Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neurosci ; 36(14): 3962-77, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27053204

ABSTRACT

Cisplatin is a commonly used antineoplastic agent that produces ototoxicity that is mediated in part by increasing levels of reactive oxygen species (ROS) via the NOX3 NADPH oxidase pathway in the cochlea. Recent studies implicate ROS generation in mediating inflammatory and apoptotic processes and hearing loss by activating signal transducer and activator of transcription (STAT1). In this study, we show that the adenosine A1 receptor (A1AR) protects against cisplatin ototoxicity by suppressing an inflammatory response initiated by ROS generation via NOX3 NADPH oxidase, leading to inhibition of STAT1. Trans-tympanic administration of the A1AR agonist R-phenylisopropyladenosine (R-PIA) inhibited cisplatin-induced ototoxicity, as measured by auditory brainstem responses and scanning electron microscopy in male Wistar rats. This was associated with reduced NOX3 expression, STAT1 activation, tumor necrosis factor-α (TNF-α) levels, and apoptosis in the cochlea. In vitro studies in UB/OC-1 cells, an organ of Corti immortalized cell line, showed that R-PIA reduced cisplatin-induced phosphorylation of STAT1 Ser(727) (but not Tyr(701)) and STAT1 luciferase activity by suppressing the ERK1/2, p38, and JNK mitogen-activated protein kinase (MAPK) pathways.R-PIA also decreased the expression of STAT1 target genes, such as TNF-α, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) and reduced cisplatin-mediated apoptosis. These data suggest that the A1AR provides otoprotection by suppressing NOX3 and inflammation in the cochlea and could serve as an ideal target for otoprotective drug therapy. SIGNIFICANCE STATEMENT: Cisplatin is a widely used chemotherapeutic agent for the treatment of solid tumors. Its use results in significant and permanent hearing loss, for which no US Food and Drug Administration-approved treatment is currently available. In this study, we targeted the cochlear adenosine A1 receptor (A1AR) by trans-tympanic injections of the agonist R-phenylisopropyladenosine (R-PIA) and showed that it reduced cisplatin-induced inflammation and apoptosis in the rat cochlea and preserved hearing. The mechanism of protection involves suppression of the NOX3 NADPH oxidase enzyme, a major target of cisplatin-induced reactive oxygen species (ROS) generation in the cochlea. ROS initiates an inflammatory and apoptotic cascade in the cochlea by activating STAT1 transcription factor, which is attenuated byR-PIA. Therefore, trans-tympanic delivery of A1AR agonists could effectively treat cisplatin ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cochlea/drug effects , Inflammation/physiopathology , NADPH Oxidases/drug effects , NADPH Oxidases/genetics , Receptor, Adenosine A1/drug effects , STAT1 Transcription Factor/drug effects , STAT1 Transcription Factor/genetics , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/administration & dosage , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Cell Line , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory/drug effects , Hearing Disorders/chemically induced , Hearing Disorders/physiopathology , MAP Kinase Signaling System/drug effects , Male , Rats , Rats, Wistar , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
2.
Antioxid Redox Signal ; 19(16): 1863-76, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23682737

ABSTRACT

AIMS: Oncogenic microRNAs (miRs) promote tumor growth and invasiveness. One of these, miR-21, contributes to carcinogenesis in prostate and other cancers. In the present study, we tested the hypothesis that NADPH oxidase-dependent reactive oxygen species (ROS) regulate the expression and function of miR-21 and its target proteins, maspin and programmed cell death 4 (PDCD4), in prostate cancer cells. RESULTS: The highly aggressive androgen receptor negative PC-3M-MM2 prostate cancer cells demonstrated high expression of miR-21 and p47(phox) (an essential subunit of NADPH oxidase). Using loss-of-function strategy, we showed that transfection of PC-3M-MM2 cells with anti-miR-21- and p47(phox) siRNA (si-p47(phox)) led to reduced expression of miR-21 with concurrent increase in maspin and PDCD4, and decreased the invasiveness of the cells. Tail-vein injections of anti-miR-21- and si-p47(phox)-transfected PC-3M-MM2 cells in severe combined immunodeficient mice reduced lung metastases. Clinical samples from patients with advanced prostate cancer expressed high levels of miR-21 and p47(phox), and low expression of maspin and PDCD4. Finally, ROS activated Akt in these cells, the inhibition of which reduced miR-21 expression. INNOVATION: The levels of NADPH oxidase-derived ROS are high in prostate cancer cells, which have been shown to be involved in their growth and migration. This study demonstrates that ROS produced by this pathway is essential for the expression and function of an onco-miR, miR-21, in androgen receptor-negative prostate cancer cells. CONCLUSION: These data demonstrate that miR-21 is an important target of ROS, which contributes to the highly invasive and metastatic phenotype of prostate cancer cells.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , NADPH Oxidases/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Humans , Male , Mice , Mice, SCID , NADPH Oxidases/antagonists & inhibitors , Neoplasm Invasiveness/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/metabolism , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Serpins/biosynthesis , Serpins/genetics , Serpins/metabolism , Tumor Cells, Cultured
3.
PLoS One ; 7(12): e51655, 2012.
Article in English | MEDLINE | ID: mdl-23272133

ABSTRACT

The consumption of foods containing resveratrol produces significant health benefits. Resveratrol inhibits cancer by reducing cell proliferation and metastasis and by inducing apoptosis. These actions could be explained by its ability to inhibit (ERK-1/2), Akt and suppressing the levels of estrogen and insulin growth factor -1 (IGF-1) receptor. How these processes are manifested into the antitumor actions of resveratrol is not clear. Using microarray studies, we show that resveratrol reduced the expression of various prostate-tumor associated microRNAs (miRs) including miR-21 in androgen-receptor negative and highly aggressive human prostate cancer cells, PC-3M-MM2. This effect of resveratrol was associated with reduced cell viability, migration and invasiveness. Additionally, resveratrol increased the expression of tumor suppressors, PDCD4 and maspin, which are negatively regulated by miR-21. Short interfering (si) RNA against PDCD4 attenuated resveratrol's effect on prostate cancer cells, and similar effects were observed following over expression of miR-21 with pre-miR-21 oligonucleotides. PC-3M-MM2 cells also exhibited high levels of phospho-Akt (pAkt), which were reduced by both resveratrol and LY294002 (a PI3-kinase inhibitor). MiR-21 expression in these cells appeared to be dependent on Akt, as LY294002 reduced the levels of miR-21 along with a concurrent increase in PDCD4 expression. These in vitro findings were further corroborated in a severe combined immunodeficient (SCID) mouse xenograft model of prostate cancer. Oral administration of resveratrol not only inhibited the tumor growth but also decreased the incidence and number of metastatic lung lesions. These tumor- and metastatic-suppressive effects of resveratrol were associated with reduced miR-21 and pAkt, and elevated PDCD4 levels. Similar anti-tumor effects of resveratrol were observed in DU145 and LNCaP prostate cancer cells which were associated with suppression of Akt and PDCD4, but independent of miR-21.These data suggest that resveratrol's anti-tumor actions in prostate cancer could be explained, in part, through inhibition of Akt/miR-21 signaling pathway.


Subject(s)
MicroRNAs/metabolism , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Stilbenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Estrogens/metabolism , Flow Cytometry/methods , Genes, Tumor Suppressor , Humans , Male , Mice , Mice, SCID , MicroRNAs/antagonists & inhibitors , Morpholines/pharmacology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Oligonucleotides/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA, Small Interfering/metabolism , Receptor, IGF Type 1/metabolism , Resveratrol , Wound Healing
4.
Cell Mol Life Sci ; 69(14): 2429-34, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22562580

ABSTRACT

Cisplatin is a widely used chemotherapeutic agent that causes significant hearing loss. Previous studies have shown that cisplatin exposure is associated with increase in reactive oxygen species (ROS) in the cochlea. The inner ear expresses a unique isoform of NADPH oxidase, NOX3. This enzyme may be the primary source of ROS generation in the cochlea. The knockdown of NOX3 by pretreatment with siRNA prevented cisplatin ototoxicity, as demonstrated by preservation of hearing thresholds and inner ear sensory cells. Trans-tympanic NOX3 siRNA reduced the expression of NOX3 and biomarkers of cochlear damage, including transient receptor vanilloid 1 (TRPV1) channel and kidney injury molecule-1 (KIM-1) in cochlear tissues. In addition, siRNA against NOX3 reduced apoptosis as demonstrated by TUNEL staining, and prevented the increased expression of Bax and abrogated the decrease in Bcl2 expression following cisplatin administration. Trans-tympanic administration of siRNA directed against NOX3 may provide a useful method of attenuating cisplatin ototoxicity. In this paper, we review recent publications dealing with the role of NOX3 in ototoxicity and the effects of siRNA against cisplatin-induced hearing loss.


Subject(s)
Hearing Loss/enzymology , Membrane Proteins/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , RNA, Small Interfering/metabolism , Cisplatin/toxicity , Ear, Inner/enzymology , Hearing Loss/chemically induced , Hearing Loss/pathology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Paraquat/toxicity , RNA Interference
5.
Curr Neuropharmacol ; 9(2): 342-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22131942

ABSTRACT

Adenosine is produced primarily by the metabolism of ATP and mediates its physiological actions by interacting primarily with adenosine receptors (ARs) on the plasma membranes of different cell types in the body. Activation of these G protein-coupled receptors promotes activation of diverse cellular signaling pathways that define their tissue-specific functions. One of the major actions of adenosine is cytoprotection, mediated primarily via two ARs - A(1) (A(1)AR) and A(3) (A(3)AR). These ARs protect cells exposed to oxidative stress and are also regulated by oxidative stress. Stress-mediated regulation of ARs involves two prominent transcription factors - activator protein-1 (AP-1) and nuclear factor (NF)-κB - that mediate the induction of genes important in cell survival. Mice that are genetically deficient in the p50 subunit of NF-κB (i.e., p50 knock-out mice) exhibit altered expression of A(1)AR and A(2A)AR and demonstrate distinct behavioral phenotypes under normal conditions or after drug challenges. These effects suggest an important role for NF-κB in dictating the level of expression of ARs in vivo, in regulating the cellular responses to stress, and in modifying behavior.

6.
Antioxid Redox Signal ; 14(6): 999-1010, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20712533

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is implicated in cisplatin ototoxicity. Activation of this channel by cisplatin increases reactive oxygen species generation, which contribute to loss of outer hair cells in the cochlea. Knockdown of TRPV1 by short interfering RNA protected against cisplatin ototoxicity. In this study, we examined the mechanism underlying TRPV1-mediated ototoxicity using cultured organ of Corti transformed cells (UB/OC-1) and rats. Trans-tympanic injections of capsaicin produced transient hearing loss within 24 h, which recovered by 72 h. In UB/OC-1 cells, capsaicin increased NOX3 NADPH oxidase activity and activation of signal transducer and activator of transcription 1 (STAT1). Intratympanic administration of capsaicin transiently increased STAT1 activity and expression of downstream proinflammatory molecules. Capsaicin produced a transient increase in CD14-positive inflammatory cells into the cochlea, which mimicked the temporal course of STAT1 activation but did not alter the expression of apoptotic genes or damage to outer hair cells. In addition, trans-tympanic administration of STAT1 short interfering RNA protected against capsaicin-induced hearing loss. These data suggest that activation of TRPV1 mediates temporary hearing loss by initiating an inflammatory process in the cochlea via activation of NOX3 and STAT1. Thus, these proteins represent reasonable targets for ameliorating hearing loss.


Subject(s)
Capsaicin/pharmacology , Hearing Loss/metabolism , Inflammation/metabolism , NADPH Oxidases/metabolism , STAT1 Transcription Factor/metabolism , TRPV Cation Channels/metabolism , Animals , Capsaicin/administration & dosage , Cells, Cultured , Cochlea/drug effects , Cochlea/metabolism , Evoked Potentials , Hearing Loss/chemically induced , Immunohistochemistry , Inflammation/genetics , Male , Mice , Microscopy, Electron, Scanning , NADPH Oxidases/genetics , RNA, Small Interfering/drug effects , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , TRPV Cation Channels/genetics
7.
Expert Opin Drug Discov ; 6(5): 491-505, 2011 May.
Article in English | MEDLINE | ID: mdl-22646075

ABSTRACT

INTRODUCTION: Sensorineural hearing loss affects a high percentage of the population. Ototoxicity is a serious and pervasive problem in patients treated with cisplatin. Strategies to ameliorate ototoxicity without compromising on antitumor activity of treatments are urgently needed. Similar problems occur with aminoglycoside antibiotic therapy for infections. Noise-induced hearing loss affects a large number of people. The use of ear protection is not always possible or effective. The prevention of hearing loss with drug therapy would have a huge impact in reducing the number of people with hearing loss from these major causes. AREAS COVERED: This review discusses significant research findings dealing with the use of protective agents against hearing loss caused by cisplatin, aminoglycoside antibiotics and noise trauma. The efficacy in animal studies and the application of these protective agents in clinical trials that are ongoing are presented. EXPERT OPINION: The reader will gain new insights into current and projected future strategies to prevent sensorineural hearing loss from cisplatin chemotherapy, aminoglycoside antibiotic therapy and noise exposure. The future appears to offer numerous agents to prevent hearing loss caused by cisplatin, aminoglycoside antibiotics and noise. Novel delivery systems will provide ways to guide these protective agents to the desired target areas in the inner ear and circumvent problems with therapeutic interference of antitumor and antibiotics agents as well as minimize undesired side effects.

8.
Antioxid Redox Signal ; 13(5): 589-98, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20214492

ABSTRACT

Cisplatin produces hearing loss in cancer patients. Reactive oxygen species (ROS) in the cochlea leads to lipid peroxidation, death of outer hair cells (OHCs), and hearing loss. The cochlea expresses a unique isoform of NADPH oxidase, NOX3, which serves as the primary source of ROS generation in the cochlea. Inhibition of NOX3 could offer a unique protective target against cisplatin ototoxicity. Here, we document that knockdown of NOX3 using short interfering (si) RNA abrogated cisplatin ototoxicity, as evidenced by protection of OHCs from damage and reduced threshold shifts in auditory brainstem responses (ABRs). Transtympanic NOX3 siRNA reduced the expression of NOX3 in OHCs, spiral ganglion (SG) cells, and stria vascularis (SV) in the rat. NOX3 siRNA also reduced the expression of transient receptor potential vanilloid 1 (TRPV1) channel and kidney injury molecule-1 (KIM-1), biomarkers of cochlear damage. Also, transtympanic NOX3 siRNA reduced the expression of Bax, abolished the decrease in expression of Bcl2, and reduced apoptosis induced by cisplatin in the cochlea. These data suggest that NOX3 regulates stress-related genes in the cochlea, such as TRPV1 and KIM-1, and initiates apoptosis in the cochlea. This appears to be the first study of the efficacy of transtympanic delivery of siRNA attenuating cisplatin ototoxicity.


Subject(s)
Cisplatin/adverse effects , Hearing Loss/chemically induced , Hearing Loss/prevention & control , NADPH Oxidases/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Animals , Apoptosis/drug effects , Auditory Threshold/drug effects , Auditory Threshold/physiology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , Cochlea/ultrastructure , Gene Expression/drug effects , Gene Expression/genetics , Hair Cells, Auditory, Outer/cytology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/metabolism , Hearing Loss/pathology , Hearing Loss/physiopathology , Male , NADPH Oxidases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , bcl-2-Associated X Protein/genetics
9.
Am J Physiol Cell Physiol ; 298(1): C56-65, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19828838

ABSTRACT

Exposure of cells to adenosine receptor (AR) agonists leads to receptor uncoupling from G proteins and downregulation of the A(1)AR. The receptor levels on the cell surface generally recover on withdrawal of the agonist, because of either translocation of the sequestered A(1)AR back to plasma membrane or de novo synthesis of A(1)AR. To examine the mechanism(s) underlying A(1)AR downregulation and recovery, we treated ductus deferens tumor (DDT(1) MF-2) cells with the agonist R-phenylisopropyladenosine (R-PIA) and showed a decrease in membrane A(1)AR levels by 24 h, which was associated with an unexpected 11-fold increase in A(1)AR mRNA. Acute exposure of these cells to R-PIA resulted in a rapid translocation of beta-arrestin1 to the plasma membrane. Knockdown of beta-arrestin1 by short interfering RNA (siRNA) blocked R-PIA-mediated downregulation of the A(1)AR, suppressed R-PIA-dependent ERK1/2 and activator protein-1 (AP-1) activity, and reduced the induction of A(1)AR mRNA. Withdrawal of the agonist after a 24-h exposure resulted in rapid recovery of plasma membrane A(1)AR. This was dependent on the de novo protein synthesis and on the activity of ERK1/2 but independent of beta-arrestin1 and nuclear factor-kappaB. Together, these data suggest that exposure to A(1)AR agonist stimulates ERK1/2 activity via beta-arrestin1, which subserves receptor uncoupling and downregulation, in addition to the induction of A(1)AR expression. We propose that such a pathway ensures both the termination of the agonist signal and recovery by priming the cell for rapid de novo synthesis of A(1)AR once the drug is terminated.


Subject(s)
Arrestins/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptor, Adenosine A1/physiology , Animals , Arrestins/genetics , Blotting, Western , Cell Culture Techniques , Cell Division , Cricetinae , Immunohistochemistry , Male , Mesocricetus , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Phenylisopropyladenosine/pharmacology , RNA, Small Interfering/genetics , Receptor, Adenosine A1/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vas Deferens/cytology , Vas Deferens/physiology , beta-Arrestins
10.
Neoplasia ; 11(11): 1132-45, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19881949

ABSTRACT

Prostate cancer is the most commonly diagnosed and second most lethal malignancy in men, due mainly to a lack of effective treatment for the metastatic disease. A number of recent studies have shown that activation of the purine nucleoside receptor, adenosine A(3) receptor (A(3)AR), attenuates proliferation of melanoma, colon, and prostate cancer cells. In the present study, we determined whether activation of the A(3)AR reduces the ability of prostate cancer cells to migrate in vitro and metastasize in vivo. Using severe combined immunodeficient mice, we show that proliferation and metastasis of AT6.1 rat prostate cancer cells were decreased by the administration of A(3)AR agonist N(6)-(3-iodobenzyl) adenosine-5'-N-methyluronamide. In vitro studies show that activation of A(3)AR decreased high basal nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity present in these cells, along with the expression of Rac1 and p47(phox) subunits of this enzyme. Inhibition of NADPH oxidase activity by the dominant-negative RacN17 or short interfering (si)RNA against p47(phox) reduced both the generation of reactive oxygen species and the invasion of these cells on Matrigel. In addition, we show that membrane association of p47(phox) and activation of NADPH oxidase is dependent on the activity of the extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein kinase pathway. We also provide evidence that A(3)AR inhibits ERK1/2 activity in prostate cancer cells through inhibition of adenylyl cyclase and protein kinase A. We conclude that activation of the A(3)AR in prostate cancer cells reduces protein kinase A-mediated stimulation of ERK1/2, leading to reduced NADPH oxidase activity and cancer cell invasiveness.


Subject(s)
NADPH Oxidases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, Adenosine A3/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , RNA, Small Interfering , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
11.
Tohoku J Exp Med ; 219(3): 177-86, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19851045

ABSTRACT

Cisplatin is a chemotherapeutic agent that is widely used to treat a variety of malignant tumors. Serious dose-limiting side effects like ototoxicity, nephrotoxicity and neurotoxicity occur with the use of this agent. This review summarizes recent important clinical and experimental investigations of cisplatin ototoxicity. It also discusses the utility of protective agents employed in patients and in experimental animals. The future strategies for limiting cisplatin ototoxicity will need to avoid interference with the therapeutic effect of cisplatin in order to enhance the quality of life of patients receiving this important anti-tumor agent.


Subject(s)
Cisplatin/adverse effects , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Animals , Cisplatin/therapeutic use , Clinical Trials as Topic , Genetic Predisposition to Disease , Hearing Loss/genetics , Humans
12.
J Neurosci ; 28(49): 13056-65, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-19052196

ABSTRACT

Cisplatin, a chemotherapeutic agent of choice for the treatment of solid tumors, produces hearing loss in approximately half a million new cancer patients annually in the United States. The hearing loss is due, in part, to increased generation of reactive oxygen species (ROS) in the cochlea, leading to lipid peroxidation and damage or death of outer hair cells in the organ of Corti. The cochlea expresses the transient receptor potential vanilloid 1 (TRPV1), which are normally expressed on small diameter neurons in the peripheral nervous system and mediate thermal sensitivity, but whose role in the cochlea is unclear. In this study, we show that TRPV1 is coregulated along with the NADPH oxidase isoform, NOX3, by cisplatin. Induction of these proteins by cisplatin is dependent on ROS generation, since it is reversed by systemic lipoic acid administration. In organ of Corti hair cell cultures (UB/OC-1 cells), cisplatin activates and induces TRPV1 and NOX3, leading to apoptosis of these cells. Inhibition of TRPV1 by capsazepine or ruthenium red reduced the apoptosis, implicating TRPV1 in this process. Treatment of UB/OC-1 cultures with short interfering RNA (siRNA) against either TRPV1 or NOX3 reduced cisplatin-induced apoptosis, while round window application of TRPV1 siRNA to rats reduced TRPV1 expression, decreased damage to outer hair cells and reduced cisplatin-induced hearing loss. These data provide a link between NOX3 and TRPV1 in cisplatin-induced hearing loss and suggest that targeting these proteins for knockdown by siRNA could serve as a novel approach in treating cisplatin ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Hair Cells, Auditory/metabolism , Hearing Loss, Sensorineural/chemically induced , Oxidative Stress , RNA Interference , TRPV Cation Channels/antagonists & inhibitors , Animals , Antioxidants/pharmacology , Apoptosis/genetics , Cells, Cultured , Disease Models, Animal , Down-Regulation/genetics , Hair Cells, Auditory/drug effects , Hearing Loss, Sensorineural/therapy , Isoenzymes/genetics , Male , NADPH Oxidases/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , TRPV Cation Channels/genetics , Thioctic Acid/pharmacology
13.
Mol Pharmacol ; 72(4): 856-67, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17609415

ABSTRACT

Human immunodeficiency virus dementia (HIV-D) is a nonfocal central nervous system manifestation characterized by cognitive, behavioral, and motor abnormalities. The pathophysiology of neuronal damage in HIV-D includes a direct toxic effect of viral proteins on neuronal cells and an indirect effect caused by the release of inflammatory mediators and neurotoxins by activated macrophages/microglia and astrocytes, culminating into neuronal apoptosis. Previous studies have documented that the nucleoside adenosine mediates neuroprotection by activating adenosine A(1) receptor subtype (A(1)AR) linked to suppression of neuronal excitability. In this study, we show that A(1)AR activation protects against HIV-1 Tat-induced toxicity in primary cultures of rat cerebellar granule neurons and in rat pheochromocytoma (PC12) cell. In PC12 cells, HIV-1 Tat increased [Ca(2+)](i) levels, release of nitric oxide (NO), and expression of inducible nitric-oxide synthase (iNOS) and A(1)AR. Activation of A(1)AR suppressed Tat-mediated increases in [Ca(2+)](i) and NO. Furthermore, A(1)AR agonists inhibited iNOS expression in a nuclear factor-kappaB (NF-kappaB)-dependent manner. It is noteworthy that activation of the A(1)AR or inhibition of NOS protected against Tat-induced apoptosis in PC12 cells and cerebellar granule cells. Moreover, activation of the A(1)AR-inhibited Tat-induced increases in the levels of proapoptotic proteins Bax and caspase-3. Taken together, our results demonstrate that the A(1)AR protects against HIV-1 toxicity by inhibiting NF-kappaB, thereby reducing the expression of iNOS and NO radicals and neuronal apoptosis.


Subject(s)
Adenosine A1 Receptor Agonists , Apoptosis/physiology , Gene Products, tat/physiology , HIV-1/physiology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Immunohistochemistry , Neurons/cytology , PC12 Cells , Rats , Reverse Transcriptase Polymerase Chain Reaction
14.
J Pharmacol Exp Ther ; 317(1): 1-10, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16322354

ABSTRACT

Pertussis toxin ADP ribosylates G(i) and G(o) transducing proteins and functionally uncouples adenosine A(1) receptor (A(1)AR) from its effectors. We hypothesized that this loss in receptor coupling could lead to de novo A(1)AR synthesis by the cell in a futile attempt to re-establish normal receptor function. To test this hypothesis, we used hamster ductus deferens tumor (DDT(1) MF-2) cells, a cell culture model for studying A(1)AR, and showed that pertussis toxin (100 ng/ml) produced a time-dependent loss in A(1)AR-G(i) interaction and abolished A(1)AR activation of extracellular signal-regulated kinase 1/2. Interestingly, pertussis toxin increased the expression of A(1)AR, as measured by real-time polymerase chain reaction, immunocytochemistry, and [(3)H]cyclopentyl-1,3-dipropylxanthine (DPCPX) binding, suggesting a compensatory response to G(i) protein inactivation. DDT(1) MF-2 cells exposed to pertussis toxin demonstrated nuclear factor kappaB (NF-kappaB) activation within 30 min of exposure, a time point that preceded the loss of function of the A(1)AR. Inhibition of NF-kappaB attenuated the increase in A(1)AR induced by pertussis toxin. Cells exposed to B-oligomer subunit of pertussis toxin, devoid of significant ADP ribosyltransferase activity, showed increased A(1)AR protein expression, preceded by activation of NF-kappaB. B-Oligomer increased intracellular Ca(2+) in DDT(1) MF-2 cells. Chelation of intracellular Ca(2+) with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid or inhibition of protein kinase C (PKC) with bisindolylmaleimide hydrochloride reduced the activation of NF-kappaB and [(3)H]DPCPX binding. We conclude that pertussis toxin promotes de novo A(1)AR synthesis by activating NF-kappaB through an ADP ribosylation-independent mechanism involving intracellular Ca(2+) release and PKC activation.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , NF-kappa B/metabolism , Pertussis Toxin/pharmacology , Receptor, Adenosine A1/biosynthesis , Signal Transduction/drug effects , Animals , Calcium/metabolism , Cell Line, Tumor , Cricetinae , Dose-Response Relationship, Drug , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Protein Binding , Protein Kinase C/metabolism
15.
J Neurochem ; 95(6): 1689-703, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16283857

ABSTRACT

Nerve growth factor (NGF) regulates the nociceptive properties of a subset of small diameter sensory neurons by increasing the expression of the heat-sensing transient receptor potential (TRP) channel, TRPV1. This action involves activation of the tyrosine kinase receptor (Trk) A/p38 MAPK pathway. Recent studies indicate that activation of TrkA promotes superoxide generation via NADPH oxidase. In this study, we determined whether the NADPH oxidase pathway is involved in NGF-stimulated TRPV1 expression using a rat pheochromocytoma 12 line and rat dorsal root ganglion neurons. Treatment of these cells with NGF (100 ng/mL) increased TRPV1 protein expression (approx. twofold) but not mRNA. This increase was mimicked by H(2)O(2) and attenuated by catalase and inhibitors of NADPH oxidase. NGF stimulated NADPH oxidase activity, while 24 h exposure further increased expression of the Rac1 and gp91(phox) subunits of the holoenzyme. Inhibition of NADPH oxidase by transient transfection of a dominant negative Rac1 mutant (RacN17) plasmid blocked NGF-stimulated TRPV1 protein expression, while expression of a constitutively active Rac1 increased basal and NGF-stimulated TRPV1 levels. Inhibition of NADPH oxidase activity also attenuated NGF-dependent p38 MAPK activation. We conclude that the Rac1/NADPH oxidase pathway regulates p38 activation and TRPV1 expression which aids in the maintenance of peripheral neuron integrity and pain perception.


Subject(s)
NADPH Oxidases/physiology , Nerve Growth Factors/biosynthesis , TRPV Cation Channels/biosynthesis , rac1 GTP-Binding Protein/physiology , Animals , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/physiology , Fluoresceins , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Immunohistochemistry , Neurons/drug effects , Neurons/metabolism , PC12 Cells , RNA, Messenger/biosynthesis , Radioligand Assay , Rats , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...