Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Mol Sci ; 25(2)2024 Jan 14.
Article in English | MEDLINE | ID: mdl-38256106

ABSTRACT

Chemotherapy and radiotherapy resistance are major obstacles in the long-term efficacy of head and neck squamous cell carcinoma (HNSCC) treatment. Secondhand smoke (SHS) exposure is common and has been proposed as an independent predictor of HNSCC recurrence and disease-free survival. However, the underlying mechanisms responsible for these negative patient outcomes are unknown. To assess the effects of SHS exposure on cisplatin efficacy in cancer cells, three distinct HNSCC cell lines were exposed to sidestream (SS) smoke, the main component of SHS, at concentrations mimicking the nicotine level seen in passive smokers' saliva and treated with cisplatin (0.01-100 µM) for 48 h. Compared to cisplatin treatment alone, cancer cells exposed to both cisplatin and SS smoke extract showed significantly lower cisplatin-induced cell death and higher cell viability, IC50, and indefinite survival capacity. However, SS smoke extract exposure alone did not change cancer cell viability, cell death, or cell proliferation compared to unexposed control cancer cells. Mechanistically, exposure to SS smoke extract significantly reduced the expression of cisplatin influx transporter CTR1, and increased the expression of multidrug-resistant proteins ABCG2 and ATP7A. Our study is the first to document that exposure to SHS can increase cisplatin resistance by altering the expression of several proteins involved in multidrug resistance, thus increasing the cells' capability to evade cisplatin-induced cell death. These findings emphasize the urgent need for clinicians to consider the potential role of SHS on treatment outcomes and to advise cancer patients and caregivers on the potential benefits of avoiding SHS exposure.


Subject(s)
Head and Neck Neoplasms , Tobacco Smoke Pollution , Humans , Tobacco Smoke Pollution/adverse effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Head and Neck Neoplasms/drug therapy , Cell Death
2.
Cells ; 12(20)2023 10 17.
Article in English | MEDLINE | ID: mdl-37887311

ABSTRACT

Fusion genes are key cancer driver genes that can be used as potential drug targets in precision therapies, and they can also serve as accurate diagnostic and prognostic biomarkers. The fusion genes can cause microRNA (miRNA/miR) aberrations in many types of cancer. Nevertheless, whether fusion genes incite miRNA aberrations as one of their many critical oncogenic functionalities for driving carcinogenesis needs further investigation. Recent discoveries of miRNA genes that are present within the regions of genomic rearrangements that initiate fusion gene-based intronic miRNA dysregulation have brought the fusion genes into the limelight and revealed their unexplored potential in the field of cancer biology. Fusion gene-based 'promoter-switch' event aberrantly activate the miRNA-related upstream regulatory signals, while fusion-based coding region alterations disrupt the original miRNA coding loci. Fusion genes can potentially regulate the miRNA aberrations regardless of the protein-coding capability of the resultant fusion transcript. Studies on out-of-frame fusion and nonrecurrent fusion genes that cause miRNA dysregulation have attracted the attention of researchers on fusion genes from an oncological perspective and therefore could have potential implications in cancer therapies. This review will provide insights into the role of fusion genes and miRNAs, and their possible interrelationships in cancer.


Subject(s)
MicroRNAs , Neoplasms , Humans , MicroRNAs/genetics , Neoplasms/genetics , Introns , Oncogenes , Carcinogenesis/genetics
3.
Cytogenet Genome Res ; 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37906989

ABSTRACT

BACKGROUND: Development of resistance upon exposure to small doses of ionizing radiation followed by higher doses is known as radiation-induced adaptive response (RIAR). Traditionally, the induction of the RIAR phenomenon at the cellular level has been examined in cell lines, animal models, and epidemiological studies where people live in high natural background radiation. SUMMARY: The primary intention of the earlier studies was to corroborate the existence of RIAR and the mechanism involved in mediating the response surveyed by exposure to a low dose of radiation (<500 mGy) as priming dose towards the radiation protection point of view. However, the investigation has shifted the focus to understand the relevance of this phenomenon at clinically relevant set-ups (high doses in the order of Gy) and can be exploited during radiotherapy as RIAR is considered a mechanism for the development of radioresistance. Although the knowledge of molecular mechanisms at the cellular level has evolved significantly in multi-fractionated radiotherapy regimes, its relevance in developing radioresistance at low doses remains elusive. The authors recapitulate the existing knowledge on RIAR at cellular levels, specifically after low-dose exposure as an adaptive dose, and discussed its potential implications in clinical radiotherapy outcomes. KEY MESSAGES: Recent studies contributed to understand the signaling molecules, pathways, and inhibitors to mitigate RIAR-mediated radiation resistance and persistent radio-tolerance at the cellular level. Monitoring the disease progression in tumor samples or liquid biopsies before, during, and after therapy with suitable biomarkers has been proposed as a strategy to translate the phenomena into clinical scenario.

4.
Curr Top Med Chem ; 22(31): 2589-2597, 2022.
Article in English | MEDLINE | ID: mdl-36380414

ABSTRACT

Endocrine cancer is an uncontrolled growth of cells in the hormone-producing glands. Endocrine cancers include the adrenal, thyroid, parathyroid, pancreas, pituitary, and ovary malignancy. Recently, there is an increase in the incidence of the most common endocrine cancer types, namely pancreatic and thyroid cancers. Cancer stem cells (CSCs) of endocrine tumors have received more attention due to their role in cancer progression, therapeutic resistance, and cancer relapse. Phytochemicals provide several health benefits and are effective in the treatment of various diseases including cancer. Therefore, finding the natural phytochemicals that target the CSCs will help to improve cancer patients' prognosis and life expectancy. Phytochemicals have been shown to have anticancer properties and are very effective in treating various cancer types. Curcumin is a common polyphenol found in turmeric, which has been shown to promote cellular drug accumulation and increase the effectiveness of chemotherapy. Moreover, various other phytochemicals such as resveratrol, genistein, and apigenin are effective against different endocrine cancers by regulating the CSCs. Thus, phytochemicals have emerged as chemotherapeutics that may have significance in preventing and treating the endocrine cancers.


Subject(s)
Genistein , Ovarian Neoplasms , Female , Humans , Genistein/pharmacology , Ovarian Neoplasms/drug therapy , Resveratrol/pharmacology , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Neoplastic Stem Cells
5.
Colloids Surf B Biointerfaces ; 217: 112612, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35738074

ABSTRACT

The cancer stem cell (CSC) hypothesis is an evolving oncogenesis concept. CSCs have a distinct ability to self-renew themselves and also give rise to a phenotypically diverse population of cells. Targeting CSCs represents a promising strategy for cancer treatment. Plant-derived compounds are potent in restricting the expansion of CSCs. DCLK1 has been already reported as a colon CSC specific marker. Nanoparticles can effectively inhibit multiple types of CSCs by targeting specific markers. We have synthesized DCLK1 functionalized folic acid conjugated hesperetin encapsulated chitosan nanoparticles (CFH-DCLK1), specifically to target CSCs. In this regard, we have performed proliferation assay, colony formation assay, cell migration assay, apoptosis assay, flow cytometry analysis, real-time RT- PCR and western blot analyses to determine the effect of CFH-DCLK1 and CFH nanoparticles in HCT116-colon cancer cells. In our study, we have determined the median inhibitory concentration (IC50) of CFH (47.8 µM) and CFH-DCLK1 (4.8 µM) nanoparticles in colon cancer cells. CFH-DCLK1 nanoparticles induced apoptosis and inhibited the migration and invasion of colon cancer cells. Real time PCR and western blot results have demonstrated that the treatment with CFH-DCLK1 nanoparticles significantly reduced the expression of CSC markers such as DCLK1, STAT1 and NOTCH1 compared to the CFH alone in HCT116 colon cancer cells. Finally, in the 3D spheroid model, CFH-DCLK1 nanoparticles significantly inhibited the colonosphere growth. Overall, our results highlight the effectiveness of CFH-DCLK1 nanoparticles in targeting the colon cancer cells and CSCs. This study would lead to the development of therapies targeting both cancer cells and CSCs simultaneously using nanoformulated drugs, which could bring changes in the current cancer treatment strategies.


Subject(s)
Chitosan , Colonic Neoplasms , Nanoparticles , Cell Line, Tumor , Cell Proliferation , Chitosan/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Doublecortin-Like Kinases , Folic Acid/metabolism , Hesperidin , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neoplastic Stem Cells , Protein Serine-Threonine Kinases
6.
Andrologia ; 53(9): e14160, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34241921

ABSTRACT

The present study was designed to establish the epigenetic mechanisms by which lactational exposure to PCBs affects the Leydig cell function in progeny rats. The lactating dams were oral gavaged with different doses of PCBs [1, 2 and 5 mg/kg or corn oil ] and Leydig cells were isolated from the testes of progeny rats at postnatal day (PND) 60. We assessed the expression of transcription factors regulating steroidogenic machinery and the promoter methylation of LHR and AR in the Leydig cells. Our results confirmed hypermethylation of SF-1, Sp1/3, LHR and AR genes. There was a significant reduction in the gene expression of SF-1 and Sp1. The mRNA expression of Sp3 was decreased. Interestingly, there was an increased gene expression levels of DNA methyltransferases (Dnmts) (Dnmt1, Dnmt3a/b and Dnmt3l) and unaltered histone deacetylase-1 (Hdac-1). Furthermore, increased percentage of 5-methylcytosine was observed in PCBs exposed Leydig cells. Taken together, our findings suggest that promoter hypermethylation of SF-1, Sp1/3, LHR and AR could have led to transcriptional repression of these genes in Leydig cells. In conclusion, our study demonstrates that lactational exposure to PCBs caused epigenetic changes in the Leydig cells which could have impaired the Leydig cell function in progeny (PND60) rats.


Subject(s)
Leydig Cells , Polychlorinated Biphenyls , Animals , Epigenesis, Genetic , Female , Lactation , Male , Polychlorinated Biphenyls/toxicity , Rats , Rats, Wistar , Testosterone
7.
Mol Biol Rep ; 48(4): 3349-3356, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33948855

ABSTRACT

Prostate cancer is the leading cause of death among men worldwide. Deregulation of microRNAs has been reported in many cancers. Expression of microRNAs miR-20a-5p, miR-21-5p, miR-100-5p, miR-125a-5p and miR-146a-5p in tissue blocks of histologically confirmed prostate cancer patients compared with BPH patients, to identify potential microRNA biomarker for prostate cancer. MicroRNA was isolated and expression was quantified by qRT-PCR using Taqman Advanced microRNA assay kits. The interactions between the microRNA:target mRNA were predicted by using bioinformatics tools such as miRwalk and miRTargetlink. The experimentally validated targets were analysed using gprofiler to identify their molecular function, biological process and related pathways. The expression analysis revealed that miR-21 and miR-100 were significantly down-regulated whereas miR-125a was up-regulated in prostate cancer patients. Comparative analysis of the expression levels with tumor grading reveal that miR-100 was significantly down-regulated (p < 0.05) in high grade tumor, indicating that miR-100 associated with prostate cancer. ROC analysis revealed that combined analysis of down-regulated miRNAs (miR-21 and miR-100) shown AUC of 0.72 (95% CI 0.65-0.79). The combined analysis of all five miRNAs showed AUC of 0.87 (95% CI 0.81-0.92). The targets prediction analysis revealed several validated targets including BCL2, ROCK1, EGFR, PTEN, MTOR, NAIF1 and VEGFA. Our results provide evidence that combined analysis of all the five miRNAs as a panel can significantly improve the prediction level of the presence of prostate cancer and may be used as a potential diagnostic biomarker.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Computational Biology , Humans , Male , MicroRNAs/analysis , Middle Aged , Prostatic Neoplasms/metabolism , ROC Curve
8.
Article in English | MEDLINE | ID: mdl-32940190

ABSTRACT

The development of colorectal cancer (CRC) is a multistage process. The inflammation of the colon as in inflammatory bowel disease (IBD) such as ulcerative colitis (UC) or Crohn's disease (CD) is often regarded as the initial trigger for the development of inflammation-associated CRC. Many cytokines such as tumor necrosis factor alpha (TNF-α) and interleukins (ILs) are known to exert proinflammatory actions, and inflammation initiates or promotes tumorigenesis of various cancers, including CRC, through differential regulation of microRNAs (miRNAs/miRs). miRNAs can be oncogenic miRNAs (oncomiRs) or anti-oncomiRs/tumor suppressor miRNAs, and they play key roles during colorectal carcinogenesis. However, the functions and molecular mechanisms of regulation of miRNAs involved in inflammation-associated CRC are still anecdotal and largely unknown. Consolidating the published results and offering perspective solutions to circumvent CRC, the current review is focused on the role of miRNAs and their regulation in the development of CRC. We have also discussed the model systems adapted by researchers to delineate the role of miRNAs in inflammation-associated CRC.


Subject(s)
Colorectal Neoplasms/genetics , Inflammatory Bowel Diseases/complications , MicroRNAs/physiology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colitis/complications , Colitis/genetics , Colitis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Signal Transduction/genetics
9.
Article in English | MEDLINE | ID: mdl-32901590

ABSTRACT

Chronic inflammation can lead to the development of many diseases, including cancer. Inflammatory bowel disease (IBD) that includes both ulcerative colitis (UC) and Crohnmp's disease (CD) are risk factors for the development of colorectal cancer (CRC). Many cytokines produced primarily by the gut immune cells either during or in response to localized inflammation in the colon and rectum are known to stimulate the complex interactions between the different cell types in the gut environment resulting in acute inflammation. Subsequently, chronic inflammation, together with genetic and epigenetic changes, have been shown to lead to the development and progression of CRC. Various cell types present in the colon, such as enterocytes, Paneth cells, goblet cells, and macrophages, express receptors for inflammatory cytokines and respond to tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), IL-6, and other cytokines. Among the several cytokines produced, TNF-α and IL-1ß are the key pro-inflammatory molecules that play critical roles in the development of CRC. The current review is intended to consolidate the published findings to focus on the role of pro-inflammatory cytokines, namely TNF-α and IL-1ß, on inflammation (and the altered immune response) in the gut, to better understand the development of CRC in IBD, using various experimental model systems, preclinical and clinical studies. Moreover, this review also highlights the current therapeutic strategies available (monotherapy and combination therapy) to alleviate the symptoms or treat inflammation-associated CRC by using monoclonal antibodies or aptamers to block pro-inflammatory molecules, inhibitors of tyrosine kinases in the inflammatory signaling cascade, competitive inhibitors of pro-inflammatory molecules, and the nucleic acid drugs like small activating RNAs (saRNAs) or microRNA (miRNA) mimics to activate tumor suppressor or repress oncogene/pro-inflammatory cytokine gene expression.


Subject(s)
Colorectal Neoplasms/etiology , Inflammation/complications , Inflammatory Bowel Diseases/complications , Animals , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colitis/complications , Colitis/pathology , Colorectal Neoplasms/pathology , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/pathology
10.
Stem Cell Investig ; 7: 20, 2020.
Article in English | MEDLINE | ID: mdl-33294429

ABSTRACT

BACKGROUND: To understand the mechanism underlying tamoxifen-induced multidrug resistance (MDR) and stem-like phenotypes in breast cancer cells, we treated the MCF-7 cells with 4-hydroxy-tamoxifen (TAM) for 6 months continuously and established MCF-7 tamoxifen resistance (TR) phenotypes. METHODS: In the present study, the following methods were used: cell viability assay, colony formation, cell cycle analysis, ALDEFLUOR assay, mammosphere formation assay, chromatin immunoprecipitation (ChIP) assay, PCR array, western blot analysis and quantitative reverse transcription polymerase chain reaction (QRT-PCR). RESULTS: The expression of ERα was significantly higher in MCF7-TR cells when compared with parental MCF-7 cells. MCF7-TR cells exposed to TAM showed a significant increase in the proliferation and rate of colony formation. The number of cancer stem cells was higher in MCF7-TR cells as observed by the increase in the number of ALDH+ cells. Furthermore, the number of mammospheres formed from the FACS-sorted ALDH+ cells was higher in MCF7-TR cells. Using PCR array analysis, we were able to identify that the long-term exposure of TAM leads to alterations in the epigenetic and MDR stem cell marker genes. Furthermore, western blot analysis demonstrated elevated levels of Notch-1 expression in MCF-TR cells compared with MCF-7 cells. Chromatin immunoprecipitation (ChIP) assay revealed that Notch-1 enhanced the cyclin D1 expression significantly in these cells. In addition, we observed that MCF7-TR cells were resistant to doxorubicin but not the MCF-7 cells. CONCLUSIONS: In the present study, we conclude that the treatment with tamoxifen induces multiple epigenetic alterations that lead to the development of MDR and stem-like phenotypes in breast cancers. Therefore, our study provides better insights to develop novel treatment regime to control the progression of breast cancer.

11.
Medicina (Kaunas) ; 56(3)2020 Feb 27.
Article in English | MEDLINE | ID: mdl-32120970

ABSTRACT

Background and Objectives: Polycystic ovary syndrome (PCOS) is one of the most prevalent disorders among women of reproductive age. It is considered as a pro-inflammatory state with chronic low-grade inflammation, one of the key factors contributing to the pathogenesis of this disorder. Polycystic ovary is a well-established criterion for PCOS. The present investigation aimed at finding the role of hyperandrogenism, the most important feature of PCOS, in the development of this inflammatory state. To address this problem, we adopted a model system that developed polycystic ovary morphology (PCOM), which could be most effectively used in order to study the role of non-aromatizable androgen in inflammation in PCOS. Materials and Methods: Six rats were used to induce PCOM in 21-days-old female Wistar albino rats by using a pre-determined release of dihydrotestosterone (DHT), a potent non-aromatizable androgen, achieved by implanting a DHT osmotic pump, which is designed to release a daily dose of 83 µg. Results: After 90 days, the rats displayed irregular estrous cycles and multiple ovarian cysts similar to human PCOS. Elevated serum inflammatory markers such as tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), and the presence of a necrotic lesion in the liver, osteoclast in the femur, multinucleated giant cells and lymphocytes in the ovary based on histopathological observation of DHT-treated rats clearly indicated the onset of inflammation in the hyperandrogenic state. Our results show no significant alterations in serum hormones such as luteinizing hormone (LH), follicle stimulating hormone (FSH), insulin, and cortisol between control and hyperandrogenised rats. DHT was significantly elevated as compared to control. mRNA studies showed an increased expression level of TNF-α and IL-1ß, further, the mRNA expression of urocortin 1 (Ucn-1) was stupendously elevated in the liver of hyperandrogenised rats. Conclusions: Thus, results from this study provide: (1) a good PCOM model system in order to study the inflammatory changes in PCOS aspects, (2) alteration of inflammatory markers in PCOM rats that could be either due to its direct effect or by the regulation of various inflammatory genes and markers in the liver of hyperandrogenic state suggesting the regulatory role of DHT, and (3) alteration in stress-related protein in the liver of PCOM rats.


Subject(s)
Cytokines/blood , Dihydrotestosterone/adverse effects , Hyperandrogenism/metabolism , Inflammation Mediators/blood , Polycystic Ovary Syndrome/metabolism , Animals , Disease Models, Animal , Female , Follicle Stimulating Hormone/blood , Hyperandrogenism/chemically induced , Insulin/blood , Liver/metabolism , Luteinizing Hormone/blood , Polycystic Ovary Syndrome/complications , Rats , Rats, Wistar , Urocortins/metabolism
12.
Curr Protein Pept Sci ; 21(1): 52-65, 2020.
Article in English | MEDLINE | ID: mdl-31702489

ABSTRACT

Hormones are known to influence various body systems that include skeletal, cardiac, digestive, excretory, and immune systems. Emerging investigations suggest the key role played by secretions of endocrine glands in immune cell differentiation, proliferation, activation, and memory attributes of the immune system. The link between steroid hormones such as glucocorticoids and inflammation is widely known. However, the role of peptide hormones and amino acid derivatives such as growth and thyroid hormones, prolactin, dopamine, and thymopoietin in regulating the functioning of the immune system remains unclear. Here, we reviewed the findings pertinent to the functional role of hormone-immune interactions in health and disease and proposed perspective directions for translational research in the field.


Subject(s)
Endocrine System Diseases/metabolism , Endocrine System/metabolism , Growth Hormone/metabolism , Immune System Diseases/metabolism , Immune System/metabolism , Prolactin/metabolism , Thymocytes/metabolism , Animals , Cell Communication , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Dopamine/genetics , Dopamine/immunology , Dopamine/metabolism , Endocrine System/cytology , Endocrine System/immunology , Endocrine System Diseases/genetics , Endocrine System Diseases/immunology , Endocrine System Diseases/pathology , Glucocorticoids/genetics , Glucocorticoids/immunology , Glucocorticoids/metabolism , Growth Hormone/genetics , Growth Hormone/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/pathology , Lactotrophs/cytology , Lactotrophs/immunology , Lactotrophs/metabolism , Prolactin/genetics , Prolactin/immunology , Receptors, Dopamine/genetics , Receptors, Dopamine/immunology , Receptors, Dopamine/metabolism , Somatotrophs/cytology , Somatotrophs/immunology , Somatotrophs/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thyroid Hormones/genetics , Thyroid Hormones/immunology , Thyroid Hormones/metabolism
13.
IET Nanobiotechnol ; 12(6): 727-732, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30104445

ABSTRACT

Research dealing with early diagnosis and efficient treatment in colon cancer to improve patient's survival is still under investigation. Chemotherapeutic agent result in high systemic toxicity due to their non-specific actions on DNA repair and/or cell replication. Traditional medicine such as Lycopodium clavatum (LC) has been claimed to have therapeutic potentials against cancer. The present study focuses on targeted drug delivery of cationic liposomal nanoformulated LC (CL-LC) in colon cancer cells (HCT15) and comparing the efficacy with an anti-colon cancer drug, 7-ethyl-10-hydroxy-camptothecin (SN38) along with its nanoformulated form (CL-SN38). The colloidal suspension of LC was made using thin film hydration method. The drugs were characterised using ultraviolet, dynamic light scattering, scanning electron microscopy, energy, dispersive X-ray spectroscopy. Invitro drug release showed kinetics of 49 and 89% of SN38 and LC, whereas CL-SN38 and CL-LC showed 73 and 74% of sustained drug release, respectively. Studies on morphological changes, cell viability, cytotoxicity, apoptosis, cancer-associated gene expression analysis of Bcl-2, Bax, p53 by real-time polymerase chain reaction and western blot analysis of Bad and p53 protein were performed. Nanoformulated LC significantly inhibited growth and increased the apoptosis of colon cancer cells indicating its potential anti-cancer activity against colon cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Liposomes/pharmacology , Lycopodium/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Apoptosis/drug effects , Cations/chemistry , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Delivery Systems , Drug Liberation , Drug Screening Assays, Antitumor , Humans , Liposomes/chemical synthesis , Liposomes/chemistry , Nanocomposites/chemistry , Tumor Cells, Cultured
14.
J Cell Biochem ; 119(7): 6181-6193, 2018 07.
Article in English | MEDLINE | ID: mdl-29626351

ABSTRACT

Parathyroid hormone (PTH) acts on osteoblasts and functions as an essential regulator of calcium homeostasis and as a mediator of bone remodeling. We previously reported that PTH stimulates the expression of matrix metalloproteinase-13 (MMP-13) in rat osteoblasts and that MMP-13 plays a key role in bone remodeling, endochondral bone formation, and bone repair. Recent evidence indicated that microRNAs (miRNAs) have regulatory functions in bone metabolism. In this study, we hypothesized that the down-regulation of miRNAs that target MMP-13 by PTH leads to the stimulation of MMP-13 expression in osteoblasts. We used various bioinformatic tools to identify miRNAs that putatively target rat MMP-13. Among these miRNAs, the expression of miR-532-5p in rat osteoblasts decreased at 4 h of PTH-treatment, whereas MMP-13 mRNA expression was maximal at the same time point. When an miR-532-5p mimic was transiently transfected into UMR-106-01 cells, MMP-13 mRNA and protein expression decreased. Using a luciferase reporter assay system, we also identified that miR-532-5p directly targeted the 3' UTRs of MMP-13 gene. Based on these results, we suggest that PTH-induced down-regulation of miR-532-5p resulted in the stimulation of MMP-13 expression in rat osteoblasts. This study identified a significant role of miRNA in controlling bone remodeling via PTH-stimulated MMP-13 expression. This finding enhances our understanding of bone metabolism and bone-related diseases and it could provide information regarding the usage of miRNAs as therapeutic agents or biomarkers.


Subject(s)
Gene Expression Regulation/drug effects , Matrix Metalloproteinase 13/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Animals , Cells, Cultured , Down-Regulation , Matrix Metalloproteinase 13/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Rats
15.
Reprod Toxicol ; 75: 73-85, 2018 01.
Article in English | MEDLINE | ID: mdl-29208485

ABSTRACT

The present study was aimed to determine the effects of lactational exposure of PCBs (Aroclor 1254) on Leydig cellular steroidogenesis in F1 progeny rats. Lactating dams were orally treated by gavage with different doses of PCBs (1, 2 and 5mg/kg b.wt./day). Male progenies were sacrificed on PND60. Our results demonstrated that exposure to PCBs decreased the body weight, testis weight and anogenital distance (AGD) index in the F1 progeny rats. Importantly, PCBs exposure reduced the serum levels of LH, testosterone and estradiol. Interestingly, PCBs caused a decrease in the Leydig cell population along with decreased activities of steroidogenic enzymes 3ß- and 17ß-HSD. Additionally, we observed a significant decrease in LHR, SR-B1, StAR protein, Cyp11a1, 3ß-HSD, Cyp17a1, 17ß-HSD, 5α-reductase, Cyp19a1 and AR gene expression in the Leydig cells of progeny rats. In conclusion, our study demonstrates that lactational exposure of PCBs alters Leydig cellular steroidogenesis in the F1 progeny rats.


Subject(s)
Endocrine Disruptors/toxicity , Estradiol/blood , Leydig Cells/drug effects , Maternal Exposure/adverse effects , Polychlorinated Biphenyls/toxicity , Testosterone/blood , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Animals , Animals, Newborn , Aromatase/genetics , Female , Gene Expression/drug effects , Lactation , Leydig Cells/metabolism , Male , Pregnancy , Rats, Wistar , Receptors, Androgen/genetics , Testis/drug effects , Testis/metabolism , Testis/pathology
16.
Int J Biol Macromol ; 107(Pt B): 1988-1998, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29032208

ABSTRACT

Cancer is one of the major causes of increased morbidity and mortality in modern society. Colorectal cancer is the third leading cause for cancer related death worldwide. Current chemotherapeutics are not very effective and have severe side effects. Hesperetin is a bioflavonoid from citrus fruits and its clinical use is restricted because of the poor water solubility. Folate receptor is overexpressed in various cancer cells. Therefore, we synthesized the chitosan folate hesperetin nanoparticle (CFH) by covalently conjugating folic acid with chitosan molecules. The size of the CFH nanoparticles is around 450nm, which is advantageous for passively targeting the cancer cell specifically due to the leaky vasculature of the tumour. Particle surface and size were observed using SEM and TEM studies. The results show that hesperetin has an IC50 value of 190µM and it induces apoptosis in HCT15 cells, however, CFH is very potent in inhibiting the proliferation with the IC50 value of 28µM. In addition, CFH inhibited colony formation and induced apoptosis by regulating the expression of proapoptotic genes expression. Therefore, the chitosan - folic acid conjugation appears to be the suitable carrier for colorectal cancer cell-specific delivery of hesperetin.


Subject(s)
Antineoplastic Agents/pharmacology , Chitosan/chemistry , Hesperidin/pharmacology , Nanoparticles/chemistry , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Drug Delivery Systems , Drug Liberation , Gene Expression Regulation, Neoplastic/drug effects , Hesperidin/chemistry , Humans , Particle Size , Powders , Spectroscopy, Fourier Transform Infrared , Static Electricity , X-Ray Diffraction
17.
PLoS One ; 12(5): e0177780, 2017.
Article in English | MEDLINE | ID: mdl-28542301

ABSTRACT

BACKGROUND: Electronic cigarette (EC) aerosols contain unique compounds in addition to toxicants and carcinogens traditionally found in tobacco smoke. Studies are warranted to understand the public health risks of ECs. OBJECTIVE: The aim of this study was to determine the genotoxicity and the mechanisms induced by EC aerosol extracts on human oral and lung epithelial cells. METHODS: Cells were exposed to EC aerosol or mainstream smoke extracts and DNA damage was measured using the primer anchored DNA damage detection assay (q-PADDA) and 8-oxo-dG ELISA assay. Cell viability, reactive oxygen species (ROS) and total antioxidant capacity (TAC) were measured using standard methods. mRNA and protein expression were evaluated by RT-PCR and western blot, respectively. RESULTS: EC aerosol extracts induced DNA damage in a dose-dependent manner, but independently of nicotine concentration. Overall, EC aerosol extracts induced significantly less DNA damage than mainstream smoke extracts, as measured by q-PADDA. However, the levels of oxidative DNA damage, as indicated by the presence of 8-oxo-dG, a highly mutagenic DNA lesion, were similar or slightly higher after exposure to EC aerosol compared to mainstream smoke extracts. Mechanistically, while exposure to EC extracts significantly increased ROS, it decreased TAC as well as the expression of 8-oxoguanine DNA glycosylase (OGG1), an enzyme essential for the removal of oxidative DNA damage. CONCLUSIONS: Exposure to EC aerosol extracts suppressed the cellular antioxidant defenses and led to significant DNA damage. These findings emphasize the urgent need to investigate the potential long-term cancer risk of exposure to EC aerosol for vapers and the general public.


Subject(s)
Antioxidants/metabolism , DNA Damage , Electronic Nicotine Delivery Systems/adverse effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Aerosols , Cell Line , Dose-Response Relationship, Drug , Humans , Smoke/adverse effects , Time Factors
18.
Reprod Toxicol ; 69: 84-98, 2017 04.
Article in English | MEDLINE | ID: mdl-28192182

ABSTRACT

The effect of gestational exposure to CrVI (occupational/environmental pollutant and target to Sertoli cells(SC)) was tested in a rat model during the testicular differentiation from the bipotential gonad may interrupt spermatogenesis by disrupting SC tight junctions(TJ) and it's proteins and hormone receptors. Pregnant Wistar rats were exposed to 50/100/200ppm CrVI through drinking water during embryonic days 9-14. On Postnatal day 120, testes were subjected to ion exchange chromatographic analysis and revealed increased level of CrIII in SCs and germ cells, serum and testicular interstitial fluid(TIF). Microscopic analyses showed seminiferous tubules atrophy and disruption of SC TJ, which also recorded decreased testosterone in TIF. mRNA and Protein expression analyses attested decreased level of Fshr, Ar, occludin and claudin-11 in SCs. Immunofluorescent detection revealed weak signal of TJ proteins. Taken together, we concluded that gestational exposure to CrVI interferes with the expression of SC TJ proteins due to attenuated expression of hormone receptors.


Subject(s)
Chromium/toxicity , Prenatal Exposure Delayed Effects , Testis/drug effects , Water Pollutants, Chemical/toxicity , Animals , Chromium/blood , Claudins/genetics , Claudins/metabolism , Female , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Male , Maternal-Fetal Exchange , Microscopy, Electron, Transmission , Occludin/genetics , Occludin/metabolism , Pregnancy , RNA, Messenger/metabolism , Rats, Wistar , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Sperm Motility/drug effects , Testis/metabolism , Testis/pathology , Testis/ultrastructure , Testosterone/blood , Water Pollutants, Chemical/blood
19.
Reprod Sci ; 23(4): 549-59, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26482208

ABSTRACT

Phthalates, a class of chemicals used as plasticizers, are economically important due to several industrial applications. Di-(2-ethylhexyl) phthalate (DEHP) is the most commonly used phthalate plasticizer, and it has been described as a potent antiandrogen in males. In this study, lactating dams were exposed via oral gavage to corn oil (vehicle) and DEHP (1, 10, and 100 mg/kg body weight) from postnatal day 1 to 21, and the effects were evaluated in the ovary and uterus of F(1) progeny. DEHP exposure significantly decreased the body weight and organ weight in a dose-dependent manner. Serum levels of estradiol, testosterone, and progesterone were decreased but anogenital distance was unaffected. The mRNA expressions of luteinizing hormone receptor, follicle-stimulating hormone receptor, androgen receptor, estrogen receptor (ERα and ERß), progesterone receptor, peroxisome proliferator-activated receptor γ, 3ß hydroxysteroid dehydrogenase, aromatase, and steroidogenic acute regulatory protein were altered in the ovary of F1 progeny rats. Our finding suggest that lactational exposure to DEHP has transgenerational effect on female reproductive system.


Subject(s)
Diethylhexyl Phthalate/toxicity , Lactation/drug effects , Ovary/drug effects , Ovary/physiology , Uterus/drug effects , Uterus/physiology , Age Factors , Animals , Dose-Response Relationship, Drug , Female , Lactation/physiology , Plasticizers/toxicity , Rats , Rats, Wistar
20.
Am J Prev Med ; 48(1 Suppl 1): S102-10, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25528699

ABSTRACT

BACKGROUND: Mainstream (MS) smoke, the main smoke inhaled by active smokers, and sidestream (SS) smoke, the main component of secondhand smoke, induce a wide range of DNA lesions. Owing to technical limitations, the in vivo levels of tobacco-induced DNA damage are unknown. Recently, the authors developed a highly sensitive primer-anchored DNA damage detection assay (PADDA) to quantify endogenous and induced DNA damage. PURPOSE: To quantify the in vivo levels of DNA damage induced by MS and SS smoke extracts in human cells using PADDA and define the strand-specific patterns of DNA damage and repair following exposure to diverse doses of MS and SS smoke. METHODS: Human epithelial cells were exposed to escalating doses of hydrogen peroxide (H2O2), MS, or SS smoke. TP53 gene DNA damage was quantified using PADDA at various time points. DNA double-strand breaks were detected by immunofluorescence analysis of phosphorylated histone H2AX (γ-H2AX). Cell viability was determined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay. Data were collected and analyzed by t-test in 2012-2014. RESULTS: A dose-dependent increase in DNA damage was detected in vivo with increasing doses of H2O2, MS, and SS smoke. Even 1 hour of exposure to very low doses of MS or SS smoke resulted in significant DNA damage (p<0.01). MS and SS smoke induced distinctive strand-specific patterns of DNA damage and DNA repair kinetics. CONCLUSIONS: Very low concentrations of MS and SS smoke induce significant DNA damage in human cells. Application of PADDA to population studies has major potential to establish biomarkers of susceptibility to tobacco-induced diseases.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Smoke/adverse effects , Tobacco Smoke Pollution/adverse effects , Cell Line , Cell Survival/drug effects , DNA Breaks, Double-Stranded/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/pathology , Histones/genetics , Humans , Hydrogen Peroxide/administration & dosage , Hydrogen Peroxide/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...