Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Sci Rep ; 12(1): 22420, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36575233

ABSTRACT

Epidermal growth factor receptor (EGFR) is an effective target for those patients with metastatic colorectal cancers that retain the wild-type RAS gene. However, its efficacy in many cancers, including bladder cancer, is unclear. Here, we studied the in vitro effects of cetuximab monoclonal antibodies (mAbs) targeting EGFR on the bladder cancer cells and role of CD46. Cetuximab was found to inhibit the growth of both colon and bladder cancer cell lines. Furthermore, cetuximab treatment inhibited AKT and ERK phosphorylation in the bladder cancer cells and reduced the expression of CD46 membrane-bound proteins. Restoration of CD46 expression protected the bladder cancer cells from cetuximab-mediated inhibition of AKT and ERK phosphorylation. We hypothesized that CD46 provides protection to the bladder cancer cells against mAb therapies. Bladder cancer cells were also susceptible to cetuximab-mediated immunologic anti-tumor effects. Further, cetuximab enhanced the cell killing by activating both antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in bladder cancer cells. Restoration of CD46 expression protected the cells from both CDC and ADCC induced by cetuximab. Together, CD46 exhibited a cancer-protective effect against both direct (by involvement of PBMC or complement) and indirect cytotoxic activity by cetuximab in bladder cancer cells. Considering its clinical importance, CD46 could be an important link in the action mechanism of ADCC and CDC intercommunication and may be used for the development of novel therapeutic strategies.


Subject(s)
Antineoplastic Agents , Urinary Bladder Neoplasms , Humans , Cetuximab/pharmacology , Leukocytes, Mononuclear/metabolism , Proto-Oncogene Proteins c-akt , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antibodies, Monoclonal/pharmacology , ErbB Receptors/metabolism , Antibody-Dependent Cell Cytotoxicity , Urinary Bladder Neoplasms/drug therapy , Membrane Cofactor Protein
2.
Am J Cancer Res ; 12(10): 4853-4864, 2022.
Article in English | MEDLINE | ID: mdl-36381334

ABSTRACT

δ-Catenin is expressed abundantly in various human cancers, including prostate, brain, breast, and lung carcinomas, and is recognized as an oncogene that promotes cancer cell growth and tumorigenesis. Although several transcriptional and post-translational pathways for δ-catenin regulation have been identified in cancer cells, the potential effects of microRNA-mediated regulation remain elusive. Here, we used a δ-catenin 3'-UTR luciferase reporter assay to identify regulatory microRNAs. Subsequent bioinformatics analyses and molecular studies revealed that overexpression of miR-122 downregulated δ-catenin expression significantly via targeted binding to a seed sequence in the 3'-UTR region of δ-catenin, and suppressed the invasion, migration, and proliferation of prostate cancer cells in vitro. In a TRAMP-C2 mouse syngeneic prostate tumor model, stable expression of miR-122 decreased both δ-catenin expression and tumor growth. Mechanistically, overexpression of miR-122 inhibited the expression of δ-catenin-mediated downstream factors significantly in prostate cancer cells, including c-myc and cyclin D1. In cells overexpressing miR-122, there was no additive or synergistic effect of siRNA-mediated knockdown of δ-catenin on cell invasiveness, and overexpression of miR-122 alone had a more pronounced suppressive effect on cell invasion than knockdown of δ-catenin alone. These results suggest that miR-122 acts as tumor suppressor in prostate cancer, mainly by downregulating δ-catenin expression, but also by targeting other factors. Indeed, subsequent experiments showed that overexpression of miR-122 reduced the levels of the mRNAs encoding myc, snail, and VEGF in prostate cancer cells. Overall, our findings demonstrate that targeting of δ-catenin by miR-122 represses the motility and tumorigenesis of prostate cancer cells, indicating a tumor suppressive effect of this miRNA in prostate cancer.

3.
Int J Mol Sci ; 21(8)2020 Apr 23.
Article in English | MEDLINE | ID: mdl-32340289

ABSTRACT

Zinc is a group IIB heavy metal. It is an important regulator of major cell signaling pathways in most mammalian cells, functions as an antioxidant and plays a role in maintaining genomic stability. Zinc deficiency leads to severe diseases in the brain, pancreas, liver, kidneys and reproductive organs. Zinc loss occurs during tumor development in a variety of cancers. The prostate normally contains abundant intracellular zinc and zinc loss is a hallmark of the development of prostate cancer development. The underlying mechanism of this loss is not clearly understood. The knowledge that excess zinc prevents the growth of prostate cancers suggests that zinc-mediated therapeutics could be an effective approach for cancer prevention and treatment, although challenges remain. This review summarizes the specific roles of zinc in several cancer types focusing on prostate cancer. The relationship between prostate cancer and the dysregulation of zinc homeostasis is examined in detail in an effort to understand the role of zinc in prostate cancer.


Subject(s)
Prostatic Neoplasms/metabolism , Zinc/metabolism , Animals , Biological Transport , Clinical Studies as Topic , Disease Susceptibility , Drug Delivery Systems , Drug Evaluation, Preclinical , Homeostasis , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Treatment Outcome , Zinc/pharmacology , Zinc/therapeutic use , Zinc Fingers
4.
Int J Mol Sci ; 19(10)2018 Oct 08.
Article in English | MEDLINE | ID: mdl-30297600

ABSTRACT

The prostate gland contains a high level of intracellular zinc, which is dramatically diminished during prostate cancer (PCa) development. Owing to the unclear role of zinc in this process, therapeutic applications using zinc are limited. This study aimed to clarify the role of zinc and its underlying mechanism in the growth of PCa. ZnCl2 suppressed the proliferation of androgen receptor (AR)-retaining PCa cells, whereas it did not affect AR-deficient PCa cells. In LNCaP and TRAMP-C2 cells, zinc downregulated the expression of AR in a dose- and time-dependent fashion. Zinc-mediated AR suppression accordingly inhibited the androgen-mediated transactivation and expression of the androgen target, prostate specific antigen (PSA). This phenomenon resulted from facilitated protein degradation, not transcriptional control. In studies using mice bearing TRAMP-C2 subcutaneous tumors, the intraperitoneal injection of zinc significantly reduced tumor size. Analyses of both xenograft tumors and normal prostates showed reduced expression of AR and increased cell death. Considering the significant loss of intracellular zinc and the dominant growth-modulating role of AR during PCa development, loss of zinc may be a critical step in the transformation of normal cells to cancer cells. This study provides the underlying mechanism by which zinc functions as a PCa suppressor, and forms the foundation for developing zinc-mediated therapeutics for PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Zinc Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Zinc Compounds/therapeutic use
5.
Int J Mol Sci ; 19(9)2018 Sep 10.
Article in English | MEDLINE | ID: mdl-30201920

ABSTRACT

CD46 is generally overexpressed in many human cancers, representing a prime target for CD46-binding adenoviruses (Ads). This could help to overcome low anti-tumoral activity by coxsackie-adenoviral receptor (CAR)-targeting cancer gene therapy viruses. However, because of scarce side-by-side information about CAR and CD46 expression levels in cancer cells, mixed observations of cancer therapeutic efficacy have been observed. This study evaluated Ad-mediated therapeutic efficacy using either CAR-targeting Ad5 or CD46-targeting Ad5/35 fiber chimera in bladder cancer cell lines. Compared with normal urothelia, bladder cancer tissue generally overexpressed both CAR and CD46. While CAR expression was not correlated with disease progression, CD46 expression was inversely correlated with tumor grade, stage, and risk grade. In bladder cancer cell lines, expression levels of CD46 and CAR were highly correlated with Ad5/35- and Ad5-mediated gene transduction and cytotoxicity, respectively. In a human EJ bladder cancer xenograft mouse model, with either overexpressed or suppressed CD46 expression levels, Ad5/35-tk followed by ganciclovir (GCV) treatment significantly affected tumor growth, whereas Ad5-tk/GCV had only minimal effects. Overall, our findings suggest that bladder cancer cells overexpress both CAR and CD46, and that adenoviral cancer gene therapy targeting CD46 represents a more suitable therapy option than a CAR-targeting therapy, especially in patients with low risk bladder cancers.


Subject(s)
Adenoviridae/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Membrane Cofactor Protein/metabolism , Thymidine Kinase/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/therapy , Aged , Animals , Cell Line, Tumor , Female , Ganciclovir/administration & dosage , Ganciclovir/pharmacology , Gene Expression Regulation, Neoplastic , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Male , Mice , Middle Aged , Neoplasm Grading , Survival Analysis , Transduction, Genetic , Up-Regulation , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Oncol Rep ; 35(4): 2011-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26781690

ABSTRACT

Many prostate cancer (PCa) patients die of recurrent disease due to the emergence of hormone-independent cancer cells of which the mechanism is not fully understood. Our previous studies demonstrated that most castration- resistant prostate cancers (CRPC) overexpress the HOXB13 transcription factor to confer positive growth signals. Since HOXB13 also suppresses p21WAF1/CIP1 (p21) expression, we studied the correlation between HOXB13 and p21 in selected samples of PCa. While there was no statistically significant correlation between expression of HOXB13 and p21, HOXB13-deficient tumors had three times higher odds for expressing p21 than HOXB13-positive tumors. Moreover, CRPC showed more negative correlation than hormone-dependent PCa (HDPC). Further in vitro proliferation assay demonstrated that androgen did not affect the growth-suppressive function of p21 in androgen-dependent PCa cells, suggesting that p21 seems to override the growth-promoting function of androgen and suppression of p21 expression by HOXB13 is an important step in PCa cell survival under no androgen influence. HOXB13 also inhibited AP-1 signals via suppressed expression of JNK/c-Jun. While HOXB13 suppressed p21 expression via regulation of JNK signals, alteration of p21 expression also affected c-Jun and AP-1 activity. Taken together, overexpression of HOXB13 in CRPC is an important step in avoiding the growth-suppressive effect of p21 in a harsh condition such as an androgen-deprived environment.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Homeodomain Proteins/metabolism , Prostatic Neoplasms/metabolism , Aged , Aged, 80 and over , Androgens/pharmacology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Male , Middle Aged , Prostatic Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL