1
|
Hata J, Harigane Y, Matsuoka K, Akaihata H, Yaginuma K, Meguro S, Hoshi S, Sato Y, Ogawa S, Uemura M, Kojima Y. Mechanism of Androgen-Independent Stromal Proliferation in Benign Prostatic Hyperplasia. Int J Mol Sci 2023; 24:11634. [PMID: 37511400 PMCID: PMC10380833 DOI: 10.3390/ijms241411634] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a chronic proliferative disease showing stromal-dominant proliferation. However, the detailed proliferation mechanism has remained unclear. Although aging and androgen have been reported as definitive risk factors for BPH, recent studies have focused on the involvement of androgen-independent factors. Androgen-independent factors include ischemia, oxidative stress, metabolic syndrome, infection, autoimmune reactions, and inflammation, with inflammation in BPH tissues playing a central role in the BPH proliferative process. Inflammation in BPH tissues by various factors finally leads to tissue remodeling and stromal proliferation through the wound healing process of the prostate. To elucidate the proliferative mechanism of BPH, a study using whole-genome gene expression analysis in a stromal-dominant BPH rat model was performed and showed that immune response-related pathways and complement classical pathways are activated. Furthermore, expression analysis using this BPH rat model showed that the autoimmune reaction triggered complement pathway activation in the proliferative process of BPH. BPH is a multifactorial disease, and understanding the role of androgen-independent factors including immune responses contributes to elucidating the pathogenesis of BPH. Androgen-independent factors may lead to new therapeutic targets for BPH, and further development of this research is expected.
Collapse
Affiliation(s)
- Junya Hata
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Yuki Harigane
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Kanako Matsuoka
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Hidenori Akaihata
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Kei Yaginuma
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Satoru Meguro
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Seiji Hoshi
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Yuichi Sato
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Soichiro Ogawa
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Motohide Uemura
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| | - Yoshiyuki Kojima
- Department of Urology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 9601295, Japan
| |
Collapse
|
2
|
Yuan YF, Zhu WX, Liu T, He JQ, Zhou Q, Zhou X, Zhang X, Yang J. Cyclopamine functions as a suppressor of benign prostatic hyperplasia by inhibiting epithelial and stromal cell proliferation via suppression of the Hedgehog signaling pathway. Int J Mol Med 2020; 46:311-319. [PMID: 32319534 PMCID: PMC7255449 DOI: 10.3892/ijmm.2020.4569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 03/12/2019] [Indexed: 12/17/2022] Open
Abstract
Stromal-epithelial interaction serves a pivotal role in normal prostate growth, as well as the onset of benign prostatic hyperplasia (BPH). The present study aimed to explore the role of cyclopamine in the proliferation and apoptosis of epithelial and stromal cells in rats with BPH by blocking the Hedgehog signaling pathway. Cyclopamine (an inhibitor of the Hedgehog signaling pathway) was administered in a rat model of BPH, and the expression of Ki67 (proliferation factor) was determined by immunohistochemistry. In addition, epithelial and stromal cells were separated and cultured in order to investigate the role of cyclopamine in the progression of BPH. The expression of Hedgehog signaling pathway- and apoptosis-related genes, including basic fibroblastic growth factor (b-FGF) and transforming growth factor β (TGF-β), was evaluated using reverse transcription-quantitative polymerase chain reaction and western blot analysis. Cell proliferation, cell cycle and apoptosis were analyzed using an MTT assay and flow cytometry. We identified upregulated Ki67 expression and activated Hedgehog signaling pathway in rats with BPH. Cyclopamine inhibited the activation of the Hedgehog signaling pathway. In response to cyclopamine treatment, epithelial and stromal cell proliferation was inhibited; this was concomitant with decreased Ki67, TGF-β, and b-FGF expression. On the other hand, epithelial cell apoptosis was enhanced, which was associated with increased Bax and reduced Bcl-2 expression. Based on these findings, we proposed that cyclopamine may serve as a potential therapeutic agent in the treatment of BPH. Cyclopamine could inhibit epithelial and stromal cell proliferation, and induce epithelial cell apoptosis by suppressing the Hedgehog signaling pathway.
Collapse
Affiliation(s)
- Yi-Feng Yuan
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Wen-Xiong Zhu
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Tao Liu
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Ju-Qiao He
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Qing Zhou
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xing Zhou
- Department of Andrology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xi Zhang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jing Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
3
|
Cunha GR, Sinclair A, Ricke WA, Robboy SJ, Cao M, Baskin LS. Reproductive tract biology: Of mice and men. Differentiation 2019; 110:49-63. [PMID: 31622789 PMCID: PMC7339118 DOI: 10.1016/j.diff.2019.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022]
Abstract
The study of male and female reproductive tract development requires expertise in two separate disciplines, developmental biology and endocrinology. For ease of experimentation and economy, the mouse has been used extensively as a model for human development and pathogenesis, and for the most part similarities in developmental processes and hormone action provide ample justification for the relevance of mouse models for human reproductive tract development. Indeed, there are many examples describing the phenotype of human genetic disorders that have a reasonably comparable phenotype in mice, attesting to the congruence between mouse and human development. However, anatomic, developmental and endocrinologic differences exist between mice and humans that (1) must be appreciated and (2) considered with caution when extrapolating information between all animal models and humans. It is critical that the investigator be aware of both the similarities and differences in organogenesis and hormone action within male and female reproductive tracts so as to focus on those features of mouse models with clear relevance to human development/pathology. This review, written by a team with extensive expertise in the anatomy, developmental biology and endocrinology of both mouse and human urogenital tracts, focusses upon the significant human/mouse differences, and when appropriate voices a cautionary note regarding extrapolation of mouse models for understanding development of human male and female reproductive tracts.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA; George M. O'Brien Center of Research Excellence, Department of Urology, University of Wisconsin, Madison, WI, 93705, USA; Department of Pathology, Duke University, Davison Building, Box 3712, Durham, NC, 27710, USA.
| | - Adriane Sinclair
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Will A Ricke
- George M. O'Brien Center of Research Excellence, Department of Urology, University of Wisconsin, Madison, WI, 93705, USA
| | - Stanley J Robboy
- Department of Pathology, Duke University, Davison Building, Box 3712, Durham, NC, 27710, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
4
|
The regulation of Hh/Gli1 signaling cascade involves Gsk3β- mediated mechanism in estrogen-derived endometrial hyperplasia. Sci Rep 2017; 7:6557. [PMID: 28747625 PMCID: PMC5529438 DOI: 10.1038/s41598-017-06370-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022] Open
Abstract
The present study was undertaken to explore the functional involvement of Hh signaling and its regulatory mechanism in endometrial hyperplasia. Differential expression of Hh signaling molecules i.e., Ihh, Shh, Gli1 or Gsk3β was observed in endometrial hyperplasial (EH) cells as compared to normal endometrial cells. Estradiol induced the expression of Hh signaling molecules and attenuated the expression of Gsk3β whereas anti-estrogen (K1) or progestin (MPA) suppressed these effects in EH cells. Cyclopamine treatment or Gli1 siRNA knockdown suppressed the growth of EH cells and reduced the expression of proliferative markers. Estradiol also induced the nuclear translocation of Gli1 which was suppressed by both MPA and K1 in EH cells. While exploring non-canonical mechanism, LY-294002 (Gsk3β activator) caused a decrease in Gli1 expression indicating the involvement of Gsk3β in Gli1 regulation. Further, Gsk3β silencing promoted the expression and nuclear translocation of Gli1 demonstrating that Gsk3β serves as a negative kinase regulator of Gli1 in EH cells. Similar attenuation of Hh signaling molecules was observed in rats with uterine hyperplasia undergoing anti-estrogen treatment. The study suggested that Hh/Gli1 cascade (canonical pathway) as well as Gsk3β-Gli1 crosstalk (non-canonical pathway) play crucial role in estrogen-dependent cell proliferation in endometrial hyperplasia.
Collapse
|
5
|
Wen S, Chang HC, Tian J, Shang Z, Niu Y, Chang C. Stromal androgen receptor roles in the development of normal prostate, benign prostate hyperplasia, and prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:293-301. [PMID: 25432062 PMCID: PMC4305176 DOI: 10.1016/j.ajpath.2014.10.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 10/13/2014] [Accepted: 10/20/2014] [Indexed: 02/05/2023]
Abstract
The prostate is an androgen-sensitive organ that needs proper androgen/androgen receptor (AR) signals for normal development. The progression of prostate diseases, including benign prostate hyperplasia (BPH) and prostate cancer (PCa), also needs proper androgen/AR signals. Tissue recombination studies report that stromal, but not epithelial, AR plays more critical roles via the mesenchymal-epithelial interactions to influence the early process of prostate development. However, in BPH and PCa, much more attention has been focused on epithelial AR roles. However, accumulating evidence indicates that stromal AR is also irreplaceable and plays critical roles in prostate disease progression. Herein, we summarize the roles of stromal AR in the development of normal prostate, BPH, and PCa, with evidence from the recent results of in vitro cell line studies, tissue recombination experiments, and AR knockout animal models. Current evidence suggests that stromal AR may play positive roles to promote BPH and PCa progression, and targeting stromal AR selectively with AR degradation enhancer, ASC-J9, may allow development of better therapies with fewer adverse effects to battle BPH and PCa.
Collapse
Affiliation(s)
- Simeng Wen
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Hong-Chiang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York
| | - Jing Tian
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjie Niu
- Chawnshang Chang Sex Hormone Research Center, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Chawnshang Chang
- Departments of Pathology and Urology, George Whipple Lab for Cancer Research, Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York; Sex Hormone Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
6
|
Hedgehog signaling in prostate epithelial-mesenchymal growth regulation. Dev Biol 2015; 400:94-104. [PMID: 25641695 DOI: 10.1016/j.ydbio.2015.01.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 12/24/2022]
Abstract
The prostate gland plays an important role in male reproduction, and is also an organ prone to diseases such as benign prostatic hyperplasia (BPH) and prostate cancer. The prostate consists of ducts with an inner layer of epithelium surrounded by stroma. Reciprocal signaling between these two cell compartments is instrumental to normal prostatic development, homeostasis, regeneration, as well as tumor formation. Hedgehog (HH) signaling is a master regulator in numerous developmental processes. In many organs, HH plays a key role in epithelial-mesenchymal signaling that regulates organ growth and tissue differentiation, and abnormal HH signaling has been implicated in the progression of various epithelial carcinomas. In this review, we focus on recent studies exploring the multipotency of endogenous postnatal and adult epithelial and stromal stem cells and studies addressing the role of HH in prostate development and cancer. We discuss the implications of the results for a new understanding of prostate development and disease. Insight into the cellular and molecular mechanisms underlying epithelial-mesenchymal growth regulation should provide a basis for devising innovative therapies to combat diseases of the prostate.
Collapse
|
7
|
NVP-LDE-225 (Erismodegib) inhibits epithelial-mesenchymal transition and human prostate cancer stem cell growth in NOD/SCID IL2Rγ null mice by regulating Bmi-1 and microRNA-128. Oncogenesis 2013; 2:e42. [PMID: 23567619 PMCID: PMC3641359 DOI: 10.1038/oncsis.2013.5] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer stem cells (CSCs) are defined by their extensive self-renewal, differentiation and tumor initiation properties. It is now clear that CSCs are involved in tumor growth and recurrence, and resistance to conventional treatments. The sonic hedgehog (Shh) pathway has a crucial role in stemness and tumorigenesis. Thus, the strategy that suppresses stemness and consequently tumorigenic potential of CSCs could be considered for the management of prostate cancer. The objectives of this study were to examine the molecular mechanisms, by which NVP-LDE-225/Erismodegib (smoothened inhibitor) regulates stem cell characteristics and tumor growth in prostate cancer. The effects of NVP-LDE-225 on CSC's viability, sphere formation, apoptosis, epithelial-mesenchymal transition (EMT) and tumor growth in NOD/SCID IL2Rγ null mice were examined. NVP-LDE-225 inhibited cell viability and spheroid formation, and induced apoptosis by activation of caspase-3 and cleavage of poly-ADP ribose polymerase (PARP). NVP-LDE-225 induced expression of Bax and Bak, and inhibited the expression of Bcl-2, Bcl-XL, XIAP, cIAP1, cIAP2 and survivin. NVP-LDE-225 inhibited Gli transcriptional activity, Gli-DNA interaction and the expression of Gli1, Gli2, Patched1 and Patched-2 in prostate CSCs. Interestingly, NVP-LDE-225 induced PDCD4 and apoptosis and inhibited cell viability by suppressing miR-21. Furthermore, NVP-LDE-225 inhibited pluripotency-maintaining factors Nanog, Oct-4, c-Myc and Sox-2. The inhibition of Bmi-1 by NVP-LDE-225 was regulated by upregulation of miR-128. NVP-LDE-225 suppressed EMT by upregulating E-cadherin and inhibiting N-cadherin, Snail, Slug and Zeb1 by regulating the miR-200 family. Finally, NVP-LDE-225 inhibited CSC tumor growth, which was associated with the suppression of Gli1, Gli2, Patched-1, Patched-2, Cyclin D1, Bmi-1 and PCNA and cleavage of caspase-3 and PARP in tumor tissues derived from NOD/SCID IL2Rγ null mice. Overall, our findings suggest that inhibition of the Shh signaling pathway could therefore be a novel therapeutic option in treating prostate cancer.
Collapse
|
8
|
Li Y, Maitah MY, Ahmad A, Kong D, Bao B, Sarkar FH. Targeting the Hedgehog signaling pathway for cancer therapy. Expert Opin Ther Targets 2012; 16:49-66. [PMID: 22243133 DOI: 10.1517/14728222.2011.617367] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Hedgehog (Hh) signaling pathway plays key roles in embryonic development, formation and maintenance of cancer stem cells (CSCs) and acquisition of epithelial-to-mesenchymal transition (EMT). Since CSCs and EMT are important biological factors responsible for cancer cell invasion, metastasis, drug resistance and tumor recurrence, the Hh signaling pathway is believed to be an important target for cancer therapy. AREAS COVERED In recent years, small-molecule inhibitors of Hh signaling have been synthesized for cancer treatment. Clinical trials using these inhibitors are being conducted to determine their toxicity profiles and efficacies. In addition, nutraceuticals (such as isoflavones, curcumin, vitamin D, etc) have been shown to inhibit cancer growth through downregulation of Hh signaling. EXPERT OPINION Inhibition of Hh signaling is important for suppression of cancer growth, invasion, metastasis and recurrence in cancer therapy. However, targeting only one molecule in Hh signaling may not be sufficient to kill cancer cells because cancers show deregulation of multiple signals. Therefore, utilizing new technologies to determine alterations in Hh and other signals for individuals and designing combination strategies with small-molecule Hh inhibitors, nutraceuticals and other chemotherapeutics in targeted personalized therapy could have a significant effect on improving the overall survival of patients with cancers.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
9
|
Hedgehog signaling plays roles in epithelial cell proliferation in neonatal mouse uterus and vagina. Cell Tissue Res 2012; 348:239-47. [PMID: 22388655 DOI: 10.1007/s00441-012-1350-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 01/20/2012] [Indexed: 10/28/2022]
Abstract
Both the uterus and vagina develop from the Müllerian duct but are quite distinct in morphology and function. To investigate factors controlling epithelial differentiation and cell proliferation in neonatal uterus and vagina, we focused on Hedgehog (HH) signaling. In neonatal mice, Sonic hh (Shh) was localized in the vaginal epithelium and Indian hh (Ihh) was slightly expressed in the uterus and vagina, whereas all Glioma-associated oncogene homolog (Gli) genes were mainly expressed in the stroma. The expression of target genes of HH signaling was high in the neonatal vagina and in the uterus, it increased with growth. Thus, in neonatal mice, Shh in the vaginal epithelium and Ihh in the uterus and vagina activated HH signaling in the stroma. Tissue recombinants showed that vaginal Shh expression was inhibited by the vaginal stroma and uterine Ihh expression was stimulated by the uterine stroma. Addition of a HH signaling inhibitor decreased epithelial cell proliferation in organ-cultured uterus and vagina and increased stromal cell proliferation in organ-cultured uterus. However, it did not affect epithelial differentiation or the expression of growth factors in organ-cultured uterus and vagina. Thus, activated HH signaling stimulates epithelial cell proliferation in neonatal uterus and vagina but inhibits stromal cell proliferation in neonatal uterus.
Collapse
|
10
|
Timms BG, Hofkamp LE. Prostate development and growth in benign prostatic hyperplasia. Differentiation 2011; 82:173-83. [DOI: 10.1016/j.diff.2011.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 06/22/2011] [Accepted: 08/04/2011] [Indexed: 11/15/2022]
|
11
|
Gioeli D, Wunderlich W, Sebolt-Leopold J, Bekiranov S, Wulfkuhle JD, Petricoin EF, Conaway M, Weber MJ. Compensatory pathways induced by MEK inhibition are effective drug targets for combination therapy against castration-resistant prostate cancer. Mol Cancer Ther 2011; 10:1581-90. [PMID: 21712477 DOI: 10.1158/1535-7163.mct-10-1033] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Targeted therapies have often given disappointing results when used as single agents in solid tumors, suggesting the importance of devising rational combinations of targeted drugs. We hypothesized that construction of such combinations could be guided by identification of growth and survival pathways whose activity or expression become upregulated in response to single-agent drug treatment. We mapped alterations in signaling pathways assessed by gene array and protein phosphorylation to identify compensatory signal transduction pathways in prostate cancer xenografts treated with a MAP/ERK kinase (MEK) inhibitor PD325901. In addition to numerous components of the extracellular signal-regulated kinase (ERK) signaling pathway, components of the IKK, hedgehog, and phosphoinositide 3-kinase/Akt/mTOR pathways were upregulated following treatment with PD325901. Combinations of PD325901 with inhibitors of any one of these upregulated pathways provided synergistically greater growth inhibition of in vitro cell growth and survival than the individual drugs alone. Thus, the identification of compensatory signal transduction pathways paves the way for rational combinatorial therapies for the effective treatment of prostate cancer.
Collapse
Affiliation(s)
- Daniel Gioeli
- Department of Microbiology, PO Box 800734, University of Virginia Health System, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
The role of p63 in cancer, stem cells and cancer stem cells. Cell Mol Biol Lett 2011; 16:296-327. [PMID: 21442444 PMCID: PMC6275999 DOI: 10.2478/s11658-011-0009-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 03/07/2011] [Indexed: 01/01/2023] Open
Abstract
The transcription factor p63 has important functions in tumorigenesis, epidermal differentiation and stem cell self-renewal. The TP63 gene encodes multiple protein isoforms that have different or even antagonistic roles in these processes. The balance of p63 isoforms, together with the presence or absence of the other p53 family members, p73 and p53, has a striking biological impact. There is increasing evidence that interactions between p53-family members, whether cooperative or antagonistic, are involved in various cell processes. This review summarizes the current understanding of the role of p63 in tumorigenesis, metastasis, cell migration and senescence. In particular, recent data indicate important roles in adult stem cell and cancer stem cell regulation and in the response of cancer cells to therapy.
Collapse
|
13
|
Abstract
Multiple cellular signaling pathways have been involved in the processes of cancer cell invasion and metastasis. Among many signaling pathways, Wnt and Hedgehog (Hh) signaling pathways are critically involved in embryonic development, in the biology of cancer stem cells (CSCs) and in the acquisition of epithelial to mesenchymal transition (EMT), and thus this article will remain focused on Wnt and Hh signaling. Since CSCs and EMT are also known to be responsible for cancer cell invasion and metastasis, the Wnt and Hedgehog signaling pathways are also intimately associated with cancer invasion and metastasis. Emerging evidence suggests the beneficial role of chemopreventive agents commonly known as nutraceutical in cancer. Among many such agents, soy isoflavones, curcumin, green tea polyphenols, 3,3'-diindolylmethane, resveratrol, lycopene, vitamin D, etc. have been found to prevent, reverse, or delay the carcinogenic process. Interestingly, these agents have also shown to prevent or delay the progression of cancer, which could in part be due to their ability to attack CSCs or EMT-type cells by attenuating the Wnt and Hedgehog signaling pathways. In this review, we summarize the current state of our knowledge on the role of Wnt and Hedgehog signaling pathways, and their targeted inactivation by chemopreventive agents (nutraceuticals) for the prevention of tumor progression and/or treatment of human malignancies.
Collapse
|
14
|
Abstract
Among many endocrine-related cancers, prostate cancer (PCa) is the most frequent male malignancy, and it is the second most common cause of cancer-related death in men in the United States. Therefore, this review focuses on summarizing the knowledge of molecular signaling pathways in PCa because, in order to better design new preventive strategies for the fight against PCa, documentation of the knowledge on the pathogenesis of PCa at the molecular level is very important. Cancer cells are known to have alterations in multiple cellular signaling pathways; indeed, the development and the progression of PCa are known to be caused by the deregulation of several selective signaling pathways such as the androgen receptor, Akt, nuclear factor-kappaB, Wnt, Hedgehog, and Notch. Therefore, strategies targeting these important pathways and their upstream and downstream signaling could be promising for the prevention of PCa progression. In this review, we summarize the current knowledge regarding the alterations in cell signaling pathways during the development and progression of PCa, and document compelling evidence showing that these are the targets of several natural agents against PCa progression and its metastases.
Collapse
Affiliation(s)
- Fazlul H Sarkar
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R Street, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
15
|
Barakat MT, Humke EW, Scott MP. Learning from Jekyll to control Hyde: Hedgehog signaling in development and cancer. Trends Mol Med 2010; 16:337-48. [PMID: 20696410 DOI: 10.1016/j.molmed.2010.05.003] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 05/13/2010] [Accepted: 05/17/2010] [Indexed: 02/07/2023]
Abstract
The Hedgehog (Hh) cascade controls cell proliferation, differentiation and patterning of tissues during embryogenesis but is largely suppressed in the adult. The Hh pathway can become reactivated in cancer. Here, we assimilate data from recent studies to understand how and when the Hh pathway is turned on to aid the neoplastic process. Hh signaling is now known to have a role in established tumors, enabling categorization of tumors based on the role Hh signaling plays in their growth. This categorization has relevance for prognosis and targeted therapeutics. In the first category, abnormal Hh signaling initiates the tumor. In the second category, Hh signaling helps maintain the tumor. In the third category, Hh signaling is implicated but its role is not yet defined.
Collapse
Affiliation(s)
- Monique T Barakat
- Department of Developmental Biology, Howard Hughes Medical Institute, Clark Center West W252, 318 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305-5439, USA
| | | | | |
Collapse
|
16
|
ten Haaf A, Bektas N, von Serenyi S, Losen I, Arweiler EC, Hartmann A, Knüchel R, Dahl E. Expression of the glioma-associated oncogene homolog (GLI) 1 in human breast cancer is associated with unfavourable overall survival. BMC Cancer 2009; 9:298. [PMID: 19706168 PMCID: PMC2753634 DOI: 10.1186/1471-2407-9-298] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Accepted: 08/25/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The transcription factor GLI1, a member of the GLI subfamily of Krüppel-like zinc finger proteins is involved in signal transduction within the hedgehog pathway. Aberrant hedgehog signalling has been implicated in the development of different human tumour entities such as colon and lung cancer and increased GLI1 expression has been found in these tumour entities as well. In this study we questioned whether GLI1 expression might also be important in human breast cancer development. Furthermore we correlated GLI1 expression with histopathological and clinical data to evaluate whether GLI1 could represent a new prognostic marker in breast cancer treatment. METHODS Applying semiquantitative realtime PCR analysis and immunohistochemistry (IHC) GLI1 expression was analysed in human invasive breast carcinomas (n = 229) in comparison to normal human breast tissues (n = 58). GLI1 mRNA expression was furthermore analysed in a set of normal (n = 3) and tumourous breast cell lines (n = 8). IHC data were statistically interpreted using SPSS version 14.0. RESULTS Initial analysis of GLI1 mRNA expression in a small cohort of (n = 5) human matched normal and tumourous breast tissues showed first tendency towards GLI1 overexpression in human breast cancers. However only a small sample number was included into these analyses and values for GLI1 overexpression were statistically not significant (P = 0.251, two-tailed Mann-Whitney U-test). On protein level, nuclear GLI1 expression in breast cancer cells was clearly more abundant than in normal breast epithelial cells (P = 0.008, two-tailed Mann-Whitney U-test) and increased expression of GLI1 protein in breast tumours significantly correlated with unfavourable overall survival (P = 0.019), but also with higher tumour stage (P < 0.001) and an increased number of tumour-positive axillar lymph nodes (P = 0.027). Interestingly, a highly significant correlation was found between GLI1 expression and the expression of SHH, a central upstream molecule of the hedgehog pathway that was previously analysed on the same tissue microarray. CONCLUSION Our study presents a systematic expression analysis of GLI1 in human breast cancer. Elevated levels of GLI1 protein in human breast cancer are associated with unfavourable prognosis and progressive stages of disease. Thus GLI1 protein expression measured e.g. by an IHC based scoring system might have an implication in future multi-marker panels for human breast cancer prognosis or molecular sub typing. The highly significant correlation between SHH and GLI1 expression characterises GLI1 as a potential functional downstream target of the hedgehog signalling pathway in human breast cancer as well. Furthermore, our study indicates that altered hedgehog signalling may represent a key disease pathway in the progression of human breast cancer.
Collapse
Affiliation(s)
- Anette ten Haaf
- Molecular Oncology Group, Institute of Pathology, University Hospital of the RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Nuran Bektas
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Sonja von Serenyi
- Molecular Oncology Group, Institute of Pathology, University Hospital of the RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Inge Losen
- Molecular Oncology Group, Institute of Pathology, University Hospital of the RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Elfriede Christel Arweiler
- Institute of Medical Statistics, University Hospital of the RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Krankenhausstrasse 12, 91054 Erlangen, Germany
| | - Ruth Knüchel
- Molecular Oncology Group, Institute of Pathology, University Hospital of the RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Edgar Dahl
- Molecular Oncology Group, Institute of Pathology, University Hospital of the RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
17
|
Laner-Plamberger S, Kaser A, Paulischta M, Hauser-Kronberger C, Eichberger T, Frischauf AM. Cooperation between GLI and JUN enhances transcription of JUN and selected GLI target genes. Oncogene 2009; 28:1639-51. [PMID: 19219074 DOI: 10.1038/onc.2009.10] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sustained Hedgehog (HH) signaling is implicated in basal cell carcinoma of the skin and other types of cancer. Here we show that GLI1 and GLI2, the main transcriptional activators of the HH pathway, directly regulate expression of the activator protein 1 (AP-1) family member JUN, a transcription factor controlling keratinocyte proliferation and skin homeostasis. Activation of the JUN promoter by GLI is dependent on a GLI-binding site and the AP-1 sites known to be involved in self-activation of JUN. Transcription of JUN is greatly enhanced in the presence of GLI and requires activated JUN protein. GLI2act is a more potent activator than GLI1 in these experiments and physical interaction with phosphorylated JUN was only detected for GLI2act. The synergistic effect of GLI and JUN extends to the activation of further GLI target genes as shown by shRNA-mediated knockdown of JUN in human keratinocytes. Some of these cooperatively activated genes are involved in cell-cycle progression, which is consistent with a significant reduction of the proliferative potential of GLI in the absence of JUN. These results suggest a novel connection between HH/GLI pathway activity and JUN, which may contribute to the oncogenic activity of HH/GLI signaling in skin.
Collapse
Affiliation(s)
- S Laner-Plamberger
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | | | | | | | | |
Collapse
|