115 results on '"Navone N"'
Search Results
2. Glutathione-S-transferase (GST) polymorphisms are associated with relapse after radical prostatectomy
- Author
-
Cotignola, J, Leonardi, D B, Shahabi, A, Acuña, A D, Stern, M C, Navone, N, Scorticati, C, De Siervi, A, Mazza, O, and Vazquez, E
- Published
- 2013
- Full Text
- View/download PDF
3. Constitutive activation of p38 MAPK in tumor cells contributes to osteolytic bone lesions in multiple myeloma
- Author
-
Yang, J, He, J, Wang, J, Cao, Y, Ling, J, Qian, J, Lu, Y, Li, H, Zheng, Y, Lan, Y, Hong, S, Matthews, J, Starbuck, M W, Navone, N M, Orlowski, R Z, Lin, P, Kwak, L W, and Yi, Q
- Published
- 2012
- Full Text
- View/download PDF
4. Heme oxygenase 1 (HO-1) challenges the angiogenic switch in prostate cancer
- Author
-
Ferrando, M., Gueron, G., Elguero, B., Giudice, J., Salles, A., Leskow, F. Coluccio, Jares-Erijman, E. A., Colombo, L., Meiss, R., Navone, N., De Siervi, A., and Vazquez, E.
- Published
- 2011
- Full Text
- View/download PDF
5. A 45-kDa ErbB3 secreted by prostate cancer cells promotes bone formation
- Author
-
Lin, S-H, Cheng, C-J, Lee, Y-C, Ye, X, Tsai, W-W, Kim, J, Pasqualini, R, Arap, W, Navone, N M, Tu, S-M, Hu, M, Yu-Lee, L-Y, and Logothetis, C J
- Published
- 2008
- Full Text
- View/download PDF
6. Low-density lipoprotein receptor-related protein 5 (LRP5) mediates the prostate cancer-induced formation of new bone
- Author
-
Li, Z G, Yang, J, Vazquez, E S, Rose, D, Vakar-Lopez, F, Mathew, P, Lopez, A, Logothetis, C J, Lin, S-H, and Navone, N M
- Published
- 2008
- Full Text
- View/download PDF
7. Supplement II: Abstracts of the international symposium on Skin Carcinogenesis in man and in experimental models. Heidelberg, 29–31 October 1991 (pp S61–S88)
- Author
-
Barrett, J. C., Afshari, C. A., Annab, L. A., Burkhart, B. A., Boyd, J. A., Owen, R. D., Futreal, P. A., Richter, K. H., Moses, H. L., Lavker, R. M., Miller, Stanley, Sun, T. -T., Stingl, G., Bianchi, A. B., Navone, N. M., Conti, C. J., Spencer, James M., Kahn, S., Weinstein, I. B., Silvers, D. S., DeLeo, V. A., Larcher, F., Bauluz, C., Quintanilla, M., Ballestin, C., Jorcano, J. L., Schön, M., Haas, M., Klein, C. E., Weber, L., Cerri, A., Tadini, G., Gitto, R., Berti, E., Cano, A., Caulín, C., Gómez, M., Gandarillas, A., Martín, M., Montes, A., Navarro, P., Bastian, B. C., Van der Piepen, U., Römisch, J., Pâques, E., Hartmann, A. A., Krieg, P., Schnapke, R., Feil, S., Fürstenberger, G., Marks, F., Missero, C., Cajal, S. Ramon y, Filvaroff, E., Dotto, G. P., Sherman, J., Albert, R. E., Baxter, C. S., Bauer, G., Höfler, P., Götschl, M., Viesel, E., Jürgensmeier, J., Schaefer, D., Picht, G., Grande, T., Real, A., Rünqer, T. M., Möller, K., Fuchs, P., Bauer, C., Epe' B., Gruner, S., Diezel, W., Macejewski, J., Weber, H., Eckert, R., Volk, H. D., Sönnichsen, N., Bavinck, Jan N. Bouwes, Vermeer, Bert J., Van Der Woude, Fokko J., Vandenbroucke, Jan P., Claas, Frans H. J., Griffin, E. F., Harris, H., Tilgen, W., Garbe, C., Østerlind, Anne, Weiss, J., Jung, E. G., Ruiter, D. J., Danen, E., Broecker, E. -B., Johnson, J. P., van Muijen, G. N. P., Halaban, R., Krüger-Krasagakes, S., Orfanos, C. E., Newton, J. A., Bataille, V., Cuzick, J., Bishop, T., Schwaaf, A., Azizi, E., Bröcker, E. B., Eberlein, B., Froschermaier, S., Gollhausen, R., Przybilla, B., Krasagakis, K., Abdel-Naser, M. B., Lopez-Bran, E., Robledo, A., Lopez-Bran, E., Heine, H., Hennig, B., Graf, G., Nährig, J., Niedner, R., Schöpf, E., Mailhammer, R., Reisbach, G., Kempkes, B., Hültner, L., Thalmeier, K., Anders, F., Zechel, C., Schleenbecker, U., Leers, J., Smith, A., Wagner, E., Burcin, U., Hug, H., Fiebich, B., Anders, A., Gröger, H., Schlatterer, B., Moll, I., Wollina, U., Leigh IM, Purkis PE, Markey A., Neill S., Proby C., Glover M., Lane EB, Klein-Szanto, A. J. P., Yaar, M., Garmyn, M., Gilani, A., Gilchrest, B. A., Bowden, G. T., Nelson, M., Levy, J., Tanooka, Hiroshi, Ootsuyama, Akira, Urbach, F., van der Leun, J. C., de Gruijl, F. R., Kripke, Margaret L., Yuspa, S. H., Glick, A., Lee, E., Diugosz, A., Balmain, A., Bums, P., Kemp, C. J., Stoler, A. B., Harks, F., Boukamp, P., Pascheberg, U., Breitkreutz, D., Hülsen, A., Altmeier, S., Tomakidi, P., Fusenig, N. E., Lowy, Douglas R., Sedman, Sylvia A., Cohen, Bruce D., Schiller, John T., Kricker, A., Armstrong, B. K., English, D., Heenan, P. J., Randell, P. L., de Gruijl, F. R., Kelfkens, G., van Weelden, H., van der Leun, J. C., Grabbe, S., Bruvers, S., Granstein, R. D., Albert, R., Miller, M., Cody, T., Baxter, C., Shukla, R., Ueda, M., Ichihashi, M., Yamamura, K., Hayashibe, K., Funasaka, Y., Mishima, Y., Fujiwara, Y., Ichihashi, M., Jimbo, T., Mishima, Y., Popanda, O., Thielmann, H. W., Jahrens, D., Edler, L., Ootsuyama, A., Tanooka, H., Sutter, C., Mukhtar, H., Strickland, P. T., Winter, H., Schweizer, J., Schmidt, R., Weber, E., Rippmann, F., Hecker, E., Kopp-Schneider, A., Lehmann, W. D., Stephan, M., Troll, W., Wei, H., Fujiki, H., Garte, S. J., Frenkel, K., Svetek, J., Schara, M., Pečar, S., Hergenhahn, M., Kinzel, V., Richards, J., Plein, P., Schiess, K., Kaszkin, M., Yamamoto, S., Wang, J. C., Kato, R., Kuroki, T., Hashimoto, Y., Osada, S., Ohno, S., Gilles, C., Piette, M., Foidart, J. -M., Ranki, A., Lassus, J., Lehmus, A., Niemi, K. -M., Friesel, H., Schneider, T., Steinbauer, B., Sorg, B., Winter, A., Krauter, G., Krauß, R., Roeser, H., Unger, Sylvia, Janiaud, Paul, Rueß, Doris, Mechler, Bernard M., Stanbridge, Eric J., Gross, Monika M., Buček, M., Klein-Bauernschmitt, P., Schlehofer, J. R., Kosters, R., Stark, H. -J., Okulov, V. B., Elgjo, K., Ushmorov, A. G., Danilov, A. O., Zubova, S. G., Furstenberger, G., and Faissner, A.
- Published
- 1991
- Full Text
- View/download PDF
8. Erratum: Constitutive activation of p38 MAPK in tumor cells contributes to osteolytic bone lesions in multiple myeloma
- Author
-
Yang, J, He, J, Wang, J, Cao, Y, Ling, J, Qian, J, Lu, Y, Li, H, Zheng, Y, Lan, Y, Hong, S, Matthews, J, Starbuck, M W, Navone, N M, Orlowski, R Z, Lin, P, Kwak, L W, and Yi, Q
- Published
- 2015
- Full Text
- View/download PDF
9. P3.11-25 Analysis Indicates Low Incremental Cost-Effectiveness Ratio for Implementation of Lung Cancer Screening in Italy
- Author
-
Veronesi, G., Ghislandi, S., Vanni, E., Dieci, E., Toschi, L., Velutti, L., Solinas, M., Novellis, P., Alloisio, M., Riboli, E., and Navone, N.
- Published
- 2018
- Full Text
- View/download PDF
10. PO-259 Identification of a clinically meaningful site-specific steroid roadmap in prostate cancer
- Author
-
Sakellakis, M., Ramachandran, S., Efstathiou, E., Mao, X., Hoang, A., Navone, N., Logothetis, C., and Titus, M.
- Published
- 2018
- Full Text
- View/download PDF
11. Detection of loss of heterozygosity in formalin-fixed paraffin-embedded tumor specimens by the polymerase chain reaction
- Author
-
Bianchi, A. B., Navone, N. M., and Conti, C. J.
- Subjects
Heterozygote ,Mice ,Skin Neoplasms ,Formaldehyde ,Histological Techniques ,Molecular Sequence Data ,Tumor Cells, Cultured ,Animals ,Chromosome Mapping ,Amino Acid Sequence ,Polymerase Chain Reaction ,Alleles ,Research Article - Abstract
A polymerase chain reaction-based procedure was used for the detection of DNA length polymorphisms generated by naturally occurring genetic deletions or insertions of known sequence. This method consists of a simple one-step assay that does not require any restriction enzyme analysis or Southern blot hybridization, allowing identification in ethidium bromide-stained gels. The procedure described here was used to detect loss of heterozygosity at various loci, including the Hbb beta-globin gene cluster, in chemically induced mouse skin tumors, using a variety of tissue preparations, including microdissection of formalin-fixed, paraffin-embedded specimens, short-term cultures, and fluorescence-activated cell sorting of epithelial populations. This approach may be useful in detecting tumor-specific reduction to homozygosity at polymorphic chromosomal loci, allowing the mapping of putative tumor-suppressor loci involved in carcinogenesis.
- Published
- 1991
12. Nuclear translocation of haeme oxygenase-1 is associated to prostate cancer.
- Author
-
Sacca, P., Meiss, R., Casas, G., Mazza, O., Calvo, J. C., Navone, N., and Vazquez, E.
- Subjects
HEME oxygenase ,PROSTATE cancer ,OXIDATIVE stress ,INFLAMMATION ,CARCINOGENESIS ,PROSTATECTOMY ,BENIGN prostatic hyperplasia ,IMMUNOHISTOCHEMISTRY ,BIOLOGICAL transport ,CELL nuclei ,COMPARATIVE studies ,RESEARCH methodology ,MEDICAL cooperation ,OXIDOREDUCTASES ,PROSTATE tumors ,RESEARCH ,EVALUATION research ,CANCER cell culture ,METALLOPORPHYRINS - Abstract
The role of oxidative stress in prostate cancer has been increasingly recognised. Acute and chronic inflammations generate reactive oxygen species that result in damage to cellular structures. Haeme oxygenase-1 (HO-1) has cytoprotective effects against oxidative damage. We hypothesise that modulation of HO-1 expression may be involved in the process of prostate carcinogenesis and prostate cancer progression. We thus studied HO-1 expression and localisation in 85 samples of organ-confined primary prostate cancer obtained via radical prostatectomy (Gleason grades 4-9) and in 39 specimens of benign prostatic hyperplasia (BPH). We assessed HO-1 expression by immunohistochemical staining. No significant difference was observed in the cytoplasmic positive reactivity among tumours (84%), non-neoplastic surrounding parenchyma (89%), or BPH samples (87%) (P=0.53). Haeme oxygenase-1 immunostaining was detected in the nuclei of prostate cancer cells in 55 of 85 (65%) patients but less often in non-neoplastic surrounding parenchyma (30 of 85, 35%) or in BPH (9 of 39, 23%) (P<0.0001). Immunocytochemical and western blot analysis showed HO-1 only in the cytoplasmic compartment of PC3 and LNCaP prostate cancer cell lines. Treatment with hemin, a well-known specific inducer of HO-1, led to clear nuclear localisation of HO-1 in both cell lines and highly induced HO-1 expression in both cellular compartments. These findings have demonstrated, for the first time, that HO-1 expression and nuclear localisation can define a new subgroup of prostate cancer primary tumours and that the modulation of HO-1 expression and its nuclear translocation could represent new avenues for therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
13. p53 protein accumulation and gene mutation in the progression of human prostate carcinoma.
- Author
-
Navone, Nora M., Troncoso, Patricia, Pisters, Louis L., Goodrow, Tamra L., Palmer, Judy L., Nichols, Warren W., Eschenbach, Andrew C. von, Conti, Claudio J., Navone, N M, Troncoso, P, Pisters, L L, Goodrow, T L, Palmer, J L, Nichols, W W, von Eschenbach, A C, and Conti, C J
- Abstract
Background: Nuclear accumulation of p53 protein has been shown to be strongly associated with missense p53 mutations. Studies of nuclear accumulation of p53 protein in prostate carcinoma cells have to date been confined to material from primary tumors.Purpose: We studied the accumulation of p53 protein in specimens obtained from primary and metastatic sites of prostate carcinoma. By examining the accumulation of this protein as a function of stage, histologic grade, and androgen responsiveness of the tumor, we hoped to determine the role of p53 mutation in the progression of prostate carcinoma.Methods: The accumulation of the p53 protein in the cell nuclei was determined by immunohistochemical methods using polyclonal antibody to human p53 CM-1. The material studied consisted of formalin-fixed, paraffin-embedded tissue obtained from primary tumors and metastases of 92 patients with prostate carcinoma. Twelve samples from 11 patients were analyzed for the presence of mutations within exons 5-8 of the p53 gene (also known as TP53) by polymerase chain reaction-single-stranded conformation polymorphism (PCR-SSCP) analysis. Sequence analysis was subsequently performed on DNA obtained by polymerase chain reaction amplification of PCR-SSCP reactions produced from six different specimens. The chi-square test, Fisher's exact test, and the Freeman Halton test were used for statistical analyses of the results.Results: All tumors with p53 accumulation were metastatic (stage D), poorly differentiated, and androgen independent. Nuclear accumulation of p53 protein was strongly associated with stage (D2 versus D1 versus A-C, P < .0001), grade (Gleason score 8-10 versus 5-7, P < .003), and androgen sensitivity (androgen independent versus dependent, P < .0001). Logistic regression analysis demonstrated that androgen sensitivity predicted p53 outcome better than did stage (P < .0001) or grade alone (P < .006). There was a perfect concordance between the results obtained by PCR-SSCP analysis and the p53 protein accumulation determined by immunohistochemistry in the 12 samples studied. Mutation of the p53 gene was confirmed by sequencing DNA obtained from six specimens positive in the PCR-SSCP assay.Conclusions: p53 gene mutation is a late event in the progression of prostate cancer and is associated with advanced (metastatic) stage, loss of differentiation, and the transition from androgen-dependent to androgen-independent growth.Implication: Testing of prostate cancer biopsy specimens from metastatic sites for p53 protein accumulation and gene mutation may provide useful prognostic information and could influence the recommended course of treatment. [ABSTRACT FROM AUTHOR]- Published
- 1993
14. Constitutive activation of p38 MAPK in tumor cells contributes to osteolytic bone lesions in multiple myeloma.
- Author
-
Yang, J, He, J, Wang, J, Cao, Y, Ling, J, Qian, J, Lu, Y, Li, H, Zheng, Y, Lan, Y, Hong, S, Matthews, J, Starbuck, M W, Navone, N M, Orlowski, R Z, Lin, P, Kwak, L W, and Yi, Q
- Subjects
MITOGEN-activated protein kinases ,MULTIPLE myeloma ,TUMORS - Abstract
A correction to the article "Constitutive activation of p38 MAPK in tumor cells contributes to osteolytic bone lesions in multiple myeloma" published in the volume 26, 2012 issue is presented.
- Published
- 2015
- Full Text
- View/download PDF
15. Prioritizing genes associated with prostate cancer development
- Author
-
Troncoso Patricia, Gorlova Olga Y, Navone Nora M, Maity Sankar N, Zhao Hongya, Sircar Kanishka, Gorlov Ivan P, Pettaway Curtis A, Byun Jin, and Logothetis Christopher J
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The genetic control of prostate cancer development is poorly understood. Large numbers of gene-expression datasets on different aspects of prostate tumorigenesis are available. We used these data to identify and prioritize candidate genes associated with the development of prostate cancer and bone metastases. Our working hypothesis was that combining meta-analyses on different but overlapping steps of prostate tumorigenesis will improve identification of genes associated with prostate cancer development. Methods A Z score-based meta-analysis of gene-expression data was used to identify candidate genes associated with prostate cancer development. To put together different datasets, we conducted a meta-analysis on 3 levels that follow the natural history of prostate cancer development. For experimental verification of candidates, we used in silico validation as well as in-house gene-expression data. Results Genes with experimental evidence of an association with prostate cancer development were overrepresented among our top candidates. The meta-analysis also identified a considerable number of novel candidate genes with no published evidence of a role in prostate cancer development. Functional annotation identified cytoskeleton, cell adhesion, extracellular matrix, and cell motility as the top functions associated with prostate cancer development. We identified 10 genes--CDC2, CCNA2, IGF1, EGR1, SRF, CTGF, CCL2, CAV1, SMAD4, and AURKA--that form hubs of the interaction network and therefore are likely to be primary drivers of prostate cancer development. Conclusions By using this large 3-level meta-analysis of the gene-expression data to identify candidate genes associated with prostate cancer development, we have generated a list of candidate genes that may be a useful resource for researchers studying the molecular mechanisms underlying prostate cancer development.
- Published
- 2010
- Full Text
- View/download PDF
16. Kinesin Facilitates Phenotypic Targeting of Therapeutic Resistance in Advanced Prostate Cancer.
- Author
-
Archer M, Begemann D, Gonzalez-Kozlova E, Nepali PR, Labanca E, Shepherd P, Dogra N, Navone N, and Kyprianou N
- Subjects
- Male, Humans, Animals, Mice, Cell Line, Tumor, Xenograft Model Antitumor Assays, Taxoids pharmacology, Taxoids therapeutic use, Epithelial-Mesenchymal Transition drug effects, Phenotype, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Prostatic Neoplasms metabolism, Drug Resistance, Neoplasm, Kinesins genetics, Kinesins metabolism, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Prostatic Neoplasms, Castration-Resistant metabolism
- Abstract
Understanding the mechanisms underlying resistance is critical to improving therapeutic outcomes in patients with metastatic castration-resistant prostate cancer. Previous work showed that dynamic interconversions between epithelial-mesenchymal transition to mesenchymal-epithelial transition defines the phenotypic landscape of prostate tumors, as a potential driver of the emergence of therapeutic resistance. In this study, we use in vitro and in vivo preclinical MDA PCa patient-derived xenograft models of resistant human prostate cancer to determine molecular mechanisms of cross-resistance between antiandrogen therapy and taxane chemotherapy, underlying the therapeutically resistant phenotype. Transcriptomic profiling revealed that resistant and sensitive prostate cancer C4-2B cells have a unique differential gene signature response to cabazitaxel. Gene pathway analysis showed that sensitive cells exhibit an increase in DNA damage, while resistant cells express genes associated with protein regulation in response to cabazitaxel. The patient-derived xenograft model specimens are from patients who have metastatic lethal castration-resistant prostate cancer, treated with androgen deprivation therapy, antiandrogens, and chemotherapy including second-line taxane chemotherapy, cabazitaxel. Immunohistochemistry revealed high expression of E-cadherin and low expression of vimentin resulting in redifferentiation toward an epithelial phenotype. Furthermore, the mitotic kinesin-related protein involved in microtubule binding and the SLCO1B3 transporter (implicated in cabazitaxel intracellular transport) are associated with resistance in these prostate tumors. Combinational targeting of kinesins (ispinesib) with cabazitaxel was more effective than single monotherapies in inducing cell death in resistant prostate tumors. Implications: Our findings are of translational significance in identifying kinesin as a novel target of cross-resistance toward enhancing therapeutic vulnerability and improved clinical outcomes in patients with advanced prostate cancer., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
17. Bone mimetic environments support engineering, propagation, and analysis of therapeutic response of patient-derived cells, ex vivo and in vivo.
- Author
-
Paindelli C, Parietti V, Barrios S, Shepherd P, Pan T, Wang WL, Satcher RL, Logothetis CJ, Navone N, Campbell MT, Mikos AG, and Dondossola E
- Subjects
- Male, Humans, Xenograft Model Antitumor Assays, Bone and Bones pathology, Osteoblasts pathology, Bone Neoplasms therapy, Bone Neoplasms secondary, Prostatic Neoplasms pathology
- Abstract
Bone metastases are the most common milestone in the lethal progression of prostate cancer and prominent in a substantial portion of renal malignancies. Interactions between cancer and bone host cells have emerged as drivers of both disease progression and therapeutic resistance. To best understand these central host-epithelial cell interactions, biologically relevant preclinical models are required. To achieve this goal, we here established and characterized tissue-engineered bone mimetic environments (BME) capable of supporting the growth of patient-derived xenograft (PDX) cells, ex vivo and in vivo. The BME consisted of a polycaprolactone (PCL) scaffold colonized by human mesenchymal stem cells (hMSCs) differentiated into osteoblasts. PDX-derived cells were isolated from bone metastatic prostate or renal tumors, engineered to express GFP or luciferase and seeded onto the BMEs. BMEs supported the growth and therapy response of PDX-derived cells, ex vivo. Additionally, BMEs survived after in vivo implantation and further sustained the growth of PDX-derived cells, their serial transplant, and their application to study the response to treatment. Taken together, this demonstrates the utility of BMEs in combination with patient-derived cells, both ex vivo and in vivo. STATEMENT OF SIGNIFICANCE: Our tissue-engineered BME supported the growth of patient-derived cells and proved useful to monitor the therapy response, both ex vivo and in vivo. This approach has the potential to enable co-clinical strategies to monitor bone metastatic tumor progression and therapy response, including identification and prioritization of new targets for patient treatment., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Eleonora Dondossola reports financial support was provided by The Prostate Cancer SPORE (P50 CA140388–07). Eleonora Dondossola reports financial support and equipment, drugs, or supplies were provided by Bayer HealthCare Pharmaceuticals (57440)., (Copyright © 2024 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Role of adenosine deaminase in prostate cancer progression.
- Author
-
Charles C, Lloyd SM, Piyarathna DWB, Gohlke J, Rasaily U, Putluri V, Simons BW, Zaslavsky A, Nallandhighal S, Michailidis G, Palanisamy N, Navone N, Jones JA, Ittmann MM, Putluri N, Rowley DR, Salami SS, Palapattu GS, and Sreekumar A
- Abstract
Prostate cancer (PCa) is the second most common cancer and constitutes about 14.7% of total cancer cases. PCa is highly prevalent and more aggressive in African-American (AA) men than in European-American (EA) men. PCa tends to be highly heterogeneous, and its complex biology is not fully understood. We use metabolomics to better understand the mechanisms behind PCa progression and disparities in its clinical outcome. Adenosine deaminase (ADA) is a key enzyme in the purine metabolic pathway; it was found to be upregulated in PCa and is associated with higher-grade PCa and poor disease-free survival. The inosine-to-adenosine ratio, which is a surrogate for ADA activity was high in PCa patient urine and higher in AA PCa compared to EA PCa. To understand the significance of high ADA in PCa, we established ADA overexpression models and performed various in vitro and in vivo studies. Our studies have revealed that an acute increase in ADA expression during later stages of tumor development enhances in vivo growth in multiple pre-clinical models. Further analysis revealed that mTOR signaling activation could be associated with this tumor growth. Chronic ADA overexpression shows alterations in the cells' adhesion machinery and a decrease in cells' ability to adhere to the extracellular matrix in vitro . Losing cell-matrix interaction is critical for metastatic dissemination which suggests that ADA could potentially be involved in promoting metastasis. This is supported by the association of higher ADA expression with higher-grade tumors and poor patient survival. Overall, our findings suggest that increased ADA expression may promote PCa progression, specifically tumor growth and metastatic dissemination., Competing Interests: None., (AJCEU Copyright © 2023.)
- Published
- 2023
19. Characterization of prostate cancer adrenal metastases: dependence upon androgen receptor signaling and steroid hormones.
- Author
-
Sakellakis MJ, Hahn AW, Ramachandran S, Zhang M, Hoang A, Song JH, Liu J, Wang F, Basu HS, Sheperd P, Wang X, Frigo DE, Lin SH, Panaretakis T, Zhang J, Navone N, Troncoso P, Logothetis CJ, and Titus MA
- Subjects
- Male, Humans, Animals, Mice, Receptors, Androgen genetics, Receptors, Androgen metabolism, Retrospective Studies, Steroids metabolism, Testosterone metabolism, DNA, Cell Line, Tumor, Tumor Microenvironment, Androgens metabolism, Prostatic Neoplasms pathology
- Abstract
Background: Prostate cancer (PCa) typically spreads to the bone, and this distribution is attributed to the central role of the microenvironment in progression. However, metastasis to the adrenal glands, while not as common, does occur. The biology that accounts for adrenal metastases may be attributed to the unique local steroid metabolome and co-clinical characterization may elucidate the role steroid biosynthesis plays in PCa progression., Methods: Three patients with metastatic PCa who had archived tumor tissue from an adrenalectomy were retrospectively identified, and one adrenal metastasis was developed into a xenograft (MDA-PCa-250). The adrenal metastases were characterized by performing somatic DNA whole exome sequencing (WES), RNA-Seq, immunohistochemistry (IHC), and steroid metabolite quantitation. The influence of steroid metabolites on adrenal metastasis cells and tumor growth was tested in vitro and in vivo., Results: Clinically, adrenalectomy was performed during castration-resistant oligometastatic disease, and two men experienced resensitization to leuprolide. Somatic DNA WES revealed heterogeneous alterations in tumor suppressor and DNA damage repair pathway genes. Adrenal metastases had active androgen receptor (AR) signaling by IHC, and RNA-Seq supported a potential role for adrenal androgen precursor metabolism in activating the AR. Steroid quantitation suggested the adrenal androgen precursors were converted into testosterone in these metastases, and stable isotope tracing of an organoid from MDA-PCa-250 confirmed the capability of adrenal metastases to biosynthesize testosterone from adrenal precursors. In vitro testing of a cell line derived from MDA-PCa-250 showed that testosterone and cortisol stimulated tumor cell growth. In vivo experiments demonstrated that MDA-PCa-250 grew in intact mice with circulating testosterone, but not in castrated mice., Conclusions: PCa adrenal metastases depend upon AR signaling driven by androgen precursors, androstenedione and dehydroepiandrosterone, available in the microenvironment, despite the presence of heterogeneous somatic DNA alterations. Moreover, MDA-PCa-250 provides a preclinical model that can recapitulate the unique androgen-dependence of adrenal metastases., Clinical Trial Registration: This study does not report the clinical results of a clinical trial, but it does use samples from a completed clinical trial that is registered with clinicaltrials.gov (NCT01254864)., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
20. Bone Progenitors Pull the Strings on the Early Metabolic Rewiring Occurring in Prostate Cancer Cells.
- Author
-
Sanchis P, Anselmino N, Lage-Vickers S, Sabater A, Lavignolle R, Labanca E, Shepherd PDA, Bizzotto J, Toro A, Mitrofanova A, Valacco MP, Navone N, Vazquez E, Cotignola J, and Gueron G
- Abstract
Metastatic prostate cancer (PCa) cells soiling in the bone require a metabolic adaptation. Here, we identified the metabolic genes fueling the seeding of PCa in the bone niche. Using a transwell co-culture system of PCa (PC3) and bone progenitor cells (MC3T3 or Raw264.7), we assessed the transcriptome of PC3 cells modulated by soluble factors released from bone precursors. In a Principal Component Analysis using transcriptomic data from human PCa samples (GSE74685), the altered metabolic genes found in vitro were able to stratify PCa patients in two defined groups: primary PCa and bone metastasis, confirmed by an unsupervised clustering analysis. Thus, the early transcriptional metabolic profile triggered in the in vitro model has a clinical correlate in human bone metastatic samples. Further, the expression levels of five metabolic genes ( VDR, PPARA, SLC16A1, GPX1 and PAPSS2 ) were independent risk-predictors of death in the SU2C-PCF dataset and a risk score model built using this lipid-associated signature was able to discriminate a subgroup of bone metastatic PCa patients with a 23-fold higher risk of death. This signature was validated in a PDX pre-clinical model when comparing MDA-PCa-183 growing intrafemorally vs. subcutaneously, and appears to be under the regulatory control of the Protein Kinase A (PKA) signaling pathway. Secretome analyses of conditioned media showcased fibronectin and type-1 collagen as critical bone-secreted factors that could regulate tumoral PKA. Overall, we identified a novel lipid gene signature, driving PCa aggressive metastatic disease pointing to PKA as a potential hub to halt progression.
- Published
- 2022
- Full Text
- View/download PDF
21. Exploiting Interdata Relationships in Prostate Cancer Proteomes: Clinical Significance of HO-1 Interactors.
- Author
-
Lage-Vickers S, Sanchis P, Bizzotto J, Toro A, Sabater A, Lavignolle R, Anselmino N, Labanca E, Paez A, Navone N, Valacco MP, Cotignola J, Vazquez E, and Gueron G
- Abstract
Prostate cancer (PCa) cells display abnormal expression of proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown the anti-tumoral role of heme oxygenase 1 (HO-1) in this disease. In this work, we undertook a mass spectrometry-based proteomics study to identify HO-1 molecular interactors that might collaborate with its modulatory function in PCa. Among the HO-1 interactors, we identified proteins with nuclear localization. Correlation analyses, using the PCa GSE70770 dataset, showed a significant and positive correlation between HMOX1 and 6 of those genes. Alternatively, HMOX1 and YWHAZ showed a negative correlation. Univariable analyses evidenced that high expression of HNRNPA2B1, HSPB1, NPM1, DDB1, HMGA1, ZC3HAV1, and HMOX1 was associated with increased relapse-free survival (RFS) in PCa patients. Further, PCa patients with high HSPB1/HMOX1 , DDB1/HMOX1 , and YWHAZ/HMOX1 showed a worse RFS compared with patients with lower ratios. Moreover, a decrease in RFS for patients with higher scores of this signature was observed using a prognostic risk score model. However, the only factor significantly associated with a higher risk of relapse was high YWHAZ . Multivariable analyses confirmed HSPB1 , DDB1 , and YWHAZ independence from PCa clinic-pathological parameters. In parallel, co-immunoprecipitation analysis in PCa cells ascertained HO-1/14-3-3ζ/δ (protein encoded by YWHAZ ) interaction. Herein, we describe a novel protein interaction between HO-1 and 14-3-3ζ/δ in PCa and highlight these factors as potential therapeutic targets.
- Published
- 2022
- Full Text
- View/download PDF
22. Prostate cancer castrate resistant progression usage of non-canonical androgen receptor signaling and ketone body fuel.
- Author
-
Labanca E, Bizzotto J, Sanchis P, Anselmino N, Yang J, Shepherd PDA, Paez A, Antico-Arciuch V, Lage-Vickers S, Hoang AG, Tang X, Raso MG, Titus M, Efstathiou E, Cotignola J, Araujo J, Logothetis C, Vazquez E, Navone N, and Gueron G
- Subjects
- Animals, Cell Line, Tumor, Disease Progression, Fatty Acids metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Mice, Neoplasm Transplantation, Prostatic Neoplasms, Castration-Resistant metabolism, Signal Transduction drug effects, Androgen Antagonists pharmacology, Ketone Bodies metabolism, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen metabolism
- Abstract
Prostate cancer (PCa) that progresses after androgen deprivation therapy (ADT) remains incurable. The underlying mechanisms that account for the ultimate emergence of resistance to ADT, progressing to castrate-resistant prostate cancer (CRPC), include those that reactivate androgen receptor (AR), or those that are entirely independent or cooperate with androgen signaling to underlie PCa progression. The intricacy of metabolic pathways associated with PCa progression spurred us to develop a metabolism-centric analysis to assess the metabolic shift occurring in PCa that progresses with low AR expression. We used PCa patient-derived xenografts (PDXs) to assess the metabolic changes after castration of tumor-bearing mice and subsequently confirmed main findings in human donor tumor that progressed after ADT. We found that relapsed tumors had a significant increase in fatty acids and ketone body (KB) content compared with baseline. We confirmed that critical ketolytic enzymes (ACAT1, OXCT1, BDH1) were dysregulated after castrate-resistant progression. Further, these enzymes are increased in the human donor tissue after progressing to ADT. In an in silico approach, increased ACAT1, OXCT1, BDH1 expression was also observed for a subset of PCa patients that relapsed with low AR and ERG (ETS-related gene) expression. Further, expression of these factors was also associated with decreased time to biochemical relapse and decreased progression-free survival. Our studies reveal the key metabolites fueling castration resistant progression in the context of a partial or complete loss of AR dependence., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
23. HO-1 Modulates Aerobic Glycolysis through LDH in Prostate Cancer Cells.
- Author
-
Cascardo F, Anselmino N, Páez A, Labanca E, Sanchis P, Antico-Arciuch V, Navone N, Gueron G, Vázquez E, and Cotignola J
- Abstract
Prostate cancer (PCa) is the second most diagnosed malignancy and the fifth leading cause of cancer associated death in men worldwide. Dysregulation of cellular energetics has become a hallmark of cancer, evidenced by numerous connections between signaling pathways that include oncoproteins and key metabolic enzymes. We previously showed that heme oxygenase 1 (HO-1), a cellular homeostatic regulator counteracting oxidative and inflammatory damage, exhibits anti-tumoral activity in PCa cells, inhibiting cell proliferation, migration, tumor growth and angiogenesis. The aim of this study was to assess the role of HO-1 on the metabolic signature of PCa. After HO-1 pharmacological induction with hemin, PC3 and C4-2B cells exhibited a significantly impaired cellular metabolic rate, reflected by glucose uptake, ATP production, lactate dehydrogenase (LDH) activity and extracellular lactate levels. Further, we undertook a bioinformatics approach to assess the clinical significance of LDHA , LDHB and HMOX1 in PCa, identifying that high LDHA or low LDHB expression was associated with reduced relapse free survival (RFS). Interestingly, the shortest RFS was observed for PCa patients with low HMOX1 and high LDHA, while an improved prognosis was observed for those with high HMOX1 and LDHB . Thus, HO-1 induction causes a shift in the cellular metabolic profile of PCa, leading to a less aggressive phenotype of the disease.
- Published
- 2021
- Full Text
- View/download PDF
24. Optimizing the diagnosis and management of ductal prostate cancer.
- Author
-
Ranasinghe W, Shapiro DD, Zhang M, Bathala T, Navone N, Thompson TC, Broom B, Aparicio A, Tu SM, Tang C, Davis JW, Pisters L, and Chapin BF
- Subjects
- Humans, Male, Carcinoma, Ductal diagnosis, Carcinoma, Ductal therapy, Prostatic Neoplasms diagnosis, Prostatic Neoplasms therapy
- Abstract
Ductal adenocarcinoma (DAC) is the most common variant histological subtype of prostate carcinoma and has an aggressive clinical course. DAC is usually characterized and treated as high-risk prostatic acinar adenocarcinoma (PAC). However, DAC has a different biology to that of acinar disease, which often poses a challenge for both diagnosis and management. DAC can be difficult to identify using conventional diagnostic modalities such as serum PSA levels and multiparametric MRI, and the optimal management for localized DAC is unknown owing to the rarity of the disease. Following definitive therapy for localized disease with radical prostatectomy or radiotherapy, the majority of DACs recur with visceral metastases at low PSA levels. Various systemic therapies that have been shown to be effective in high-risk PAC have limited use in treating DAC. Although current understanding of the biology of DAC is limited, genomic analyses have provided insights into the pathology behind its aggressive behaviour and potential future therapeutic targets.
- Published
- 2021
- Full Text
- View/download PDF
25. Radium-223 Treatment Increases Immune Checkpoint Expression in Extracellular Vesicles from the Metastatic Prostate Cancer Bone Microenvironment.
- Author
-
Vardaki I, Corn P, Gentile E, Song JH, Madan N, Hoang A, Parikh N, Guerra L, Lee YC, Lin SC, Yu G, Santos E, Melancon MP, Troncoso P, Navone N, Gallick GE, Efstathiou E, Subudhi SK, Lin SH, Logothetis CJ, and Panaretakis T
- Subjects
- Animals, Bone Neoplasms mortality, Bone Neoplasms pathology, Cell Line, Tumor, Exosomes genetics, Gene Expression Profiling, Humans, Male, Mice, Prostatic Neoplasms mortality, RNA genetics, Survival Rate, Bone Neoplasms secondary, Extracellular Vesicles metabolism, Gene Expression drug effects, Gene Expression genetics, Immune Checkpoint Proteins genetics, Immune Checkpoint Proteins metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Radiopharmaceuticals pharmacology, Radiopharmaceuticals therapeutic use, Radium pharmacology, Radium therapeutic use, Tumor Microenvironment drug effects, Tumor Microenvironment genetics
- Abstract
Purpose: Radium-223 prolongs survival in a fraction of men with bone metastatic prostate cancer (PCa). However, there are no markers for monitoring response and resistance to Radium-223 treatment. Exosomes are mediators of intercellular communication and may reflect response of the bone microenvironment to Radium-223 treatment. We performed molecular profiling of exosomes and compared the molecular profile in patients with favorable and unfavorable overall survival., Experimental Design: We performed exosomal transcriptome analysis in plasma derived from our preclinical models (MDA-PCa 118b tumors, TRAMP-C2/BMP4 PCa) and from the plasma of 25 patients (paired baseline and end of treatment) treated with Radium-223. All samples were run in duplicate, and array data analyzed with fold changes +2 to -2 and P < 0.05., Results: We utilized the preclinical models to establish that genes derived from the tumor and the tumor-associated bone microenvironment (bTME) are differentially enriched in plasma exosomes upon Radium-223 treatment. The mouse transcriptome analysis revealed changes in bone-related and DNA damage repair-related pathways. Similar findings were observed in plasma-derived exosomes from patients treated with Radium-223 detected changes. In addition, exosomal transcripts detected immune-suppressors (e.g., PD-L1) that were associated with shorter survival to Radium-223. Treatment of the Myc-CaP mouse model with a combination of Radium-223 and immune checkpoint therapy (ICT) resulted in greater efficacy than monotherapy., Conclusions: These clinical and coclinical analyses showed that RNA profiling of plasma exosomes may be used for monitoring the bTME in response to treatment and that ICT may be used to increase the efficacy of Radium-223., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
26. Ductal Prostate Cancers Demonstrate Poor Outcomes with Conventional Therapies.
- Author
-
Ranasinghe W, Shapiro DD, Hwang H, Wang X, Reichard CA, Elsheshtawi M, Achim MF, Bathala T, Tang C, Aparicio A, Tu SM, Navone N, Thompson TC, Pisters L, Troncoso P, Davis JW, and Chapin BF
- Subjects
- Aged, Humans, Male, Middle Aged, Retrospective Studies, Risk Assessment, Treatment Outcome, Adenocarcinoma therapy, Prostatic Neoplasms therapy
- Abstract
Background: Ductal prostate adenocarcinoma (DAC) is a rare, aggressive, histologic variant of prostate cancer that is treated with conventional therapies, similar to high-risk prostate adenocarcinoma (PAC)., Objective: To assess the outcomes of men undergoing definitive therapy for DAC or high-risk PAC and to explore the effects of androgen deprivation therapy (ADT) in improving the outcomes of DAC., Design, Setting, and Participants: A single-center retrospective review of all patients with cT1-4/N0-1 DAC from 2005 to 2018 was performed. Those undergoing radical prostatectomy (RP) or radiotherapy (RTx) for DAC were compared with cohorts of high-risk PAC patients., Outcome Measurements and Statistical Analysis: Metastasis-free survival (MFS) and overall survival (OS) rates were analyzed using Kaplan-Meier and Cox regression models., Results and Limitations: A total of 228 men with DAC were identified; 163 underwent RP, 34 underwent RTx, and 31 had neoadjuvant therapy prior to RP. In this study, 163 DAC patients and 155 PAC patients undergoing RP were compared. Similarly, 34 DAC patients and 74 PAC patients undergoing RTx were compared. DAC patients undergoing RP or RTx had worse 5-yr MFS (75% vs 95% and 62% vs 93%, respectively, p < 0.001) and 5-yr OS (88% vs 97% and 82% vs 100%, respectively, p < 0.05) compared with PAC patients. In the 76 men who received adjuvant/salvage ADT after RP, DAC also had worse MFS and OS than PAC (p < 0.01). A genomic analysis revealed that 10/11 (91%) DACs treated with ADT had intrinsic upregulation of androgen-resistant pathways. Further, none of the DAC patients (0/15) who received only neoadjuvant ADT prior to RP had any pathologic downgrading. The retrospective nature was a limitation., Conclusions: Men undergoing RP or RTx for DAC had worse outcomes than PAC patients, regardless of the treatment modality. Upregulation of several intrinsic resistance pathways in DAC rendered ADT less effective. Further evaluation of the underlying biology of DAC with clinical trials is needed., Patient Summary: This study demonstrated worse outcomes among patients with ductal adenocarcinoma of the prostate than among high-grade prostate adenocarcinoma patients, regardless of the treatment modality., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
27. The expression of YWHAZ and NDRG1 predicts aggressive outcome in human prostate cancer.
- Author
-
Lage-Vickers S, Bizzotto J, Valacco MP, Sanchis P, Nemirovsky S, Labanca E, Scorticati C, Mazza O, Mitrofanova A, Navone N, Vazquez E, Cotignola J, and Gueron G
- Subjects
- Adenocarcinoma mortality, Adult, Aged, Biomarkers, Tumor metabolism, Humans, Male, Mass Spectrometry, Middle Aged, Prostatic Neoplasms mortality, Proteome, Risk Assessment, 14-3-3 Proteins metabolism, Adenocarcinoma metabolism, Cell Cycle Proteins metabolism, Intracellular Signaling Peptides and Proteins metabolism, Prostatic Hyperplasia metabolism, Prostatic Neoplasms metabolism
- Abstract
Some prostate cancers (PCas) are histo-pathologically grouped within the same Gleason Grade (GG), but can differ significantly in outcome. Herein, we aimed at identifying molecular biomarkers that could improve risk prediction in PCa. LC ESI-MS/MS was performed on human PCa and benign prostatic hyperplasia (BPH) tissues and peptide data was integrated with omic analyses. We identified high YWHAZ and NDRG1 expression to be associated with poor PCa prognosis considering all Gleason scores (GS). YWHAZ and NDRG1 defined two subpopulations of PCa patients with high and intermediate risk of death. Multivariable analyses confirmed their independence from GS. ROC analysis unveiled that YWHAZ outperformed GS beyond 60 months post-diagnosis. The genomic analysis of PCa patients with YWHAZ amplification, or increased mRNA or protein levels, revealed significant alterations in key DNA repair genes. We hereby state the relevance of YWHAZ in PCa, showcasing its role as an independent strong predictor of aggressiveness.
- Published
- 2021
- Full Text
- View/download PDF
28. Enhanced Viability for Ex vivo 3D Hydrogel Cultures of Patient-Derived Xenografts in a Perfused Microfluidic Platform.
- Author
-
Sablatura LK, Bircsak KM, Shepherd P, Queiroz K, Farach-Carson MC, Constantinou PE, Saleh A, Navone N, and Harrington DA
- Subjects
- Animals, Cell Survival, Centrifugation, Density Gradient, Disease Models, Animal, Humans, Image Processing, Computer-Assisted, Mice, Staining and Labeling, Cell Culture Techniques, Heterografts, Hydrogels chemistry, Microfluidics
- Abstract
Patient-derived xenografts (PDX), generated when resected patient tumor tissue is engrafted directly into immunocompromised mice, remain biologically stable, thereby preserving molecular, genetic, and histological features, as well as heterogeneity of the original tumor. However, using these models to perform a multitude of experiments, including drug screening, is prohibitive both in terms of cost and time. Three-dimensional (3D) culture systems are widely viewed as platforms in which cancer cells retain their biological integrity through biochemical interactions, morphology, and architecture. Our team has extensive experience culturing PDX cells in vitro using 3D matrices composed of hyaluronic acid (HA). In order to separate mouse fibroblast stromal cells associated with PDXs, we use rotation culture, where stromal cells adhere to the surface of tissue culture-treated plates while dissociated PDX tumor cells float and self-associate into multicellular clusters. Also floating in the supernatant are single, often dead cells, which present a challenge in collecting viable PDX clusters for downstream encapsulation into hydrogels for 3D cell culture. In order to separate these single cells from live cell clusters, we have employed density step gradient centrifugation. The protocol described here allows for the depletion of non-viable single cells from the healthy population of cell clusters that will be used for further in vitro experimentation. In our studies, we incorporate the 3D cultures in microfluidic plates which allow for media perfusion during culture. After assessing the resultant cultures using a fluorescent image-based viability assay of purified versus non-purified cells, our results show that this additional separation step substantially reduced the number of non-viable cells from our cultures.
- Published
- 2020
- Full Text
- View/download PDF
29. Myxovirus Resistance Protein 1 (MX1), a Novel HO-1 Interactor, Tilts the Balance of Endoplasmic Reticulum Stress towards Pro-Death Events in Prostate Cancer.
- Author
-
Ortiz E, Sanchis P, Bizzotto J, Lage-Vickers S, Labanca E, Navone N, Cotignola J, Vazquez E, and Gueron G
- Subjects
- Case-Control Studies, Cell Proliferation, Data Mining, Databases, Genetic, Endoplasmic Reticulum Stress, Gene Expression Regulation, Neoplastic, Heme Oxygenase-1 genetics, Humans, Male, Myxovirus Resistance Proteins genetics, PC-3 Cells, Prostatic Neoplasms genetics, Survival Analysis, Tumor Microenvironment, Computational Biology methods, Heme Oxygenase-1 metabolism, Myxovirus Resistance Proteins metabolism, Prostatic Neoplasms metabolism
- Abstract
The inflammatory tumor microenvironment is a fertile niche accelerating prostate cancer (PCa). We have reported that heme-oxygenase (HO-1) had a strong anti-tumoral effect in PCa. We previously undertook an in-depth proteomics study to build the HO-1 interactome in PCa. In this work, we used a bioinformatics approach to address the biological significance of HO-1 interactors. Open-access PCa datasets were mined to address the clinical significance of the HO-1 interactome in human samples. HO-1 interactors were clustered into groups according to their expression profile in PCa patients. We focused on the myxovirus resistance gene ( MX1 ) as: (1) it was significantly upregulated under HO-1 induction; (2) it was the most consistently downregulated gene in PCa vs. normal prostate; (3) its loss was associated with decreased relapse-free survival in PCa; and (4) there was a significant positive correlation between MX1 and HMOX1 in PCa patients. Further, MX1 was upregulated in response to endoplasmic reticulum stress (ERS), and this stress triggered apoptosis and autophagy in PCa cells. Strikingly, MX1 silencing reversed ERS. Altogether, we showcase MX1 as a novel HO-1 interactor and downstream target, associated with ERS in PCa and having a high impact in the clinical setting.
- Published
- 2020
- Full Text
- View/download PDF
30. Heme Oxygenase-1 Is a Pivotal Modulator of Bone Turnover and Remodeling: Molecular Implications for Prostate Cancer Bone Metastasis.
- Author
-
Anselmino N, Starbuck M, Labanca E, Cotignola J, Navone N, Gueron G, Zenclussen AC, and Vazquez E
- Subjects
- Animals, Bone Neoplasms secondary, Bone Regeneration, Bone Remodeling, Heme Oxygenase-1 deficiency, Male, Membrane Proteins deficiency, Mice, Mice, Inbred BALB C, Mice, Knockout, Bone Neoplasms metabolism, Heme Oxygenase-1 metabolism, Membrane Proteins metabolism, Prostatic Neoplasms metabolism
- Abstract
Aims: Bone is the most frequent site of prostate cancer (PCa) metastasis. Tumor cells interact with the bone microenvironment interrupting tissue balance. Heme oxygenase-1 (HO-1; encoded by Hmox1 ) appears as a potential target in PCa maintaining the cellular homeostasis. Our hypothesis is that HO-1 is implicated in bone physiology and modulates the communication with PCa cells. Here we aimed at (i) assessing the physiological impact of Hmox1 gene knockout (KO) on bone metabolism in vivo and (ii) determining the alterations of the transcriptional landscape associated with tumorigenesis and bone remodeling in cells growing in coculture (PCa cells with primary mouse osteoblasts [PMOs] from BALB/c Hmox1
+/+ , Hmox1+/- , and Hmox1-/- mice). Results: Histomorphometric analysis of Hmox1-/- mice bones exhibited significantly decreased bone density with reduced remodeling parameters. A positive correlation between Hmox1 expression and Runx2 , Col1a1 , Csf1 , and Opg genes was observed in PMOs. Flow cytometry studies revealed two populations of PMOs with different reactive oxygen species (ROS) levels. The high ROS population was increased in PMOs Hmox1+/- compared with Hmox1+/+ , but was significantly reduced in PMOs Hmox1-/- , suggesting restrained ROS tolerance in KO cells. Gene expression was altered in PMOs upon coculture with PCa cells, showing a pro-osteoclastic profile. Moreover, HO-1 induction in PCa cells growing in coculture with PMOs resulted in a significant modulation of key bone markers such as PTHrP and OPG . Innovation and Conclusion: We here demonstrate the direct implications of HO-1 expression in bone remodeling and how it participates in the alterations in the communication between bone and prostate tumor cells.- Published
- 2020
- Full Text
- View/download PDF
31. Favorable incremental cost-effectiveness ratio for lung cancer screening in Italy.
- Author
-
Veronesi G, Navone N, Novellis P, Dieci E, Toschi L, Velutti L, Solinas M, Vanni E, Alloisio M, and Ghislandi S
- Subjects
- Aged, Aged, 80 and over, Combined Modality Therapy, Female, Follow-Up Studies, Humans, Italy epidemiology, Lung Neoplasms diagnosis, Lung Neoplasms epidemiology, Lung Neoplasms therapy, Male, Middle Aged, Prognosis, Survival Rate, Cost-Benefit Analysis, Early Detection of Cancer economics, Health Care Costs statistics & numerical data, Lung Neoplasms economics, Quality-Adjusted Life Years
- Abstract
Objectives: Lung cancer detection by low-dose computed tomographic screening reduces mortality. However, it is essential to assess cost-effectiveness. We present a cost-effectiveness analysis of screening in Italians at high risk of lung cancer, from the point of view of the Italian tax-payer., Materials and Methods: We used a decision model to estimate the cost-effectiveness of annual screening for 5 years in smokers (≥30 pack-years) of 55-79 years. Patients diagnosed in the COSMOS study were the screening arm; patients diagnosed and treated for lung cancer in the Lombardy Region, Italy, constituted the usual care arm. Treatment costs were extracted from our hospital database. Lung cancer survival in screened patients was adjusted for 2-year lead-time bias. Life-years and quality-adjusted life-years were estimated by stage at diagnosis, from which incremental cost-effectiveness ratios per life-year and quality-adjusted life-year gained were estimated., Results: Base-case incremental cost-effectiveness ratios were 3297 and 2944 euro per quality-adjusted life-year and life-year gained, respectively. Deterministic sensitivity analysis indicated that these values were particularly sensitive to lung cancer prevalence, screening sensitivity and specificity, screening cost, and treatment costs for stage I and IV disease. From the probabilistic sensitivity analysis incremental cost-effectiveness ratios had a 98 % probability of being <25,000 euro (widely-accepted threshold) and a 55 % probability of being <5000 euro., Conclusions: Low-dose computed tomographic screening is associated with an incremental cost of 2944 euro per life-year gained in high risk population, implying that screening can be introduced in Italy at contained cost, saving the lives of many lung cancer patients., Competing Interests: Declaration of Competing Interest GV has financial relationships with AB Medica SpA, Johnson & Johnson Medical SpA, and Medtronic Italia SpA, unrelated to the content of this study. NN received a research grant from the Italian National Insurance Institute for Workplace Injuries (INAIL). None of the other authors has any conflict of interest to declare., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
32. HO-1 Interactors Involved in the Colonization of the Bone Niche: Role of ANXA2 in Prostate Cancer Progression.
- Author
-
Anselmino N, Bizzotto J, Sanchis P, Lage-Vickers S, Ortiz E, Valacco P, Paez A, Labanca E, Meiss R, Navone N, Cotignola J, Vazquez E, and Gueron G
- Subjects
- Animals, Bone Neoplasms pathology, Bone and Bones metabolism, Bone and Bones pathology, Humans, Male, Mice, Neoplasm Metastasis, PC-3 Cells, Prostatic Neoplasms pathology, RAW 264.7 Cells, Annexin A2 metabolism, Bone Neoplasms metabolism, Bone Neoplasms secondary, Heme Oxygenase-1 metabolism, Neoplasm Proteins metabolism, Prostatic Neoplasms metabolism, Tumor Microenvironment
- Abstract
Background: Prostate cancer (PCa) dissemination shows a tendency to develop in the bone, where heme oxygenase 1 (HO-1) plays a critical role in bone remodeling. Previously by LC/ESI-MSMS, we screened for HO-1 interacting proteins and identified annexin 2 (ANXA2). The aim of this study was to analyze the relevance of ANXA2/HO-1 in PCa and bone metastasis., Methods: We assessed ANXA2 levels using a co-culture transwell system of PC3 cells (pre-treated or not with hemin, an HO-1 specific inducer) and the pre-osteoclastic Raw264.7 cell line., Results: Under co-culture conditions, ANXA2 mRNA levels were significantly modulated in both cell lines. Immunofluorescence analysis unveiled a clear ANXA2 reduction in cell membrane immunostaining for Raw264.7 under the same conditions. This effect was supported by the detection of a decrease in Ca
2+ concentration in the conditioned medium. HO-1 induction in tumor cells prevented both, the ANXA2 intracellular relocation and the decrease in Ca2+ concentration. Further, secretome analysis revealed urokinase (uPA) as a key player in the communication between osteoclast progenitors and PC3 cells. To assess the clinical significance of ANXA2/HO-1, we performed a bioinformatics analysis and identified that low expression of each gene strongly associated with poor prognosis in PCa regardless of the clinico-pathological parameters assessed. Further, these genes appear to behave in a dependent manner., Conclusions: ANXA2/HO-1 rises as a critical axis in PCa., Competing Interests: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.- Published
- 2020
- Full Text
- View/download PDF
33. Oncogenic and osteolytic functions of histone demethylase NO66 in castration-resistant prostate cancer.
- Author
-
Sinha KM, Bagheri-Yarmand R, Lahiri S, Lu Y, Zhang M, Amra S, Rizvi Y, Wan X, Navone N, Ozpolat B, Logothetis C, Gagel RF, and Huard J
- Subjects
- Animals, Bone Neoplasms genetics, Bone Neoplasms metabolism, Bone Neoplasms secondary, Cell Line, Tumor, Cell Transformation, Neoplastic metabolism, Dioxygenases genetics, Epigenesis, Genetic genetics, Gene Expression Regulation, Neoplastic, HEK293 Cells, Histone Demethylases genetics, Histones metabolism, Humans, Male, Mice, Mice, SCID, NIH 3T3 Cells, Osteolysis pathology, PC-3 Cells, Prostatic Neoplasms, Castration-Resistant metabolism, Cell Transformation, Neoplastic genetics, Dioxygenases physiology, Histone Demethylases physiology, Osteolysis genetics, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology
- Abstract
Epigenetic changes that cause dysregulated gene expression during progression of androgen-independent prostate cancer (PCa) and metastatic skeletal lesions remain elusive. Here, we explored the role of histone demethylase NO66 in the pathogenesis of PCa and bone metastasis-related skeletal lesions. Tissue and cDNA microarrays of PCa were analyzed for NO66 mRNA and protein levels. We examined the effects of gain and loss of NO66 function on cell viability, colony formation, migration, invasion, and tumor-induced skeletal lesions in femoral bone. RNAseq and ChIPseq were performed to elucidate NO66-target genes in PCa. We report that NO66 levels were upregulated in advanced primary prostate tumors compared to normal tissue or tumors with low Gleason scores. Forced expression of NO66 promoted cell survival and invasion of PCa cells; whereas, knockdown of NO66 resulted in decreased cell survival and increased sensitivity to docetaxel. NO66-overexpressing PC3 cells implanted into the femoral bone of male SCID mice caused massive bone loss and stimulation of mouse osteoclast-promoting genes, including Dickkopf1, Cathepsin K, Nf-kβ,; and Calcr, suggesting a role for NO66 in tumor growth in bone and osteoclast activity. Combined RNAseq and ChIP-seq revealed that NO66 activates the survival gene MCL1, the invasion-associated genes IGFBP5 and MMP3, the pro-oncogenic genes CTNNB1 and CCND1, and the epigenetic modifier gene KMT2A in androgen-independent PCa. Our findings uncover the role of NO66 as a key oncogenic driver in PCa, causing osteolytic lesions through upstream epigenetic regulation of key genes for survival, invasion and metastasis, and pro-osteoclastic factors.
- Published
- 2019
- Full Text
- View/download PDF
34. ERR1 and PGC1α associated mitochondrial alterations correlate with pan-cancer disparity in African Americans.
- Author
-
Piyarathna DWB, Balasubramanian A, Arnold JM, Lloyd SM, Karanam B, Castro P, Ittmann MM, Putluri N, Navone N, Jones JA, Yu W, Sandulache VC, Sikora AG, Michailidis G, and Sreekumar A
- Subjects
- Female, Humans, Male, Mitochondria metabolism, Mitochondria pathology, Neoplasm Proteins metabolism, Neoplasms metabolism, Neoplasms pathology, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Receptors, Estrogen metabolism, Transcription, Genetic, White People genetics, ERRalpha Estrogen-Related Receptor, Black or African American genetics, Databases, Nucleic Acid, Mitochondria genetics, Neoplasm Proteins genetics, Neoplasms genetics, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha genetics, Receptors, Estrogen genetics
- Abstract
Background: African American (AA) patients have higher cancer mortality rates and shorter survival times compared to European American (EA) patients. Despite a significant focus on socioeconomic factors, recent findings strongly argue the existence of biological factors driving this disparity. Most of these factors have been described in a cancer-type specific context rather than a pan-cancer setting., Methods: A novel in silico approach based on Gene Set Enrichment Analysis (GSEA) coupled to Transcription Factor enrichment was carried out to identify common biological drivers of pan-cancer racial disparity using The Cancer Genome Atlas (TCGA) dataset. Mitochondrial content in patient tissues was examined using a multi-cancer tissue microarray approach (TMA)., Results: Mitochondrial oxidative phosphorylation was uniquely enriched in AA tumors compared to EA tumors across various cancer types. AA tumors also showed strong enrichment for the ERR1-PGC1α-mediated transcriptional program, which has been implicated in mitochondrial biogenesis. TMA analysis revealed that AA cancers harbor significantly more mitochondria compared to their EA counterparts., Conclusions: These findings highlight changes in mitochondria as a common distinguishing feature between AA and EA tumors in a pan-cancer setting, and provide the rationale for the repurposing of mitochondrial inhibitors to treat AA cancers.
- Published
- 2019
- Full Text
- View/download PDF
35. Engineered bone for probing organotypic growth and therapy response of prostate cancer tumoroids in vitro.
- Author
-
Paindelli C, Navone N, Logothetis CJ, Friedl P, and Dondossola E
- Subjects
- Animals, Bone Neoplasms drug therapy, Bone Neoplasms secondary, Cell Culture Techniques methods, Cell Proliferation drug effects, Docetaxel pharmacology, Humans, Male, Mice, SCID, PC-3 Cells, Prostatic Neoplasms pathology, Radium pharmacology, Spheroids, Cellular drug effects, Spheroids, Cellular pathology, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured pathology, Tumor Microenvironment drug effects, Antineoplastic Agents pharmacology, Drug Screening Assays, Antitumor methods, Prostatic Neoplasms drug therapy
- Abstract
Mechanistic analysis of metastatic prostate cancer (PCa) biology and therapy response critically depends upon clinically relevant three-dimensional (3D) bone-like, organotypic culture. We here combine an engineered bone-mimetic environment (BME) with longitudinal microscopy to test the growth and therapy response of 3D PCa tumoroids. Besides promoting both tumor-cell autonomous and microenvironment-dependent growth in PCa cell lines and patient-derived xenograft cells, the BME enables in vivo-like tumor cell response to therapy, and reveals bone stroma dependent resistance to chemotherapy and BME-targeted localization and induction of cytoxicity by Radium-223. The BME platform will allow the propagation, compound screening and mechanistic dissection of patient-derived bone tumor isolates and applications toward personalized medicine., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
36. The estrogen receptor variants β2 and β5 induce stem cell characteristics and chemotherapy resistance in prostate cancer through activation of hypoxic signaling.
- Author
-
Faria M, Shepherd P, Pan Y, Chatterjee SS, Navone N, Gustafsson JÅ, and Strom A
- Abstract
Chemotherapy resistant prostate cancer is a major clinical problem. When the prostate cancer has become androgen deprivation resistant, one of the few treatment regimens left is chemotherapy. There is a strong connection between a cancer's stem cell like characteristics and drug resistance. By performing RNA-seq we observed several factors associated with stem cells being strongly up-regulated by the estrogen receptor β variants, β2 and β5. In addition, most of these factors were also up-regulated by hypoxia. One mechanism of chemotherapy resistance was expression of the hypoxia-regulated, drug transporter genes, where especially ABCG2 and MDR1 were shown to be expressed in recurrent prostate cancer and to cause chemotherapy resistance by efficiently transporting drugs like docetaxel out of the cells. Another mechanism was expression of the hypoxia-regulated Notch3 gene, which causes chemotherapy resistance in urothelial carcinoma, although the mechanism is unknown. It is well known that hypoxic signaling is involved in increasing chemotherapy resistance. Regulation of the hypoxic factors, HIF-1α and HIF-2α is very complex and extends far beyond hypoxia itself. We have recently shown that two of the estrogen receptor β variants, estrogen receptor β2 and β5, bind to and stabilize both HIF-1α and HIF-2α proteins leading to expression of HIF target genes. This study suggests that increased expression of the estrogen receptor β variants, β2 and β5, could be involved in development of a cancer's stem cell characteristics and chemotherapy resistance, indicating that targeting these factors could prevent or reverse chemotherapy resistance and cancer stem cell expansion., Competing Interests: CONFLICTS OF INTEREST The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the work reported.
- Published
- 2018
- Full Text
- View/download PDF
37. Intravital microscopy of osteolytic progression and therapy response of cancer lesions in the bone.
- Author
-
Dondossola E, Alexander S, Holzapfel BM, Filippini S, Starbuck MW, Hoffman RM, Navone N, De-Juan-Pardo EM, Logothetis CJ, Hutmacher DW, and Friedl P
- Subjects
- Animals, Bone Marrow blood supply, Bone Marrow pathology, Bone Neoplasms drug therapy, Bone Neoplasms pathology, Cathepsin K metabolism, Cell Line, Tumor, Diphosphonates pharmacology, Diphosphonates therapeutic use, Female, Humans, Male, Mice, Mice, Nude, Miniaturization, Stromal Cells pathology, Tissue Engineering, Tissue Scaffolds chemistry, Treatment Outcome, Zoledronic Acid pharmacology, Zoledronic Acid therapeutic use, Bone Neoplasms therapy, Disease Progression, Intravital Microscopy methods, Osteolysis pathology
- Abstract
Intravital multiphoton microscopy (iMPM) in mice provides access to cellular and molecular mechanisms of metastatic progression of cancers and the underlying interactions with the tumor stroma. Whereas iMPM of malignant disease has been performed for soft tissues, noninvasive iMPM of solid tumor in the bone is lacking. We combined miniaturized tissue-engineered bone constructs in nude mice with a skin window to noninvasively and repetitively monitor prostate cancer lesions by three-dimensional iMPM. In vivo ossicles developed large central cavities containing mature bone marrow surrounded by a thin cortex and enabled tumor implantation and longitudinal iMPM over weeks. Tumors grew inside the bone cavity and along the cortical bone interface and induced niches of osteoclast activation (focal osteolysis). Interventional bisphosphonate therapy reduced osteoclast kinetics and osteolysis without perturbing tumor growth, indicating dissociation of the tumor-stroma axis. The ossicle window, with its high cavity-to-cortex ratio and long-term functionality, thus allows for the mechanistic dissection of reciprocal epithelial tumor-bone interactions and therapy response., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
38. Game-changing restraint of Ros-damaged phenylalanine, upon tumor metastasis.
- Author
-
Gueron G, Anselmino N, Chiarella P, Ortiz EG, Lage Vickers S, Paez AV, Giudice J, Contin MD, Leonardi D, Jaworski F, Manzano V, Strazza A, Montagna DR, Labanca E, Cotignola J, D Accorso N, Woloszynska-Read A, Navone N, Meiss RP, Ruggiero R, and Vazquez E
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Down-Regulation, Drug Resistance, Neoplasm, Humans, Male, Mice, Nude, Prostatic Neoplasms pathology, Serum, Signal Transduction, Subcutaneous Tissue pathology, Tyrosine metabolism, Xenograft Model Antitumor Assays, Neoplasm Metastasis pathology, Phenylalanine metabolism, Reactive Oxygen Species metabolism
- Abstract
An abrupt increase in metastatic growth as a consequence of the removal of primary tumors suggests that the concomitant resistance (CR) phenomenon might occur in human cancer. CR occurs in murine tumors and ROS-damaged phenylalanine, meta-tyrosine (m-Tyr), was proposed as the serum anti-tumor factor primarily responsible for CR. Herein, we demonstrate for the first time that CR happens in different experimental human solid tumors (prostate, lung anaplastic, and nasopharyngeal carcinoma). Moreover, m-Tyr was detected in the serum of mice bearing prostate cancer (PCa) xenografts. Primary tumor growth was inhibited in animals injected with m-Tyr. Further, the CR phenomenon was reversed when secondary implants were injected into mice with phenylalanine (Phe), a protective amino acid highly present in primary tumors. PCa cells exposed to m-Tyr in vitro showed reduced cell viability, downregulated NFκB/STAT3/Notch axis, and induced autophagy; effects reversed by Phe. Strikingly, m-Tyr administration also impaired both, spontaneous metastasis derived from murine mammary carcinomas (4T1, C7HI, and LMM3) and PCa experimental metastases. Altogether, our findings propose m-Tyr delivery as a novel approach to boost the therapeutic efficacy of the current treatment for metastasis preventing the escape from tumor dormancy.
- Published
- 2018
- Full Text
- View/download PDF
39. Targeting the MYCN-PARP-DNA Damage Response Pathway in Neuroendocrine Prostate Cancer.
- Author
-
Zhang W, Liu B, Wu W, Li L, Broom BM, Basourakos SP, Korentzelos D, Luan Y, Wang J, Yang G, Park S, Azad AK, Cao X, Kim J, Corn PG, Logothetis CJ, Aparicio AM, Chinnaiyan AM, Navone N, Troncoso P, and Thompson TC
- Subjects
- Animals, Aurora Kinase A metabolism, Carcinoma, Neuroendocrine drug therapy, Carcinoma, Neuroendocrine metabolism, Carcinoma, Neuroendocrine pathology, Cell Line, Tumor, Computational Biology methods, Disease Models, Animal, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Heterografts, Humans, Male, Mice, Mitosis genetics, N-Myc Proto-Oncogene Protein metabolism, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerases metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Transcriptome, Carcinoma, Neuroendocrine genetics, DNA Damage, N-Myc Proto-Oncogene Protein genetics, Poly(ADP-ribose) Polymerases genetics, Prostatic Neoplasms genetics, Signal Transduction
- Abstract
Purpose: We investigated MYCN-regulated molecular pathways in castration-resistant prostate cancer (CRPC) classified by morphologic criteria as adenocarcinoma or neuroendocrine to extend the molecular phenotype, establish driver pathways, and identify novel approaches to combination therapy for neuroendocrine prostate cancer (NEPC). Experimental Design and Results: Using comparative bioinformatics analyses of CRPC-Adeno and CRPC-Neuro RNA sequence data from public data sets and a panel of 28 PDX models, we identified a MYCN-PARP-DNA damage response (DDR) pathway that is enriched in CRPC with neuroendocrine differentiation (NED) and CRPC-Neuro. ChIP-PCR assay revealed that N-MYC transcriptionally activates PARP1, PARP2, BRCA1, RMI2, and TOPBP1 through binding to the promoters of these genes. MYCN or PARP1 gene knockdown significantly reduced the expression of MYCN-PARP-DDR pathway genes and NED markers, and inhibition with MYCNsi and/or PARPsi, BRCA1si, or RMI2si significantly suppressed malignant activities, including cell viability, colony formation, and cell migration, in C4-2b4 and NCI-H660 cells. Targeting this pathway with AURKA inhibitor PHA739358 and PARP inhibitor olaparib generated therapeutic effects similar to those of gene knockdown in vitro and significantly suppressed tumor growth in both C4-2b4 and MDACC PDX144-13C subcutaneous models in vivo Our results identify a novel MYCN-PARP-DDR pathway that is driven by N-MYC in a subset of CRPC-Adeno and in NEPC. Targeting this pathway using Conclusions: Our results identify a novel MYCN-PARP-DDR pathway that is driven by N-MYC in a subset of CRPC-Adeno and in NEPC. Targeting this pathway using in vitro CRPC-Adeno and CRPC-Neuro models demonstrated a novel therapeutic strategy for NEPC. Further investigation of N-MYC-regulated DDR gene targets and the biological and clinical significance of MYCN-PARP-DDR signaling will more fully elucidate the importance of the MYCN-PARP-DDR signaling pathway in the development and maintenance of NEPC. in vivo CRPC-Adeno and CRPC-Neuro models demonstrated a novel therapeutic strategy for NEPC. Further investigation of N-MYC-regulated DDR gene targets and the biological and clinical significance of MYCN-PARP-DDR signaling will more fully elucidate the importance of the MYCN-PARP-DDR signaling pathway in the development and maintenance of NEPC. Clin Cancer Res; 24(3); 696-707. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
40. In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development.
- Author
-
Jaworski FM, Gentilini LD, Gueron G, Meiss RP, Ortiz EG, Berguer PM, Ahmed A, Navone N, Rabinovich GA, Compagno D, Laderach DJ, and Vazquez ES
- Subjects
- Animals, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes pathology, Cell Proliferation drug effects, Disease Models, Animal, Galectin 1 antagonists & inhibitors, Gene Expression Regulation, Neoplastic drug effects, Heme Oxygenase-1 antagonists & inhibitors, Human Umbilical Vein Endothelial Cells drug effects, Humans, Male, Mice, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic genetics, Neovascularization, Pathologic pathology, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Xenograft Model Antitumor Assays, Galectin 1 genetics, Heme Oxygenase-1 genetics, Hemin administration & dosage, Prostatic Neoplasms drug therapy
- Abstract
Purpose: Conditioning strategies constitute a relatively unexplored and exciting opportunity to shape tumor fate by targeting the tumor microenvironment. In this study, we assessed how hemin, a pharmacologic inducer of heme oxygenase-1 (HO-1), has an impact on prostate cancer development in an in vivo conditioning model. Experimental Design: The stroma of C57BL/6 mice was conditioned by subcutaneous administration of hemin prior to TRAMP-C1 tumor challenge. Complementary in vitro and in vivo assays were performed to evaluate hemin effect on both angiogenesis and the immune response. To gain clinical insight, we used prostate cancer patient-derived samples in our studies to assess the expression of HO-1 and other relevant genes. Results: Conditioning resulted in increased tumor latency and decreased initial growth rate. Histologic analysis of tumors grown in conditioned mice revealed impaired vascularization. Hemin-treated human umbilical vein endothelial cells (HUVEC) exhibited decreased tubulogenesis in vitro only in the presence of TRAMP-C1-conditioned media. Subcutaneous hemin conditioning hindered tumor-associated neovascularization in an in vivo Matrigel plug assay. In addition, hemin boosted CD8
+ T-cell proliferation and degranulation in vitro and antigen-specific cytotoxicity in vivo A significant systemic increase in CD8+ T-cell frequency was observed in preconditioned tumor-bearing mice. Tumors from hemin-conditioned mice showed reduced expression of galectin-1 (Gal-1), key modulator of tumor angiogenesis and immunity, evidencing persistent remodeling of the microenvironment. We also found a subset of prostate cancer patient-derived xenografts and prostate cancer patient samples with mild HO-1 and low Gal-1 expression levels. Conclusions: These results highlight a novel function of a human-used drug as a means of boosting the antitumor response. Clin Cancer Res; 23(17); 5135-48. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
41. Targeting DNA Damage Response in Prostate Cancer by Inhibiting Androgen Receptor-CDC6-ATR-Chk1 Signaling.
- Author
-
Karanika S, Karantanos T, Li L, Wang J, Park S, Yang G, Zuo X, Song JH, Maity SN, Manyam GC, Broom B, Aparicio AM, Gallick GE, Troncoso P, Corn PG, Navone N, Zhang W, Li S, and Thompson TC
- Subjects
- Animals, Apoptosis drug effects, Ataxia Telangiectasia Mutated Proteins metabolism, Biomarkers metabolism, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, DNA Damage drug effects, DNA Replication drug effects, DNA-Binding Proteins metabolism, Humans, Male, Mice, Mice, Nude, Phosphorylation drug effects, Prostatic Neoplasms drug therapy, Signal Transduction drug effects, Thiophenes pharmacology, Urea analogs & derivatives, Urea pharmacology, Androgen Receptor Antagonists pharmacology, Cell Cycle Proteins metabolism, Checkpoint Kinase 1 metabolism, DNA Damage physiology, Nuclear Proteins metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism
- Abstract
Cell division cycle 6 (CDC6), an androgen receptor (AR) target gene, is implicated in regulating DNA replication and checkpoint mechanisms. CDC6 expression is increased during prostate cancer (PCa) progression and positively correlates with AR in PCa tissues. AR or CDC6 knockdown, together with AZD7762, a Chk1/2 inhibitor, results in decreased TopBP1-ATR-Chk1 signaling and markedly increased ataxia-telangiectasia-mutated (ATM) phosphorylation, a biomarker of DNA damage, and synergistically increases treatment efficacy. Combination treatment with the AR signaling inhibitor enzalutamide (ENZ) and the Chk1/2 inhibitor AZD7762 demonstrates synergy with regard to inhibition of AR-CDC6-ATR-Chk1 signaling, ATM phosphorylation induction, and apoptosis in VCaP (mutant p53) and LNCaP-C4-2b (wild-type p53) cells. CDC6 overexpression significantly reduced ENZ- and AZD7762-induced apoptosis. Additive or synergistic therapeutic activities are demonstrated in AR-positive animal xenograft models. These findings have important clinical implications, since they introduce a therapeutic strategy for AR-positive, metastatic, castration-resistant PCa, regardless of p53 status, through targeting AR-CDC6-ATR-Chk1 signaling., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
42. Heme oxygenase-1 in the forefront of a multi-molecular network that governs cell-cell contacts and filopodia-induced zippering in prostate cancer.
- Author
-
Paez AV, Pallavicini C, Schuster F, Valacco MP, Giudice J, Ortiz EG, Anselmino N, Labanca E, Binaghi M, Salierno M, Martí MA, Cotignola JH, Woloszynska-Read A, Bruno L, Levi V, Navone N, Vazquez ES, and Gueron G
- Subjects
- Animals, Cell Communication drug effects, Cell Line, Tumor, Cell Movement drug effects, Coculture Techniques, Crystallography, X-Ray, Culture Media, Conditioned pharmacology, Cytoskeleton drug effects, Cytoskeleton metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Male, Mice, Oligonucleotide Array Sequence Analysis, Prostatic Neoplasms genetics, Protein Binding drug effects, Proteomics, Pseudopodia drug effects, Sequence Analysis, RNA, Tandem Mass Spectrometry, Transcriptome drug effects, Transcriptome genetics, Cell Communication genetics, Gene Regulatory Networks drug effects, Heme Oxygenase-1 metabolism, Prostatic Neoplasms enzymology, Prostatic Neoplasms pathology, Pseudopodia metabolism
- Abstract
Prostate cancer (PCa) cells display abnormal expression of cytoskeletal proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown that heme oxygenase 1 (HO-1) is implicated in cell morphology regulation in PCa. Here, through a multi 'omics' approach we define the HO-1 interactome in PCa, identifying HO-1 molecular partners associated with the integrity of the cellular cytoskeleton. The bioinformatics screening for these cytoskeletal-related partners reveal that they are highly misregulated in prostate adenocarcinoma compared with normal prostate tissue. Under HO-1 induction, PCa cells present reduced frequency in migration events, trajectory and cell velocity and, a significant higher proportion of filopodia-like protrusions favoring zippering among neighboring cells. Moreover forced expression of HO-1 was also capable of altering cell protrusions in transwell co-culture systems of PCa cells with MC3T3 cells (pre-osteoblastic cell line). Accordingly, these effects were reversed under siHO. Transcriptomics profiling evidenced significant modulation of key markers related to cell adhesion and cell-cell communication under HO-1 induction. The integration from our omics-based research provides a four molecular pathway foundation (ANXA2/HMGA1/POU3F1; NFRSF13/GSN; TMOD3/RAI14/VWF; and PLAT/PLAU) behind HO-1 regulation of tumor cytoskeletal cell compartments. The complementary proteomics and transcriptomics approaches presented here promise to move us closer to unravel the molecular framework underpinning HO-1 involvement in the modulation of cytoskeleton pathways, pushing toward a less aggressive phenotype in PCa.
- Published
- 2016
- Full Text
- View/download PDF
43. Association of HO-1 and BRCA1 Is Critical for the Maintenance of Cellular Homeostasis in Prostate Cancer.
- Author
-
Labanca E, De Luca P, Gueron G, Paez A, Moiola CP, Massillo C, Porretti J, Giudice J, Zalazar F, Navone N, Vazquez E, and De Siervi A
- Subjects
- Animals, Cell Line, Tumor, DNA Damage genetics, Heme Oxygenase-1 metabolism, Heterografts, Humans, Male, Mice, NF-E2-Related Factor 2 metabolism, Oxidative Stress genetics, Prostatic Neoplasms pathology, Protein Binding, Transcriptional Activation, BRCA1 Protein metabolism, Heme Oxygenase-1 genetics, Prostatic Neoplasms metabolism
- Abstract
Unlabelled: Prostate cancer is the second leading cause of cancer-related death in men worldwide. Many factors that participate in the development of prostate cancer promote imbalance in the redox state of the cell. Accumulation of reactive oxygen species causes injury to cell structures, ultimately leading to cancer development. The antioxidant enzyme heme oxygenase 1 (HMOX1/HO-1) is responsible for the maintenance of the cellular homeostasis, playing a critical role in the oxidative stress and the regulation of prostate cancer development and progression. In the present study, the transcriptional regulation of HO-1 was investigated in prostate cancer. Interestingly, the tumor suppressor BRCA1 binds to the HO-1 promoter and modulates HO-1, inducing its protein levels through both the increment of its promoter activity and the induction of its transcriptional activation. In addition, in vitro and in vivo analyses show that BRCA1 also controls HO-1-negative targets: MMP9, uPA, and Cyclin D1. HO-1 transcriptional regulation is also modulated by oxidative and genotoxic agents. Induction of DNA damage by mitoxantrone and etoposide repressed HO-1 transcription, whereas hydrogen peroxide and doxorubicin induced its expression. Xenograft studies showed that HO-1 regulation by doxorubicin also occurs in vivo. Immunofluorescence analysis revealed that BRCA1 overexpression and/or doxorubicin exposure induced the cytoplasmic retention of HO-1. Finally, the transcription factor NRF2 cooperates with BRCA1 protein to activate HO-1 promoter activity. In summary, these results show that the activation of BRCA1-NRF2/HO-1 axis defines a new mechanism for the maintenance of the cellular homeostasis in prostate cancer., Implications: Oxidative and genotoxic stress converge on HO-1 transcriptional activity through the combined actions of BRCA1 and NRF2., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
44. Monocytes and CD4+ T cells contribution to the under-expression of NR4A2 and TNFAIP3 genes in patients with multiple sclerosis.
- Author
-
Navone ND, Perga S, Martire S, Berchialla P, Malucchi S, and Bertolotto A
- Subjects
- Adult, DNA-Binding Proteins genetics, Disability Evaluation, Female, Humans, Intracellular Signaling Peptides and Proteins genetics, Male, Middle Aged, Nuclear Proteins genetics, Nuclear Receptor Subfamily 4, Group A, Member 2 genetics, RNA, Messenger metabolism, Statistics, Nonparametric, Tumor Necrosis Factor alpha-Induced Protein 3, Young Adult, CD4-Positive T-Lymphocytes metabolism, DNA-Binding Proteins metabolism, Gene Expression Regulation physiology, Intracellular Signaling Peptides and Proteins metabolism, Monocytes metabolism, Multiple Sclerosis pathology, Nuclear Proteins metabolism, Nuclear Receptor Subfamily 4, Group A, Member 2 metabolism
- Abstract
We recently found a gene signature for multiple sclerosis (MS) that reverted to normal during pregnancy in MS patients and included NR4A2 and TNFAIP3, key molecules in anti-inflammatory processes. Here we focus on the expression levels of these two genes in monocytes and CD4+ T cells from healthy controls and treatment-naïve RRMS patients. Our findings show that monocytes play a key role in the dysregulated anti-inflammatory response, being the expression of both genes down-regulated in these cells in RRMS patients with respect to healthy individuals. CD4+ T cells seem to have only a marginal part, because we can observe only a slight down-regulation in NR4A2., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
45. Heme-oxygenase-1 implications in cell morphology and the adhesive behavior of prostate cancer cells.
- Author
-
Gueron G, Giudice J, Valacco P, Paez A, Elguero B, Toscani M, Jaworski F, Leskow FC, Cotignola J, Marti M, Binaghi M, Navone N, and Vazquez E
- Subjects
- Animals, Cell Adhesion, Down-Regulation, Humans, Male, Mice, Mice, Nude, Prostatic Neoplasms metabolism, Reactive Oxygen Species, Signal Transduction, Xenograft Model Antitumor Assays, Cadherins metabolism, Heme Oxygenase-1 genetics, Prostatic Neoplasms genetics, beta Catenin metabolism
- Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men. Although previous studies in PCa have focused on cell adherens junctions (AJs), key players in metastasis, they have left the molecular mechanisms unexplored. Inflammation and the involvement of reactive oxygen species (ROS) are critical in the regulation of cell adhesion and the integrity of the epithelium. Heme oxygenase-1 (HO-1) counteracts oxidative and inflammatory damage. Here, we investigated whether HO-1 is implicated in the adhesive and morphological properties of tumor cells. Genes differentially regulated by HO-1 were enriched for cell motility and adhesion biological processes. HO-1 induction, increased E-cadherin and β-catenin levels. Immunofluorescence analyses showed a striking remodeling of E-cadherin/ β-catenin based AJs under HO-1 modulation. Interestingly, the enhanced levels of E-cadherin and β-catenin coincided with a markedly change in cell morphology. To further our analysis we sought to identify HO-1 binding proteins that might participate in the regulation of cell morphology. A proteomics approach identified Muskelin, as a novel HO-1 partner, strongly implicated in cell morphology regulation. These results define a novel role for HO-1 in modulating the architecture of cell-cell interactions, favoring a less aggressive phenotype and further supporting its anti-tumoral function in PCa.
- Published
- 2014
- Full Text
- View/download PDF
46. Targeting poly(ADP-ribose) polymerase and the c-Myb-regulated DNA damage response pathway in castration-resistant prostate cancer.
- Author
-
Li L, Chang W, Yang G, Ren C, Park S, Karantanos T, Karanika S, Wang J, Yin J, Shah PK, Takahiro H, Dobashi M, Zhang W, Efstathiou E, Maity SN, Aparicio AM, Li Ning Tapia EM, Troncoso P, Broom B, Xiao L, Lee HS, Lee JS, Corn PG, Navone N, and Thompson TC
- Subjects
- Animals, BRCA1 Protein genetics, BRCA1 Protein metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Castration, Cell Cycle Checkpoints drug effects, Cell Cycle Checkpoints genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Humans, Male, Mice, Mice, Nude, Nuclear Proteins genetics, Nuclear Proteins metabolism, Poly(ADP-ribose) Polymerases genetics, Poly(ADP-ribose) Polymerases metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Proto-Oncogene Proteins c-myb genetics, Proto-Oncogene Proteins c-myb metabolism, Receptors, Androgen genetics, Receptors, Androgen metabolism, Urea pharmacology, Xenograft Model Antitumor Assays, DNA Damage, Phthalazines pharmacology, Piperazines pharmacology, Poly(ADP-ribose) Polymerase Inhibitors, Prostatic Neoplasms drug therapy, Proto-Oncogene Proteins c-myb antagonists & inhibitors, Thiophenes pharmacology, Urea analogs & derivatives
- Abstract
Androgen deprivation is the standard treatment for advanced prostate cancer (PCa), but most patients ultimately develop resistance and tumor recurrence. We found that MYB is transcriptionally activated by androgen deprivation therapy or genetic silencing of the androgen receptor (AR). MYB silencing inhibited PCa growth in culture and xenografts in mice. Microarray data revealed that c-Myb and AR shared a subset of target genes that encode DNA damage response (DDR) proteins, suggesting that c-Myb may supplant AR as the dominant regulator of their common DDR target genes in AR inhibition-resistant or AR-negative PCa. Gene signatures including AR, MYB, and their common DDR-associated target genes positively correlated with metastasis, castration resistance, tumor recurrence, and decreased survival in PCa patients. In culture and in xenograft-bearing mice, a combination strategy involving the knockdown of MYB, BRCA1, or TOPBP1 or the abrogation of cell cycle checkpoint arrest with AZD7762, an inhibitor of the checkpoint kinase Chk1, increased the cytotoxicity of the poly[adenosine 5'-diphosphate (ADP)-ribose] polymerase (PARP) inhibitor olaparib in PCa cells. Our results reveal new mechanism-based therapeutic approaches for PCa by targeting PARP and the DDR pathway involving c-Myb, TopBP1, ataxia telangiectasia mutated- and Rad3-related (ATR), and Chk1., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
47. PCAT-1, a long noncoding RNA, regulates BRCA2 and controls homologous recombination in cancer.
- Author
-
Prensner JR, Chen W, Iyer MK, Cao Q, Ma T, Han S, Sahu A, Malik R, Wilder-Romans K, Navone N, Logothetis CJ, Araujo JC, Pisters LL, Tewari AK, Canman CE, Knudsen KE, Kitabayashi N, Rubin MA, Demichelis F, Lawrence TS, Chinnaiyan AM, and Feng FY
- Subjects
- 3' Untranslated Regions, Animals, Antineoplastic Agents pharmacology, BRCA2 Protein metabolism, Cell Death drug effects, Cell Line, Tumor, DNA Damage, Humans, Male, Mice, Mice, SCID, Phthalazines pharmacology, Piperazines pharmacology, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerase Inhibitors, Poly(ADP-ribose) Polymerases metabolism, Prostatic Neoplasms metabolism, RNA Interference, RNA, Long Noncoding metabolism, Xenograft Model Antitumor Assays, BRCA2 Protein genetics, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms genetics, RNA, Long Noncoding genetics, Recombinational DNA Repair
- Abstract
Impairment of double-stranded DNA break (DSB) repair is essential to many cancers. However, although mutations in DSB repair proteins are common in hereditary cancers, mechanisms of impaired DSB repair in sporadic cancers remain incompletely understood. Here, we describe the first role for a long noncoding RNA (lncRNA) in DSB repair in prostate cancer. We identify PCAT-1, a prostate cancer outlier lncRNA, which regulates cell response to genotoxic stress. PCAT-1 expression produces a functional deficiency in homologous recombination through its repression of the BRCA2 tumor suppressor, which, in turn, imparts a high sensitivity to small-molecule inhibitors of PARP1. These effects reflected a posttranscriptional repression of the BRCA2 3'UTR by PCAT-1. Our observations thus offer a novel mechanism of "BRCAness" in sporadic cancers., (©2014 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
48. Heme oxygenase-1 (HO-1) expression in prostate cancer cells modulates the oxidative response in bone cells.
- Author
-
Ferrando M, Wan X, Meiss R, Yang J, De Siervi A, Navone N, and Vazquez E
- Subjects
- Animals, Blotting, Western, Cell Cycle drug effects, Cell Line, Cell Line, Tumor, Cell Proliferation drug effects, Cells, Cultured, Flow Cytometry, Heme Oxygenase-1 genetics, Hemin pharmacology, Humans, Immunohistochemistry, Male, Mice, Mice, SCID, Organ Culture Techniques, Osteoblasts drug effects, Prostatic Neoplasms genetics, Heme Oxygenase-1 metabolism, Osteoblasts metabolism, Prostatic Neoplasms metabolism
- Abstract
Prostate cancer (PCa) is a leading cause of death among males. It is currently estimated that inflammatory responses are linked to 15-20% of all deaths from cancer worldwide. PCa is dominated by complications arising from metastasis to the bone where the tumor cells interact with the bone microenvironment impairing the balance between bone formation and degradation. However, the molecular nature of this interaction is not completely understood. Heme oxygenase-1 (HO-1) counteracts oxidative damage and inflammation. Previous studies from our laboratory showed that HO-1 is implicated in PCa, demonstrating that endogenous HO-1 inhibits bone derived-prostate cancer cells proliferation, invasion and migration and decreases tumor growth and angiogenesis in vivo. The aim of this work was to analyze the impact of HO-1 modulated PCa cells on osteoblasts proliferation in vitro and on bone remodeling in vivo. Using a co-culture system of PC3 cells with primary mice osteoblasts (PMOs), we demonstrated that HO-1 pharmacological induction (hemin treatment) abrogated the diminution of PMOs proliferation induced by PCa cells and decreased the expression of osteoclast-modulating factors in osteoblasts. No changes were detected in the expression of genes involved in osteoblasts differentiation. However, co-culture of hemin pre-treated PC3 cells (PC3 Hem) with PMOs provoked an oxidative status and activated FoxO signaling in osteoblasts. The percentage of active osteoblasts positive for HO-1 increased in calvarias explants co-cultured with PC3 Hem cells. Nuclear HO-1 expression was detected in tumors generated by in vivo bone injection of HO-1 stable transfected PC3 (PC3HO-1) cells in the femur of SCID mice. These results suggest that HO-1 has the potential to modify the bone microenvironment impacting on PCa bone metastasis.
- Published
- 2013
- Full Text
- View/download PDF
49. Targeting fibroblast growth factor pathways in prostate cancer.
- Author
-
Corn PG, Wang F, McKeehan WL, and Navone N
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Humans, Male, Prostatic Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Receptors, Fibroblast Growth Factor metabolism, Fibroblast Growth Factors metabolism, Prostatic Neoplasms metabolism, Signal Transduction drug effects
- Abstract
Advanced prostate cancer carries a poor prognosis and novel therapies are needed. Research has focused on identifying mechanisms that promote angiogenesis and cellular proliferation during prostate cancer progression from the primary tumor to bone-the principal site of prostate cancer metastases. One candidate pathway is the fibroblast growth factor (FGF) axis. Aberrant expression of FGF ligands and FGF receptors leads to constitutive activation of multiple downstream pathways involved in prostate cancer progression including mitogen-activated protein kinase, phosphoinositide 3-kinase, and phospholipase Cγ. The involvement of FGF pathways in multiple mechanisms relevant to prostate tumorigenesis provides a rationale for the therapeutic blockade of this pathway, and two small-molecule tyrosine kinase inhibitors-dovitinib and nintedanib-are currently in phase II clinical development for advanced prostate cancer. Preliminary results from these trials suggest that FGF pathway inhibition represents a promising new strategy to treat castrate-resistant disease.
- Published
- 2013
- Full Text
- View/download PDF
50. Unveiling the association of STAT3 and HO-1 in prostate cancer: role beyond heme degradation.
- Author
-
Elguero B, Gueron G, Giudice J, Toscani MA, De Luca P, Zalazar F, Coluccio-Leskow F, Meiss R, Navone N, De Siervi A, and Vazquez E
- Subjects
- Animals, Cell Line, Tumor, Cytoplasm metabolism, Disease Models, Animal, Gene Expression, Heme Oxygenase-1 genetics, Humans, Male, Matrix Metalloproteinase 9 metabolism, Mice, Promoter Regions, Genetic, Prostate-Specific Antigen genetics, Prostatic Neoplasms genetics, Protein Binding, Protein Transport, Receptors, Androgen metabolism, Signal Transduction, Transplantation, Heterologous, Heme metabolism, Heme Oxygenase-1 metabolism, Prostatic Neoplasms metabolism, STAT3 Transcription Factor metabolism
- Abstract
Activation of the androgen receptor (AR) is a key step in the development of prostate cancer (PCa). Several mechanisms have been identified in AR activation, among them signal transducer and activator of transcription 3 (STAT3) signaling. Disruption of STAT3 activity has been associated to cancer progression. Recent studies suggest that heme oxygenase 1 (HO-1) may play a key role in PCa that may be independent of its catalytic function. We sought to explore whether HO-1 operates on AR transcriptional activity through the STAT3 axis. Our results display that HO-1 induction in PCa cells represses AR activation by decreasing the prostate-specific antigen (PSA) promoter activity and mRNA levels. Strikingly, this is the first report to show by chromatin immunoprecipitation analysis that HO-1 associates to gene promoters, revealing a novel function for HO-1 in the nucleus. Furthermore, HO-1 and STAT3 directly interact as determined by co-immunoprecipitation studies. Forced expression of HO-1 increases STAT3 cytoplasmic retention. When PCa cells were transfected with a constitutively active STAT3 mutant, PSA and STAT3 downstream target genes were abrogated under hemin treatment. Additionally, a significant decrease in pSTAT3 protein levels was detected in the nuclear fraction of these cells. Confocal microscopy images exhibit a decreased rate of AR/STAT3 nuclear co-localization under hemin treatment. In vivo studies confirmed that STAT3 nuclear delimitation was significantly decreased in PC3 tumors overexpressing HO-1 grown as xenografts in nude mice. These results provide a novel function for HO-1 down-modulating AR transcriptional activity in PCa, interfering with STAT3 signaling, evidencing its role beyond heme degradation.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.