induced by H2O2. ER stress was induced by oxidative stress and can be suppressed by taurine.


Subject(s)
Antioxidants/pharmacology , Endoplasmic Reticulum/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Taurine/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Survival/drug effects , Dose-Response Relationship, Drug , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/pharmacology , Membrane Proteins/metabolism , Oxidants/pharmacology , PC12 Cells/drug effects , PC12 Cells/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Transcription Factor CHOP/metabolism , bcl-2-Associated X Protein/metabolism
20.
J Biomed Sci ; 17 Suppl 1: S18, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20804592

ABSTRACT

BACKGROUND: Both taurine, an inhibitory neurotransmitter and granulocyte colony-stimulating factor (G-CSF), a growth factor, possess neuroprotective and neurotrophic properties in vitro. However, the mechanisms of their underlying neuroprotective effects are not fully understood. METHODS: In the present study, we investigated the potential protective benefits of taurine, G-CSF and the combination of taurine and G-CSF against excitotoxicity induced by glutamate in primary cortical neuronal cultures. RESULTS: 25 mM taurine, 25 ng/ml G-CSF and the combination of 25 mM taurine and 25 ng/ml G-CSF showed a protective effect reaching 75%, 75% and 88%, respectively. Furthermore, taurine exerted its protective effect through down-regulation of expression of GRP 78, CHOP, Bim and caspase 12. CONCLUSION: The results showed that all of these treatments, taurine, G-CSF and the combination of taurine and G-CSF, protected primary cortical neurons against excitotoxicity induced by glutamate. ER stress is suppressed by taurine after glutamate toxicity.


Subject(s)
Cell Death/drug effects , Cerebral Cortex/cytology , Glutamic Acid/toxicity , Granulocyte Colony-Stimulating Factor/pharmacology , Neurons/cytology , Neurons/drug effects , Taurine/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Caspase 12/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Female , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Neurons/physiology , Pregnancy , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor CHOP/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL