Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Biochem Mol Toxicol ; 35(5): e22744, 2021 May.
Article in English | MEDLINE | ID: mdl-33604948

ABSTRACT

Saturated fatty acids, whose circulating levels are markedly increased in the body, significantly affect the growth and functions of skeletal muscle. These fatty acids may exert a detrimental effect on the undifferentiated skeletal myoblasts that may adversely affect their differentiation. In the present study, the exposure of myoblasts to excess palmitic acid caused an elevation of tumor necrosis factor-α expression and an increase in reactive oxygen species levels consistent with the enhanced inflammation and oxidative stress. Various concentrations of palmitic acid significantly decreased the mitochondrial membrane potential, induced the programmed cell death by an increase in the caspase-3 expression, and DNA fragmentation in the myoblasts. These findings suggest that the increased concentrations of saturated fatty acid in the myoblasts increase lipotoxicity by increasing inflammation and oxidative stress, decreasing the mitochondrial function, and inducing apoptosis.


Subject(s)
Caspase 3/biosynthesis , Gene Expression Regulation/drug effects , Mitochondria, Muscle/metabolism , Myoblasts/metabolism , Oxidative Stress/drug effects , Palmitic Acid/toxicity , Tumor Necrosis Factor-alpha/biosynthesis , Cell Line , DNA Fragmentation/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Muscle/pathology , Myoblasts/pathology
2.
Article in English | MEDLINE | ID: mdl-31967962

ABSTRACT

Mild cognitive impairment (MCI) is a modifiable risk factor in progression of several diseases including dementia and type 2 diabetes. If cognitive impairments are not reversed at an early stage of appearance of symptoms, then the prolonged pathogenesis can lead to dementia and Alzheimer's disease (AD). Therefore, it is necessary to detect the risk factors and mechanism of prevention of cognitive dysfunction at an early stage of disease. Poor lifestyle, age, hyperglycemia, hypercholesterolemia, and inflammation are some of the major risk factors that contribute to cognitive and memory impairments in diabetic patients. Mild cognitive impairment was seen in those individuals of type 2 diabetes, who are on an unhealthy diet. Physical inactivity, frequent alcohol consumptions, and use of packed food products that provides an excess of cheap calories are found associated with cognitive impairment and depression in diabetic patients. Omega fatty acids (FAs) and polyphenol-rich foods, especially flavonoids, can reduce the bad effects of an unhealthy lifestyle; therefore, the consumption of omega FAs and flavonoids may be beneficial in maintaining normal cognitive function. These functional foods may improve cognitive functions by targeting many enzymes and molecules in cells chiefly through their anti-inflammatory, antioxidant, or signaling actions. Here, we provide the current concepts on the risk factors of cognitive impairments in type 2 diabetes and the mechanism of prevention, using omega FAs and bioactive compounds obtained from fruits and vegetables. The knowledge derived from such studies may assist physicians in managing the health care of patients with cognitive difficulties.


Subject(s)
Cognitive Dysfunction/etiology , Diabetes Mellitus, Type 2/complications , Functional Food , Age Factors , Cognitive Dysfunction/prevention & control , Diabetes Mellitus, Type 2/psychology , Diet , Humans , Life Style , Risk Factors
3.
Antioxidants (Basel) ; 8(7)2019 Jun 29.
Article in English | MEDLINE | ID: mdl-31261915

ABSTRACT

Flavonoids, one of the most abundant phytochemicals in a diet rich in fruits and vegetables, have been recognized as possessing anti-proliferative, antioxidant, anti-inflammatory, and estrogenic activities. Numerous cellular and animal-based studies show that flavonoids can function as antioxidants by preventing DNA damage and scavenging reactive oxygen radicals, inhibiting formation of DNA adducts, enhancing DNA repair, interfering with chemical damage by induction of Phase II enzymes, and modifying signaling pathways. Recent evidence also shows their ability to regulate the immune system. However, findings from clinical trials have been mixed with no clear consensus on dose, frequency, or type of flavonoids best suited to elicit many of the beneficial effects. Delivery of these bioactive compounds to their biological targets through "targeted designed" food processing strategies is critical to reach effective concentration in vivo. Thus, the identification of novel approaches that optimize flavonoid bioavailability is essential for their successful clinical application. In this review, we discuss the relevance of increasing flavonoid bioavailability, by agricultural engineering and "targeted food design" in the context of the immune system and cancer.

4.
J Biochem Mol Toxicol ; 33(8): e22337, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30958623

ABSTRACT

Elevated levels of saturated fatty acids show a strong cytotoxic effect in liver cells. Sirtuin 3 (SIRT3), a mitochondrially localized member of NAD+ -dependent deacetylase has been shown to protect hepatocytes against the oxidative stress. The role of SIRT3 on the cytotoxicity caused by fatty acids in liver cells is not fully understood. The aim of this study was to evaluate the expression level of SIRT3, oxidative stress, and mitochondrial impairments in human hepatoma HepG2 cells exposed to palmitic acid (PA). Our results showed that PA treatment caused the deposition of lipid droplets and resulted in an increased expression of tumor necrosis factor-α in a dose-dependent manner. Excessive accumulation of PA induces the reactive oxygen species formation and apoptosis while dissipating the mitochondrial transmembrane potential. The level of SIRT3 expression in both nuclear and mitochondrial fractions in HepG2 cells was decreased with the increase in PA concentrations. However, in the cytosolic fraction, the SIRT3 was undetectable. In conclusion, our results showed that PA caused an increase in inflammation and oxidative stress in HepG2 cells. The exposure of PA also resulted in the decline in transmembrane potential and an increase in apoptosis. The underexpression of nuclear and mitochondrial SIRT3 by PA suggests that the PA target the process that regulates the stress-related gene expression and mitochondrial functions.


Subject(s)
Apoptosis/drug effects , Down-Regulation/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Palmitic Acid/pharmacology , Sirtuin 3/metabolism , Hep G2 Cells , Humans , Lipid Metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Immunology ; 144(4): 611-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25322815

ABSTRACT

Monocytes, key components of the immune system, are a heterogeneous population comprised of classical monocytes (CD16(-) ) and non-classical monocytes (CD16(+) ). Monocytes are short lived and undergo spontaneous apoptosis, unless stimulated. Dysregulation of monocyte numbers contribute to the pathophysiology of inflammatory diseases, yet the contribution of each subset remains poorly characterized. Protein kinase C (PKC) family members are central to monocyte biology; however, their role in regulating lifespan and immune function of CD16(-) and CD16(+) monocytes has not been studied. Here, we evaluated the contribution of PKCδ and PKCε in the lifespan and immune response of both monocyte subsets. We showed that CD16(+) monocytes are more susceptible to spontaneous apoptosis because of the increased caspase-3, -8 and -9 activities accompanied by higher kinase activity of PKCδ. Silencing of PKCδ reduced apoptosis in both CD16(+) and CD16(-) monocytes. CD16(+) monocytes express significantly higher levels of PKCε and produce more tumour necrosis factor-α in CD16(+) compared with CD16(-) monocytes. Silencing of PKCε affected the survival and tumour necrosis factor-α production. These findings demonstrate a complex network with similar topography, yet unique regulatory characteristics controlling lifespan and immune response in each monocyte subset, helping define subset-specific coordination programmes controlling monocyte function.


Subject(s)
Monocytes/enzymology , Monocytes/immunology , Protein Kinase C-delta/immunology , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/immunology , Protein Kinase C-epsilon/metabolism , Apoptosis , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Survival , Cells, Cultured , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/immunology , Humans , Monocytes/classification , Monocytes/pathology , Protein Kinase C-delta/genetics , Protein Kinase C-epsilon/genetics , RNA Interference , Receptors, IgG/deficiency , Receptors, IgG/immunology , Signal Transduction , Time Factors , Transfection , Tumor Necrosis Factor-alpha/blood
6.
Int J Mol Sci ; 14(9): 17664-79, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23989609

ABSTRACT

Acute and chronic inflammation is characterized by increased reactive oxygen species (ROS) production, dysregulation of mitochondrial metabolism and abnormal immune function contributing to cardiovascular diseases and sepsis. Clinical and epidemiological studies suggest potential beneficial effects of dietary interventions in inflammatory diseases but understanding of how nutrients work remains insufficient. In the present study, we evaluated the effects of apigenin, an anti-inflammatory flavonoid abundantly found in our diet, in endothelial cells during inflammation. Here, we show that apigenin reduced lipopolysaccharide (LPS)-induced apoptosis by decreasing ROS production and the activity of caspase-3 in endothelial cells. Apigenin conferred protection against LPS-induced mitochondrial dysfunction and reestablished normal mitochondrial complex I activity, a major site of electron leakage and superoxide production, suggesting its ability to modulate endothelial cell metabolic function during inflammation. Collectively, these findings indicate that the dietary compound apigenin stabilizes mitochondrial function during inflammation preventing endothelial cell damage and thus provide new translational opportunities for the use of dietary components in the prevention and treatment of inflammatory diseases.


Subject(s)
Apigenin/pharmacology , Caspase 3/metabolism , Inflammation/chemically induced , Lipopolysaccharides/pharmacology , Animals , Apoptosis/drug effects , Cattle , Cell Survival/drug effects , Cells, Cultured , Inflammation/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
7.
Proc Natl Acad Sci U S A ; 110(24): E2153-62, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23697369

ABSTRACT

Flavonoids constitute the largest class of dietary phytochemicals, adding essential health value to our diet, and are emerging as key nutraceuticals. Cellular targets for dietary phytochemicals remain largely unknown, posing significant challenges for the regulation of dietary supplements and the understanding of how nutraceuticals provide health value. Here, we describe the identification of human cellular targets of apigenin, a flavonoid abundantly present in fruits and vegetables, using an innovative high-throughput approach that combines phage display with second generation sequencing. The 160 identified high-confidence candidate apigenin targets are significantly enriched in three main functional categories: GTPase activation, membrane transport, and mRNA metabolism/alternative splicing. This last category includes the heterogeneous nuclear ribonucleoprotein A2 (hnRNPA2), a factor involved in splicing regulation, mRNA stability, and mRNA transport. Apigenin binds to the C-terminal glycine-rich domain of hnRNPA2, preventing hnRNPA2 from forming homodimers, and therefore, it perturbs the alternative splicing of several human hnRNPA2 targets. Our results provide a framework to understand how dietary phytochemicals exert their actions by binding to many functionally diverse cellular targets. In turn, some of them may modulate the activity of a large number of downstream genes, which is exemplified here by the effects of apigenin on the alternative splicing activity of hnRNPA2. Hence, in contrast to small-molecule pharmaceuticals designed for defined target specificity, dietary phytochemicals affect a large number of cellular targets with varied affinities that, combined, result in their recognized health benefits.


Subject(s)
Apigenin/pharmacology , Cell Membrane/metabolism , GTP Phosphohydrolases/metabolism , RNA, Messenger/metabolism , Alternative Splicing/drug effects , Amino Acid Sequence , Apigenin/metabolism , Base Sequence , Biological Transport/drug effects , Cell Line, Tumor , Diet , Enzyme Activation/drug effects , Flavonoids/metabolism , Flavonoids/pharmacology , GTP Phosphohydrolases/genetics , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Humans , Molecular Sequence Data , Peptide Library , Protein Binding , Protein Multimerization/drug effects , RNA Stability/drug effects , RNA Transport/drug effects , RNA, Messenger/genetics , Sequence Analysis, DNA
8.
Biochem Pharmacol ; 84(12): 1571-80, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22985621

ABSTRACT

Apigenin, an abundant plant flavonoid, exhibits anti-proliferative and anti-carcinogenic activities through mechanisms yet not fully defined. In the present study, we show that the treatment of leukemia cells with apigenin resulted in the induction of DNA damage preceding the activation of the apoptotic program. Apigenin-induced DNA damage was mediated by p38 and protein kinase C-delta (PKCδ), yet was independent of reactive oxygen species or caspase activity. Treatment of monocytic leukemia cells with apigenin induced the phosphorylation of the ataxia-telangiectasia mutated (ATM) kinase and histone H2AX, two key regulators of the DNA damage response, without affecting the ataxia-telangiectasia mutated and Rad-3-related (ATR) kinase. Silencing and pharmacological inhibition of PKCδ abrogated ATM and H2AX phosphorylation, whereas inhibition of p38 reduced H2AX phosphorylation independently of ATM. We established that apigenin delayed cell cycle progression at G1/S and increased the number of apoptotic cells. In addition, genome-wide mRNA analyses showed that apigenin-induced DNA damage led to down-regulation of genes involved in cell-cycle control and DNA repair. Taken together, the present results show that the PKCδ-dependent activation of ATM and H2AX define the signaling networks responsible for the regulation of DNA damage promoting genome-wide mRNA alterations that result in cell cycle arrest, hence contributing to the anti-carcinogenic activities of this flavonoid.


Subject(s)
Apigenin/pharmacology , Cell Cycle Proteins/metabolism , Cell Cycle , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Down-Regulation , Histones/metabolism , Protein Kinase C-delta/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Caspase 3/metabolism , Cell Line, Tumor , Humans , Phosphorylation , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Biochim Biophys Acta ; 1800(7): 646-54, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20382204

ABSTRACT

BACKGROUND: Ceramides are intracellular lipid mediator implicated in various cellular responses, including oxidative stress and programmed cell death. Studies demonstrated strong links between ceramide and the mitochondria in the regulation of apoptosis. However, the mechanism of apoptosis induced by ceramides is not fully understood. The present study delineates importance of the redox state of cytochrome c for release of cytochrome c and apoptosis of human mammary adenocarcinoma MCF-7 and MDA-MB-231 cells induced by ceramides. METHODS: The study uses MCF-7 and MDA-MB-231 cells, isolated mitochondria, submitochondrial particles, and oxidized and reduced cytochrome c. Methods used include flow cytometry, immunoblotting, spectroscopy, and respirometry. RESULTS: We show that ceramides induce mitochondrial oxidative stress and release of cytochrome c from the mitochondria of these cells. Our findings show that ceramides react with oxidized cytochrome c whereas reduced cytochrome c does not react with ceramides. We also show that oxidized cytochrome c reacted with ceramides exerts lower reducibility and function to support mitochondrial respiration. Furthermore, our data show that glutathione protects cytochrome c of reacting with ceramides by increasing the reduced state of cytochrome c. CONCLUSIONS: Ceramides induce oxidative stress and apoptosis in human mammary adenocarcinoma cells by interacting with oxidized cytochrome c leading to the release of cytochrome c from the mitochondria. Our findings suggest a novel mechanism for protective role of glutathione. GENERAL SIGNIFICANCE: Our study suggests that the redox state of cytochrome c is important in oxidative stress and apoptosis induced by ceramides.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/drug effects , Breast Neoplasms/pathology , Ceramides/pharmacology , Cytochromes c/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cytochromes c/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Female , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Oxidation-Reduction
10.
J Immunol ; 184(10): 5582-8, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20385876

ABSTRACT

Differences in CD8(+)CD57(-) and CD8(+)CD57(+) lymphocyte lifespan have been documented. Lower numbers and shorter lifespan are characteristic of CD8(+)CD57(+) in normal individuals. However, CD8(+)CD57(+) are expanded in certain disease states including T cell large granular leukemia and other hematologic malignancies. The mechanisms responsible for the differences in CD8(+)CD57(-) and CD8(+)CD57(+) lifespan remain elusive. In this study, we demonstrate that the small heat shock protein (Hsp) 27 is a key regulator of CD8(+)CD57(+) lymphocyte lifespan. We found that Hsp27 expression is significantly lower in CD8(+)CD57(+) than in CD8(+)CD57(-) lymphocytes. In contrast, Hsp60 and Hsp70 are expressed at comparable levels. Unlike other antiapoptotic Bcl-2-like molecules, the expression of Hsp27 tightly correlates with CD8(+)CD57(+) and CD8(+)CD57(-) lifespan. We demonstrate that Hsp27 overexpression in CD8(+)CD57(+) lymphocytes to levels found normally in CD8(+)CD57(-) lymphocytes decreased apoptosis. Accordingly, silencing of Hsp27 in CD8(+)CD57(-) lymphocytes increased apoptosis. Collectively these results demonstrate that Hsp27 is a critical regulator of normal CD8(+)CD57(+) lifespan supporting its use as a marker of lifespan in this lineage, and suggest a mechanism responsible for the decreased apoptosis and clonal expansion characteristic of certain disease states.


Subject(s)
CD57 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , HSP27 Heat-Shock Proteins/physiology , Apoptosis/genetics , Apoptosis/immunology , CD57 Antigens/genetics , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Clone Cells , Down-Regulation/genetics , Down-Regulation/immunology , Gene Expression Regulation/immunology , Genetic Markers/genetics , HSP27 Heat-Shock Proteins/biosynthesis , HSP27 Heat-Shock Proteins/genetics , Humans , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
J Innate Immun ; 2(3): 204-15, 2010.
Article in English | MEDLINE | ID: mdl-20375558

ABSTRACT

Monocytes and macrophages are central cells of the innate immune system, responsible for defending against diverse pathogens. While they originate from a common myeloid precursor and share functions in innate immunity, each has a very distinct life span finely tuned by the apoptotic caspases. Normally, circulating monocytes are short-lived and undergo spontaneous apoptosis on a daily basis. Macrophages, however, have a longer life span. In chronic inflammatory diseases and, as recently recognized, in the tumor microenvironment, the inhibition of the apoptotic program promotes monocyte survival contributing to the accumulation of macrophages and the persistence of an inflammatory milieu. A complex network of differentiation factors and inflammatory stimuli determine monocyte/macrophage life span by blocking the apoptotic pathway and activating a myriad of survival pathways. Our understanding of apoptosis has flourished over the last decade, and its relevance in the regulation of the immune system is now indisputable. Nevertheless, how the complicated networks of survival and apoptotic regulators are integrated to determine cellular life span remains elusive. This review summarizes the contribution of the caspases and their regulators in monocyte/macrophage cell fate and discusses how these molecules orchestrate the initiation, maintenance, and resolution of inflammation. More provocatively, we discuss possible strategies to control inflammation by manipulating leukocyte life span.


Subject(s)
Autoimmune Diseases/immunology , Immunity, Innate , Macrophages/metabolism , Monocytes/metabolism , Neoplasms/immunology , Animals , Apoptosis/immunology , Autoimmune Diseases/pathology , Caspases/immunology , Caspases/metabolism , Cell Communication/immunology , Cell Survival/immunology , Gene Expression Regulation , Humans , Inflammation , Inhibitor of Apoptosis Proteins/immunology , Inhibitor of Apoptosis Proteins/metabolism , Macrophages/immunology , Monocytes/immunology , Neoplasms/pathology , Signal Transduction
12.
PLoS One ; 4(9): e7021, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19753113

ABSTRACT

Chronic pain is a major clinical problem and opiates are often the only treatment, but they cause significant problems ranging from sedation to deadly respiratory depression. Resiniferatoxin (RTX), a potent agonist of Transient Receptor Potential Vanilloid 1 (TRPV1), causes a slow, sustained and irreversible activation of TRPV1 and increases the frequency of spontaneous excitatory postsynaptic currents, but causes significant depression of evoked EPSCs due to nerve terminal depolarization block. Intrathecal administration of RTX to rats in the short-term inhibits nociceptive synaptic transmission, and in the long-term causes a localized, selective ablation of TRPV1-expressing central sensory nerve terminals leading to long lasting analgesia in behavioral models. Since RTX actions are selective for central sensory nerve terminals, other efferent functions of dorsal root ganglion neurons can be preserved. Preventing nociceptive transmission at the level of the spinal cord can be a useful strategy to treat chronic, debilitating and intractable pain.


Subject(s)
Analgesia , Spinal Cord/drug effects , Spinal Cord/metabolism , TRPV Cation Channels/metabolism , Animals , Capsaicin/pharmacology , Diterpenes/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Ganglia, Spinal/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Pain , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
13.
Int J Biochem Cell Biol ; 41(10): 2015-24, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19460457

ABSTRACT

Overexpression of alpha-synuclein and oxidative stress has been implicated in the neuronal cell death in Parkinson's disease. Alpha-synuclein associates with mitochondria and excessive accumulation of alpha-synuclein causes impairment of mitochondrial functions. However, the mechanism of mitochondrial impairment caused by alpha-synuclein is not fully understood. We recently reported that alpha-synuclein associates with mitochondria and that overexpression of alpha-synuclein causes nitration of mitochondrial proteins and release of cytochrome c from the mitochondria [Parihar M.S., Parihar A., Fujita M., Hashimoto M., Ghafourifar P. Mitochondrial association of alpha-synuclein causes oxidative stress. Cell Mol Life Sci. 2008a;65:1272-1284]. The present study shows that overexpression of alpha-synuclein A53T or A30P mutants or wild-type in human neuroblastoma cells augmented aggregation of alpha-synuclein. Immunoblotting and immuno-gold electron transmission microscopy show localization of alpha-synuclein aggregates within the mitochondria of overexpressing cells. Overexpressing cells show increased mitochondrial reactive oxygen species, increased protein tyrosine nitration, decreased mitochondrial transmembrane potential, and hampered cellular respiration. These findings suggest an important role for mitochondria in cellular responses to alpha-synuclein.


Subject(s)
Gene Expression , Mitochondria/metabolism , Neuroblastoma/metabolism , Oxidative Stress/physiology , alpha-Synuclein/physiology , Animals , Cell Line, Tumor , Fluorescent Antibody Technique , Humans , Immunoblotting , Membrane Potentials , Microscopy, Electron, Transmission , Mitochondria/pathology , Neuroblastoma/genetics , Rats , Reactive Oxygen Species/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
14.
Life Sci ; 82(21-22): 1077-82, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18456285

ABSTRACT

mAtNOS1 is a novel gene recently reported in mammalian genome with functions that are not fully understood. The present study shows that in human mammary adenocarcinoma MCF-7 cells, mAtNOS1 expression increases mitochondrial nitric oxide and calcium. Our study further shows that overexpression of mAtNOS1 induces apoptosis in MCF-7 cells by increasing mitochondrial protein tyrosine nitration and cytochrome c release. The present study suggests a novel function for mAtNOS1 in regulating mitochondrial nitric oxide and calcium and inducing apoptosis of MCF-7 cells.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Arabidopsis Proteins/biosynthesis , Breast Neoplasms/pathology , Nitric Oxide Synthase/biosynthesis , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Calcium/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Female , Humans , Immunochemistry , Mitochondria/metabolism , Nitric Oxide/metabolism , Transfection , Tyrosine/metabolism
15.
IUBMB Life ; 60(1): 64-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18379993

ABSTRACT

The present study shows that nitric oxide (NO) irreversibly inhibits purified cytochrome oxidase in a reverse oxygen concentration-dependent manner. The inhibition is dramatically protected by a peroxynitrite scavenger, suggesting that peroxynitrite is formed from the reaction of NO with cytochrome oxidase at low oxygen concentration, and that peroxynitrite is involved in irreversible cytochrome oxidase inactivation. Production of nitroxyl anion or superoxide was tested as potential mechanisms underlying the conversion of NO to peroxynitrite. A nitroxyl anion scavenger potently protected the irreversible inhibition, whereas a superoxide dismutase did not provide protective effect, suggesting that the peroxynitrite was formed from nitroxyl anion rather than the reaction of NO with superoxide.


Subject(s)
Electron Transport Complex IV/antagonists & inhibitors , Mitochondria, Liver/metabolism , Nitric Oxide/pharmacology , Oxygen/administration & dosage , Peroxynitrous Acid/biosynthesis , Animals , Mitochondria, Liver/drug effects , Rats
16.
Methods Enzymol ; 440: 317-34, 2008.
Article in English | MEDLINE | ID: mdl-18423228

ABSTRACT

Nitric oxide (NO) is a reactive radical synthesized by members of the NO synthase (NOS) family, including mitochondrial-specific NOS (mtNOS). Some of the assays used for the determination of cytoplasmic NOS activity have been utilized to detect mtNOS activity. However, it seems that many of those assays need to be adjusted and optimized to detect NO in the unique environment of mitochondria. Additionally, most mtNOS detection assays are designed and optimized for isolated mitochondria and may exert inherent pitfalls and limitations once used in living cells. This chapter describes several assays used commonly for mtNOS detection in isolated mitochondria and in mitochondria of live cells. Those include colorimetric and spectrophotometric methods, Griess reaction, radioassay, and polarographic and chemiluminescence assays. It also describes fluorescent-based assays for the detection of mitochondrial NO in live cells. Advantages and limitations of each assay are discussed.


Subject(s)
Mitochondria/enzymology , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Animals , Enzyme Activation/physiology , Humans
17.
Biochim Biophys Acta ; 1780(6): 921-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18359297
18.
Int J Mol Med ; 21(3): 317-24, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18288379

ABSTRACT

In the present study, we tested the significance of mitochondria for apoptosis upon exposure to tamoxifen and etoposide using two human breast cancer cell lines, MCF-7 and MDA-MB-231. We showed that both tamoxifen and etoposide induced apoptosis, increased intramitochondrial calcium and nitric oxide, and decreased mitochondrial transmembrane potential in both cell lines. Both drugs increased mitochondrial protein tyrosine nitration and caused release of cytochrome c from the mitochondria of both cell lines. This study suggests that tamoxifen and etoposide utilize a common mechanism to induce apoptosis in MCF-7 and MDA-MB-231 cells.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Calcium/metabolism , Etoposide/pharmacology , Mitochondria/metabolism , Nitric Oxide/metabolism , Tamoxifen/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Time Factors , Tyrosine/metabolism
19.
Biochem Biophys Res Commun ; 367(4): 761-7, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18191636

ABSTRACT

We recently demonstrated that mitochondrial nitric oxide synthase (mtNOS) functionally couples with mitochondrial respiratory chain complex I to produce nitric oxide [M.S. Parihar, R.R. Nazarewicz, E. Kincaid, U. Bringold, P. Ghafourifar, Association of mitochondrial nitric oxide synthase activity with respiratory chain complex I, Biochem. Biophys. Res. Commun. 366 (2008) 23-28]. The present report shows that inactivation of complex I leads mtNOS to become pro-oxidative. Our findings suggest a crucial role for mtNOS in oxidative stress caused by mitochondrial complex I inactivation.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria/metabolism , Nitric Oxide Synthase/metabolism , Oxidative Stress/physiology , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Animals , Cell Respiration/physiology , Cells, Cultured , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
20.
Mol Pharmacol ; 73(3): 995-1004, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18089839

ABSTRACT

Streptozotocin (STZ) is a diabetogenic agent extensively used to induce diabetes and to study complications including diabetic peripheral neuropathy (DPN). While studying the influence of transient receptor potential vanilloid 1 (TRPV1) on DPN in the STZ-induced diabetic mouse model, we found that a proportion of STZ-treated mice was nondiabetic but still exhibited hyperalgesia. To understand the mechanism underlying this phenomenon, dorsal root ganglion (DRG) neurons and stably TRPV1 expressing human embryonic kidney (HEK) 293T cells were used to study the expression and function of TRPV1. Incubation of DRG neurons with STZ resulted in a significant increase in the amplitude of capsaicin-induced TRPV1-mediated current and Ca(2+) influx compared with vehicle-treated sister cultures. It was also found that STZ treatment induced higher levels of reactive oxygen species, which was abolished with concomitant treatment with catalase. Treatment of cells with H(2)O(2) mimicked the effects of STZ. Western blot analysis revealed an increase in TRPV1 protein content and phospho p38 (p-p38) mitogen-activated protein kinase (MAPK) levels in DRG of STZ-injected diabetic and nondiabetic hyperalgesic mice compared with control mice. Furthermore, in stably TRPV1-expressing HEK 293T cells, STZ treatment induced an increase in TRPV1 protein content and p-p38 MAPK levels, which was abolished with concomitant treatment with catalase or p38 MAPK inhibitor. These results reveal that STZ has a direct action on neurons and modulates the expression and function of TRPV1, a nociceptive ion channel that is responsible for inflammatory thermal pain.


Subject(s)
Hot Temperature , Hyperalgesia/physiopathology , Neurons, Afferent/drug effects , Streptozocin/pharmacology , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Cells, Cultured , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Electrophysiology , Embryo, Mammalian , Female , Ganglia, Spinal/cytology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Kidney/cytology , Male , Mice , Neurons, Afferent/metabolism , Pain , Patch-Clamp Techniques , Pregnancy , Rats , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...