Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
2.
Front Cell Dev Biol ; 9: 777836, 2021.
Article in English | MEDLINE | ID: mdl-34957108

ABSTRACT

Programmed cell death (PCD) plays a critical role in the development and maturation of the cochlea. Significant remodeling occurs among cells of the greater epithelial ridge (GER) of Kölliker's organ, leading to tissue regression and formation of the inner sulcus. In mice, this event normally occurs between postnatal days 5-15 (P5-15) and is regulated by thyroid hormone (T3). During this developmental time period, the cochlea also contains a large population of macrophages. Macrophages are frequently involved in the phagocytic clearance of dead cells, both during development and after injury, but the role of macrophages in the developing cochlea is unknown. This study examined the link between developmental cell death in the GER and the recruitment of macrophages into this region. Cell death in the basal GER begins at P5 and enhanced numbers of macrophages were observed at P7. This pattern of macrophage recruitment was unchanged in mice that were genetically deficient for CX3CR1, the receptor for fractalkine (a known macrophage chemoattractant). We found that injection of T3 at P0 and P1 caused GER cell death to begin at P3, and this premature PCD was accompanied by earlier recruitment of macrophages. We further found that depletion of macrophages from the developing cochlea (using CX3CR1DTR/+ mice and treatment with the CSF1R antagonist BLZ945) had no effect on the pattern of GER regression. Together, these findings suggest that macrophages are recruited into the GER region after initiation of developmental PCD, but that they are not essential for GER regression during cochlear remodeling.

3.
Sci Rep ; 11(1): 2140, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33495483

ABSTRACT

The Hippo signaling pathway is a key regulator of tissue development and regeneration. Activation of the Hippo pathway leads to nuclear translocation of the YAP1 transcriptional coactivator, resulting in changes in gene expression and cell cycle entry. Recent studies have demonstrated the nuclear translocation of YAP1 during the development of the sensory organs of the inner ear, but the possible role of YAP1 in sensory regeneration of the inner ear is unclear. The present study characterized the cellular localization of YAP1 in the utricles of mice and chicks, both under normal conditions and after HC injury. During neonatal development, YAP1 expression was observed in the cytoplasm of supporting cells, and was transiently expressed in the cytoplasm of some differentiating hair cells. We also observed temporary nuclear translocation of YAP1 in supporting cells of the mouse utricle after short periods in organotypic culture. However, little or no nuclear translocation of YAP1 was observed in the utricles of neonatal or mature mice after ototoxic injury. In contrast, substantial YAP1 nuclear translocation was observed in the chicken utricle after streptomycin treatment in vitro and in vivo. Together, these data suggest that differences in YAP1 signaling may partially account for the differing regenerative abilities of the avian vs. mammalian inner ear.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Saccule and Utricle/embryology , Saccule and Utricle/injuries , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chickens , Diphtheria Toxin/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Protein Transport/drug effects , Saccule and Utricle/metabolism , Saccule and Utricle/pathology , Transcription Factor Brn-3C/metabolism
4.
Neuroscience ; 422: 146-160, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31678344

ABSTRACT

The vestibular system of the inner ear contains Type I and Type II hair cells (HCs) generated from sensory progenitor cells; however, little is known about how the HC subtypes are formed. Sox2 (encoding SRY-box 2) is expressed in Type II, but not in Type I, HCs. The present study aimed to investigate the role of SOX2 in cell fate determination in Type I vs. Type II HCs. First, we confirmed that Type I HCs developed from Sox2-expressing cells through lineage tracing of Sox2-positive cells using a CAG-tdTomato reporter mouse crossed with a Sox2-CreER mouse. Then, Sox2 loss of function was induced in HCs, using Sox2flox transgenic mice crossed with a Gfi1-Cre driver mouse. Knockout of Sox2 in HCs increased the number of Type I HCs and decreased the number of Type II HCs, while the total number of HCs and Sox2-positive supporting cells did not change. In addition, the effect of Sox2-knockout persisted into adulthood, resulting in an increased number of Type I HCs. These results demonstrate that SOX2 plays a critical role in the determination of Type II vs. Type I HC fate. The results suggested that Sox2 is a potential target for generating Type I HCs, which may be important for regenerative strategies for balance disorders.


Subject(s)
Aging/physiology , Cell Differentiation/physiology , Hair Cells, Vestibular/physiology , SOXB1 Transcription Factors/physiology , Animals , Cell Count , Cell Lineage/physiology , Mice , Mice, Knockout , Mice, Transgenic , SOXB1 Transcription Factors/genetics , Saccule and Utricle/cytology
5.
Sci Rep ; 9(1): 4131, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858408

ABSTRACT

Capsaicin, the spicy component of hot chili peppers activates the TRPV1 pain receptors, and causes rapid desensitization. Capsaicin also ameliorates cisplatin-induced nephrotoxicity. Cisplatin, a commonly used anti-neoplastic agent for solid tumors causes significant hearing loss, nephrotoxicity and peripheral neuropathy. Upregulation of cochlear TRPV1 expression is related to cisplatin-mediated ototoxicity. Here we report that direct TRPV1 activation by localized trans-tympanic (TT) or oral administration of capsaicin (TRPV1 agonist) prevents cisplatin ototoxicity by sustained increased activation of pro-survival transcription factor signal transducer and activator of transcription (STAT3) in the Wistar rat. Cisplatin treatment produced prolonged activation of pro-apoptotic Ser727 p-STAT1 and suppressed Tyr705-p-STAT3 for up to 72 h in the rat cochlea. Our data indicate that capsaicin causes a transient STAT1 activation via TRPV1 activation, responsible for the previously reported temporary threshold shift. Additionally, we found that capsaicin increased cannabinoid receptor (CB2) in the cochlea, which leads to pro-survival Tyr705-p-STAT3 activation. This tilts the delicate balance of p-STAT3/p-STAT1 towards survival. Furthermore, capsaicin mediated protection is lost when CB2 antagonist AM630 is administered prior to capsaicin treatment. In conclusion, capsaicin otoprotection appears to be mediated by activation of CB2 receptors in the cochlea which are coupled to both STAT1 and STAT3 activation.


Subject(s)
Antineoplastic Agents/toxicity , Capsaicin/pharmacology , Cisplatin/toxicity , Cochlea/metabolism , Ototoxicity/prevention & control , Receptor, Cannabinoid, CB2/metabolism , Sensory System Agents/pharmacology , Animals , Cannabinoid Receptor Antagonists/pharmacology , Capsaicin/therapeutic use , Cell Line , Cochlea/drug effects , Indoles/pharmacology , Male , Mice , Mice, SCID , Ototoxicity/drug therapy , Rats , Rats, Wistar , Receptor, Cannabinoid, CB2/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Sensory System Agents/therapeutic use , TRPV Cation Channels/metabolism
6.
Front Cell Neurosci ; 12: 271, 2018.
Article in English | MEDLINE | ID: mdl-30186120

ABSTRACT

Previous studies have demonstrated the presence of cannabinoid 2 receptor (CB2R) in the rat cochlea which was induced by cisplatin. In an organ of Corti-derived cell culture model, it was also shown that an agonist of the CB2R protected these cells against cisplatin-induced apoptosis. In the current study, we determined the distribution of CB2R in the mouse and rat cochleae and examined whether these receptors provide protection against cisplatin-induced hearing loss. In a knock-in mouse model expressing the CB2R tagged with green fluorescent protein, we show distribution of CB2R in the organ of Corti, stria vascularis, spiral ligament and spiral ganglion cells. A similar distribution of CB2R was observed in the rat cochlea using a polyclonal antibody against CB2R. Trans-tympanic administration of (2-methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone (JWH015), a selective agonist of the CB2R, protected against cisplatin-induced hearing loss which was reversed by blockade of this receptor with 6-iodo-2-methyl-1-[2-(4-morpholinyl)ethyl]-1H-indol-3-yl](4-methoxyphenyl)methanone (AM630), an antagonist of CB2R. JWH015 also reduced the loss of outer hair cells (OHCs) in the organ of Corti, loss of inner hair cell (IHC) ribbon synapses and loss of Na+/K+-ATPase immunoreactivity in the stria vascularis. Administration of AM630 alone produced significant hearing loss (measured by auditory brainstem responses) which was not associated with loss of OHCs, but led to reductions in the levels of IHC ribbon synapses and strial Na+/K+-ATPase immunoreactivity. Furthermore, knock-down of CB2R by trans-tympanic administration of siRNA sensitized the cochlea to cisplatin-induced hearing loss at the low and middle frequencies. Hearing loss induced by cisplatin and AM630 in the rat was associated with increased expression of genes for oxidative stress and inflammatory proteins in the rat cochlea. In vitro studies indicate that JWH015 did not alter cisplatin-induced killing of cancer cells suggesting this agent could be safely used during cisplatin chemotherapy. These data unmask a protective role of the cochlear endocannabinoid/CB2R system which appears tonically active under normal conditions to preserve normal hearing. However, an exogenous agonist is needed to boost the activity of endocannabinoid/CB2R system for protection against a more traumatic cochlear insult, as observed with cisplatin administration.

7.
Cell Death Dis ; 8(7): e2921, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28703809

ABSTRACT

Cisplatin-induced ototoxicity is one of the major factors limiting cisplatin chemotherapy. Ototoxicity results from damage to outer hair cells (OHCs) and other regions of the cochlea. At the cellular level, cisplatin increases reactive oxygen species (ROS) leading to cochlear inflammation and apoptosis. Thus, ideal otoprotective drugs should target oxidative stress and inflammatory mechanisms without interfering with cisplatin's chemotherapeutic efficacy. In this study, we show that epigallocatechin-3-gallate (EGCG) is a prototypic agent exhibiting these properties of an effect otoprotective agent. Rats administered oral EGCG demonstrate reduced cisplatin-induced hearing loss, reduced loss of OHCs in the basal region of the cochlea and reduced oxidative stress and apoptotic markers. EGCG also protected against the loss of ribbon synapses associated with inner hair cells and Na+/K+ ATPase α1 in the stria vascularis and spiral ligament. In vitro studies showed that EGCG reduced cisplatin-induced ROS generation and ERK1/2 and signal transducer and activator of transcription-1 (STAT1) activity, but preserved the activity of STAT3 and Bcl-xL. The increase in STAT3/STAT1 ratio appears critical for mediating its otoprotection. EGCG did not alter cisplatin-induced apoptosis of human-derived cancer cells or cisplatin antitumor efficacy in a xenograft tumor model in mice because of its inability to rescue the downregulation of STAT3 in these cells. These data suggest that EGCG is an ideal otoprotective agent for treating cisplatin-induced hearing loss without compromising its antitumor efficacy.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Catechin/analogs & derivatives , Cisplatin/toxicity , Cochlea/drug effects , Animals , Catechin/pharmacology , Cell Line , Cochlea/metabolism , Cochlea/pathology , HCT116 Cells , Hearing Loss/etiology , Hearing Loss/metabolism , Hearing Loss/pathology , Humans , Male , Mice , Phosphorylation/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Synapses/pathology
8.
PLoS One ; 12(5): e0177198, 2017.
Article in English | MEDLINE | ID: mdl-28467474

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer deaths in men. A better understanding of the molecular basis of prostate cancer proliferation and metastasis should enable development of more effective treatments. In this study we focused on the lncRNA, prostate cancer associated transcript 29 (PCAT29), a putative tumor suppressive gene. Our data show that the expression of PCAT29 was reduced in prostate cancer tumors compared to paired perinormal prostate tissues. We also observed substantially lower levels of PCAT29 in DU145 and LNCaP cells compared to normal prostate (RWPE-1) cells. IL-6, a cytokine which is elevated in prostate tumors, reduced the expression of PCAT29 in both DU145 and LNCaP cells by activating signal transducer and activator of transcription 3 (STAT3). One downstream target of STAT3 is microRNA (miR)-21, inhibition of which enhanced basal PCAT29 expression. In addition, we show that resveratrol is a potent stimulator of PCAT29 expression under basal condition and reversed the down regulation of this lncRNA by IL-6. Furthermore, we show that knock down of PCAT29 expression by siRNA in DU145 and LNCaP cells increased cell viability while increasing PCAT29 expression with resveratrol decreased cell viability. Immunohistochemistry studies showed increased levels of STAT3 and IL-6, but low levels of programmed cell death protein 4 (PDCD4), in prostate tumor epithelial cells compared to adjacent perinormal prostate epithelial cells. These data show that the IL-6/STAT3/miR-21 pathway mediates tonic suppression of PCAT29 expression and function. Inhibition of this signaling pathway by resveratrol induces PCAT29 expression and tumor suppressor function.


Subject(s)
Genes, Tumor Suppressor/physiology , Interleukin-6/physiology , Prostatic Neoplasms/metabolism , RNA, Long Noncoding/physiology , Signal Transduction/drug effects , Stilbenes/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , MicroRNAs/physiology , Real-Time Polymerase Chain Reaction , Resveratrol , STAT3 Transcription Factor/physiology , Signal Transduction/physiology
9.
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
10.
Expert Opin Investig Drugs ; 24(2): 201-17, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25243609

ABSTRACT

INTRODUCTION: Sensorineural hearing loss (HL) is becoming a global phenomenon at an alarming rate. Nearly 600 million people have been estimated to have significant HL in at least one ear. There are several different causes of sensorineural HL included in this review of new investigational drugs for HL. They are noise-induced, drug-induced, sudden sensorineural HL, presbycusis and HL due to cytomegalovirus infections. AREAS COVERED: This review presents trends in research for new investigational drugs encompassing a variety of causes of HL. The studies presented here are the latest developments either in the research laboratories or in preclinical, Phase 0, Phase I or Phase II clinical trials for drugs targeting HL. EXPERT OPINION: While it is important that prophylactic measures are developed, it is extremely crucial that rescue strategies for unexpected or unavoidable cochlear insult be established. To achieve this goal for the development of drugs for HL, innovative strategies and extensive testing are required for progress from the bench to bedside. However, although a great deal of research needs to be done to achieve the ultimate goal of protecting the ear against acquired sensorineural HL, we are likely to see exciting breakthroughs in the near future.


Subject(s)
Drug Design , Drugs, Investigational/therapeutic use , Hearing Loss, Sensorineural/drug therapy , Animals , Clinical Trials as Topic , Cytomegalovirus Infections/complications , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/physiopathology , Humans
11.
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
12.
Genet Test Mol Biomarkers ; 15(4): 215-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21198350

ABSTRACT

AIM: The present study was undertaken to investigate a length polymorphism of (GT)n repeats of the heme oxygenase-1 (HMOX-1) gene and its association with serum bilirubin levels in apparently healthy adults. METHODS: A total of 211 individuals (normal hematology and liver function test) with bilirubin levels of 1.7 to 22.2 µM were studied. The (GT)n repeats were analyzed by PCR and subsequent sizing by capillary electrophoresis on the ABI Prism 310 Genetic Analyzer. RESULTS: Polymorphisms of the (GT)n repeats were grouped into three classes: short (S) alleles (<20 repeats), intermediate (M) alleles (20-28 repeats), and long (L) alleles (≥ 29 repeats). The frequencies of the S, M, and L allele groups were 0.10, 0.49, and 0.41, respectively. Carriers of short alleles had significantly higher mean bilirubin levels (13.8 ± 5.10 µM) compared with others (9.18 ± 3.73 µM, p < 0.001). CONCLUSION: Short (GT)n alleles of the HMOX-1 gene promoter could be a genetic risk factor for hyperbilirubinemia.


Subject(s)
Bilirubin/blood , Dinucleotide Repeats/genetics , Heme Oxygenase-1/genetics , Hyperbilirubinemia/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Adult , Alleles , Electrophoresis, Capillary , Gene Frequency , Humans , India , Middle Aged , Polymerase Chain Reaction/methods , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL