1
|
Hsu JY, Lin YS, Huang LH, Tsao TY, Hsu CY, Ou YC, Tung MC. Concurrent occurrence of adenocarcinoma and urothelial carcinoma of the prostate gland: A case report. World J Clin Cases 2024; 12:5952-5959. [PMID: 39286382 PMCID: PMC11287516 DOI: 10.12998/wjcc.v12.i26.5952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/15/2024] [Accepted: 07/15/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Adenocarcinoma is the most common subtype of prostate cancer. Prostatic urothelial carcinoma (UC) typically originates from the prostatic urethra. The concurrent occurrence of adenocarcinoma and UC of the prostate gland is uncommon. CASE SUMMARY We present the case of an 82-year-old male patient with simultaneous adenocarcinoma and UC of the prostate gland. The patient underwent a transrectal ultrasound-guided biopsy, and the pathology test revealed UC. Subsequently, transurethral laser prostatectomy was performed, and the pathology test indicated adenocarcinoma of the prostate with a Gleason score of 3 + 4 and high-grade UC. Therefore, the patient was treated with androgen deprivation therapy, systemic chemotherapy, and immunotherapy. Magnetic resonance imaging performed during follow-up revealed a prostate tumor classified as cT2cN1M0, stage IVA. Therefore, the patient underwent robotic-assisted radical prostatectomy and bilateral pelvic lymph node dissection. The final pathology test of the prostate gland revealed acinar-type adenocarcinoma, Gleason pattern 4 + 3, pT2N0M0, and high-grade UC. The patient regularly presented to the clinic for postoperative follow-up evaluations. He did not experience any urinary discomfort. CONCLUSION According to our literature review, this is the first reported case of coexisting adenocarcinoma and UC of the prostate gland.
Collapse
Affiliation(s)
- Jhe Yuan Hsu
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Yi Sheng Lin
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Li Hua Huang
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Tang Yi Tsao
- Department of Pathology, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Chao Yu Hsu
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Yen Chuan Ou
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Min Che Tung
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| |
Collapse
|
2
|
Verma P, Shukla N, Kumari S, Ansari M, Gautam NK, Patel GK. Cancer stem cell in prostate cancer progression, metastasis and therapy resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188887. [PMID: 36997008 DOI: 10.1016/j.bbcan.2023.188887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/18/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
Prostate cancer (PCa) is the most diagnosed malignancy in the men worldwide. Cancer stem cells (CSCs) are the sub-population of cells present in the tumor which possess unique properties of self-renewal and multilineage differentiation thus thought to be major cause of therapy resistance, disease relapse, and mortality in several malignancies including PCa. CSCs have also been shown positive for the common stem cells markers such as ALDH EZH2, OCT4, SOX2, c-MYC, Nanog etc. Therefore, isolation and characterization of CSCs specific markers which may discriminate CSCs and normal stem cells are critical to selectively eliminate CSCs. Rapid advances in the field offers a theoretical explanation for many of the enduring uncertainties encompassing the etiology and an optimism for the identification of new stem-cell targets, development of reliable and efficient therapies in the future. The emerging reports have also provided unprecedented insights into CSCs plasticity, quiescence, renewal, and therapeutic response. In this review, we discuss the identification of PCa stem cells, their unique properties, stemness-driving pathways, new diagnostics, and therapeutic interventions.
Collapse
|
3
|
Liu Q, Li Q, Zhu S, Yi Y, Cao Q. B lymphoma Moloney murine leukemia virus insertion region 1: An oncogenic mediator in prostate cancer. Asian J Androl 2020; 21:224-232. [PMID: 29862993 PMCID: PMC6498728 DOI: 10.4103/aja.aja_38_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
B lymphoma Moloney murine leukemia virus insertion region 1 (BMI1), a core member of polycomb repressive complex 1 (PRC1), has been intensely investigated in the field of cancer epigenetics for decades. Widely known as a critical regulator in cellular physiology, BMI1 is essential in self-renewal and differentiation in different lineages of stem cells. BMI1 also plays a significant role in cancer etiology for its involvement in pathological progress such as epithelial–mesenchymal transition (EMT) and cancer stem cell maintenance, propagation, and differentiation. Importantly, overexpression of BMI1 is predictive for drug resistance, tumor recurrence, and eventual therapy failure of various cancer subtypes, which renders the pharmacological targeting at BMI1 as a novel and promising therapeutic approach. The study on prostate cancer, a prevalent hormone-related cancer among men, has promoted enormous research advancements in cancer genetics and epigenetics. This review summarizes the role of BMI1 as an oncogenic and epigenetic regulator in tumor initiation, progression, and relapse of prostate cancer.
Collapse
Affiliation(s)
- Qipeng Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Qiaqia Li
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Xiangya School of Medicine, Central South University, Changsha 410008, China
| | - Sen Zhu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yang Yi
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qi Cao
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA.,Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
4
|
Asadzadeh Z, Mansoori B, Mohammadi A, Aghajani M, Haji‐Asgarzadeh K, Safarzadeh E, Mokhtarzadeh A, Duijf PHG, Baradaran B. microRNAs in cancer stem cells: Biology, pathways, and therapeutic opportunities. J Cell Physiol 2018; 234:10002-10017. [DOI: 10.1002/jcp.27885] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Marjan Aghajani
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | | | - Elham Safarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Microbiology & Immunology Faculty of Medicine, Ardabil University of Medical Sciences Ardabil Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Pascal H. G. Duijf
- Translational Research Institute, University of Queensland Diamantina Institute, The University of Queensland Brisbane Queensland Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
5
|
Liao CP, Lin TP, Li PC, Geary LA, Chen K, Vaikari VP, Wu JB, Lin CH, Gross ME, Shih JC. Loss of MAOA in epithelia inhibits adenocarcinoma development, cell proliferation and cancer stem cells in prostate. Oncogene 2018; 37:5175-5190. [PMID: 29844571 DOI: 10.1038/s41388-018-0325-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/31/2018] [Accepted: 04/26/2018] [Indexed: 12/17/2022]
Abstract
Monoamine oxidase A (MAOA) is a mitochondrial enzyme, which degrades monoamine neurotransmitters and dietary amines and produces H2O2. Recent studies have shown increased MAOA expression in prostate cancer (PCa), glioma, and classical Hodgkin lymphoma. However, the biological function of MAOA in cancer development remains unknown. In this study, we investigated the role of MAOA in the development of prostate adenocarcinoma by creating a prostate-specific Pten/MAOA knockout (KO) mouse model, in which MAOA-floxP mouse was crossed with the conditional Pten KO PCa mouse that develops invasive PCa. In contrast to Pten KO mice, age-matched Pten/MAOA KO mice exhibited a significant decrease in both prostate size and the incidence of invasive cancer. We observed a significant decline in AKT phosphorylation and Ki67 expression in Pten/MAOA KO mice, which reduced epithelial cell growth and proliferation. As cancer stem cells (CSCs) are required for tumor initiation and growth, we investigated expression of OCT4 and NANOG in the setting of decreased MAOA expression. We found that both OCT4 and NANOG were significantly attenuated in the prostate epithelia of Pten/MAOA KO mice compared to Pten KO mice, which was confirmed with targeted knockdown of MAOA with a short-hairpin(sh) vector targeting MAOA compared to cells transfected with a control vector. Expression of other markers associated with the a stem cell phenotype, including CD44, α2β1, and CD133 as well as HIF-1α+CD44+ stem cells were all decreased in shMAOA PCa cells compared with empty vector-transfected control cells. We also found spheroid formation ability in PCa cells was decreased when endogenous MAOA was suppressed by siRNA or MAOA inhibitor clorgyline in a colony formation assay. Using the TCGA database, elevated MAOA expression was associated with reduced Pten levels in high Gleason grade in patient samples. Further, we found that Pten-positive PCa cells were more resistant to clorgyline treatments than Pten-null cells in tumorigenicity and stemness. Taken together, these studies suggest that MAOA expression promotes PCa development by increasing cell proliferation and CSCs and highlights the potential use of MAOA inhibitors for the treatment of PCa.
Collapse
Affiliation(s)
- Chun-Peng Liao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA.,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA.,Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033-9075, USA
| | - Tzu-Ping Lin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA.,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, WA, 11221, Taiwan
| | - Pei-Chuan Li
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA.,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Lauren A Geary
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA.,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Kevin Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA.,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Vijaya Pooja Vaikari
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA.,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA
| | - Jason Boyang Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, 99210-1495, USA
| | - Chi-Hung Lin
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, WA, 11221, Taiwan
| | - Mitchell E Gross
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033-9075, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, 90089-9176, CA, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA. .,USC-Taiwan Center for Translation Research, University of Southern California, Los Angeles, CA, 90089-9121, USA. .,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, 90089-9176, CA, USA. .,Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089-9037, USA. .,College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
6
|
Kwon OJ, Zhang B, Zhang L, Xin L. High fat diet promotes prostatic basal-to-luminal differentiation and accelerates initiation of prostate epithelial hyperplasia originated from basal cells. Stem Cell Res 2016; 16:682-91. [PMID: 27107344 DOI: 10.1016/j.scr.2016.04.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 01/03/2023] Open
Abstract
Recent lineage tracing studies showed that the prostate basal and luminal cells in adult mice are two independent lineages under the physiological condition, but basal cells are capable of generating luminal progenies during bacterial infection-induced prostatitis. Because acute bacterial infection in human prostate tissues is relatively rare, the disease relevance of the bacterial infection-induced basal-to-luminal differentiation is uncertain. Herein we employ a high fat diet-induced sterile prostate inflammation model to determine whether basal-to-luminal differentiation can be induced by inflammation irrespective of the underlying etiologies. A K14-CreER model and a fluorescent report line are utilized to specifically label basal cells with the green fluorescent protein. We show that high fat diet promotes immune cell infiltration into the prostate tissues and basal-to-luminal differentiation. Increased cell proliferation accompanies basal-to-luminal differentiation, suggesting a concurrent regulation of basal cell proliferation and differentiation. This study demonstrates that basal-to-luminal differentiation can be induced by different types of prostate inflammation evolved with distinct etiologies. Finally, high fat diet also accelerates initiation and progression of prostatic intraepithelial neoplasia that are originated from basal cells with loss-of-function of the tumor suppressor Pten. Because prostate cancer originated from basal cells tends to be invasive, our study also provides an alternative explanation for the association between obesity and aggressive prostate cancer.
Collapse
Affiliation(s)
- Oh-Joon Kwon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Boyu Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Li Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States
| | - Li Xin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, United States; Dan L. Duncan Cancer Center, Baylor College of Medicine, United States; Department of Pathology and Immunology.
| |
Collapse
|
7
|
Li Y, Sarkar FH. Role of BioResponse 3,3'-Diindolylmethane in the Treatment of Human Prostate Cancer: Clinical Experience. Med Princ Pract 2015; 25 Suppl 2:11-7. [PMID: 26501150 PMCID: PMC4848191 DOI: 10.1159/000439307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/11/2015] [Indexed: 01/09/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) progression after androgen deprivation therapy shows upregulated expression of androgen receptor (AR) splice variants, induced epithelial-to-mesenchymal transition phenotypes and enhanced stem cell characteristics, all of which are associated with resistance to enzalutamide. Since there is no curative treatment for CRPC, innovative treatments are urgently needed. In our recent study, we found that resistance to enzalutamide was partly due to deregulated expression of microRNAs such as miR-34a, miR-124, miR-27b, miR-320 and let-7, which play important roles in regulating AR and stem cell marker gene expression that appears to be linked with resistance to enzalutamide. Importantly, we found that BioResponse 3,3'-diindolylmethane (BR-DIM) treatment in vitro and in vivo caused downregulation in the expression of wild-type AR. The AR splice variants, Lin28B and EZH2, appear to be deregulated through the re-expression of let-7, miR-27b, miR-320 and miR-34a in human prostate cancer (PCa). BR-DIM administered in clinical trials was well tolerated, and 93% of patients had detectable prostatic DIM levels. The inhibitory effects of BR-DIM on AR and AR target gene such as prostate-specific antigen were also observed in the clinical trial. Our preclinical and clinical studies provide the scientific basis for a 'proof-of-concept' clinical trial in CRPC patients treated with enzalutamide in combination with BR-DIM. This strategy could be expanded in future clinical trials in patients with PCa to determine whether or not they could achieve a better treatment outcome which could be partly mediated by delaying or preventing the development of CRPC.
Collapse
Affiliation(s)
- Yiwei Li
- Department of University School of Medicine, Detroit, Mich., USA
| | - Fazlul H. Sarkar
- Department of University School of Medicine, Detroit, Mich., USA
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Mich., USA
| |
Collapse
|
8
|
Mayer MJ, Klotz LH, Venkateswaran V. Metformin and prostate cancer stem cells: a novel therapeutic target. Prostate Cancer Prostatic Dis 2015. [PMID: 26215782 DOI: 10.1038/pcan.2015.35] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prostate cancer is the second most frequently diagnosed cancer in the world. Localized disease can be effectively treated with radiation therapy or radical prostatectomy. However, advanced prostate cancer is more difficult to treat and if metastatic, is incurable. There is a need for more effective therapy for advanced prostate cancer. One potential target is the cancer stem cell (CSC). CSCs have been described in several solid tumors, including prostate cancer, and contribute to therapeutic resistance and tumor recurrence. Metformin, a common oral biguanide used to treat type 2 diabetes, has been demonstrated to have anti-neoplastic effects. Specifically, metformin targets CSCs in breast cancer, pancreatic cancer, glioblastoma and colon cancer. Metformin acts directly on the mitochondria to inhibit oxidative phosphorylation and reduce mitochondrial ATP production. This forces tumor cells to compensate by increasing the rate of glycolysis. CSCs rely heavily on mitochondrial oxidative phosphorylation for energy production. The glycolytic switch results in an energy crisis in these cells. Metformin could be used to exploit this metabolic weakness in CSCs. This would increase CSC sensitivity to conventional cancer therapies, circumventing treatment resistance and enhancing treatment efficacy. This review will explore the characteristics of prostate CSCs, their role in tumor propagation and therapeutic resistance and the role of metformin as a potential prostate CSC sensitizer to current anticancer therapies.
Collapse
Affiliation(s)
- M J Mayer
- Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - L H Klotz
- Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - V Venkateswaran
- Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Belair CD, Paikari A, Moltzahn F, Shenoy A, Yau C, Dall'Era M, Simko J, Benz C, Blelloch R. DGCR8 is essential for tumor progression following PTEN loss in the prostate. EMBO Rep 2015. [PMID: 26206718 DOI: 10.15252/embr.201439925] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In human prostate cancer, the microRNA biogenesis machinery increases with prostate cancer progression. Here, we show that deletion of the Dgcr8 gene, a critical component of this complex, inhibits tumor progression in a Pten-knockout mouse model of prostate cancer. Early stages of tumor development were unaffected, but progression to advanced prostatic intraepithelial neoplasia was severely inhibited. Dgcr8 loss blocked Pten null-induced expansion of the basal-like, but not luminal, cellular compartment. Furthermore, while late-stage Pten knockout tumors exhibit decreased senescence-associated beta-galactosidase activity and increased proliferation, the simultaneous deletion of Dgcr8 blocked these changes resulting in levels similar to wild type. Sequencing of small RNAs in isolated epithelial cells uncovered numerous miRNA changes associated with PTEN loss. Consistent with a Pten-Dgcr8 association, analysis of a large cohort of human prostate tumors shows a strong correlation between Akt activation and increased Dgcr8 mRNA levels. Together, these findings uncover a critical role for microRNAs in enhancing proliferation and enabling the expansion of the basal cell compartment associated with tumor progression following Pten loss.
Collapse
Affiliation(s)
- Cassandra D Belair
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California - San Francisco, San Francisco, CA, USA Center for Reproductive Sciences, University of California - San Francisco, San Francisco, CA, USA Department of Urology, University of California - San Francisco, San Francisco, CA, USA
| | - Alireza Paikari
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California - San Francisco, San Francisco, CA, USA Center for Reproductive Sciences, University of California - San Francisco, San Francisco, CA, USA
| | - Felix Moltzahn
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California - San Francisco, San Francisco, CA, USA Department of Urology, University of California - San Francisco, San Francisco, CA, USA
| | - Archana Shenoy
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California - San Francisco, San Francisco, CA, USA Department of Urology, University of California - San Francisco, San Francisco, CA, USA
| | - Christina Yau
- Department of Medicine, University of California - San Francisco, San Francisco, CA, USA Buck Institute for Research on Aging, Novato, CA, USA
| | - Marc Dall'Era
- Department of Urology, University of California - San Francisco, San Francisco, CA, USA
| | - Jeff Simko
- Department of Urology, University of California - San Francisco, San Francisco, CA, USA Department of Anatomic Pathology, University of California - San Francisco, San Francisco, CA, USA
| | - Christopher Benz
- Department of Medicine, University of California - San Francisco, San Francisco, CA, USA Buck Institute for Research on Aging, Novato, CA, USA
| | - Robert Blelloch
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California - San Francisco, San Francisco, CA, USA Center for Reproductive Sciences, University of California - San Francisco, San Francisco, CA, USA Department of Urology, University of California - San Francisco, San Francisco, CA, USA Department of Anatomic Pathology, University of California - San Francisco, San Francisco, CA, USA
| |
Collapse
|
10
|
Fang X, Gyabaah K, Nickkholgh B, Cline JM, Balaji K. Novel In Vivo model for combinatorial fluorescence labeling in mouse prostate. Prostate 2015; 75:988-1000. [PMID: 25753731 PMCID: PMC4515139 DOI: 10.1002/pros.22984] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/22/2015] [Indexed: 01/26/2023]
Abstract
BACKGROUND The epithelial layer of prostate glands contains several types of cells, including luminal and basal cells. Yet there is paucity of animal models to study the cellular origin of normal or neoplastic development in the prostate to facilitate the treatment of heterogenous prostate diseases by targeting individual cell lineages. METHODS We developed a mouse model that expresses different types of fluorescent proteins (XFPs) specifically in prostatic cells. Using an in vivo stochastic fluorescent protein combinatorial strategy, XFP signals were expressed specifically in prostate of Protein Kinase D1 (PKD1) knock-out, K-Ras(G) (12) (D) knock-in, and Phosphatase and tensin homolog (PTEN) and PKD1 double knock-out mice under the control of PB-Cre promoter. RESULTS In vivo XFP signals were observed in prostate of PKD1 knock-out, K-Ras(G) (12) (D) knock-in, and PTEN PKD1 double knock-out mice, which developed normal, hyperplastic, and neoplastic prostate, respectively. The patchy expression pattern of XFPs in neoplasia tissue indicated the clonal origin of cancer cells in the prostate. CONCLUSIONS The transgenic mouse models demonstrate combinatorial fluorescent protein expression in normal and cancerous prostatic tissues. This novel prostate-specific fluorescent labeled mouse model, which we named Prorainbow, could be useful in studying benign and malignant pathology of prostate.
Collapse
Affiliation(s)
- Xiaolan Fang
- Department of Cancer Biology, Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, North Carolina
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Kenneth Gyabaah
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Bita Nickkholgh
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - J. Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - K.C. Balaji
- Department of Cancer Biology, Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, North Carolina
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
- Department of Urology, Wake Forest University Health Sciences, Winston-Salem, North Carolina
- W. G. (Bill) Hefner Veterans Administration Medical Center, Salisbury, North Carolina
- Correspondence to: K. C. Balaji, Department of Urology, Cancer Biology and Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157.
| |
Collapse
|
11
|
Uygur B, Abramo K, Leikina E, Vary C, Liaw L, Wu WS. SLUG is a direct transcriptional repressor of PTEN tumor suppressor. Prostate 2015; 75:907-16. [PMID: 25728608 PMCID: PMC4654464 DOI: 10.1002/pros.22974] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/08/2015] [Indexed: 11/06/2022]
Abstract
BACKGROUND PTEN/AKT signaling plays a key role in prostate cancer development and maintenance of prostate cancer stem cells. How other oncogenes or tumor suppressors interact with this pathway remain to be elucidated. SLUG is an zinc finger transcription factor of the Snail superfamily, and it promotes cancer metastasis and determines the mammary stem cell state. METHODS SLUG was overexpressed in cells by retroviral vector and knockdown of SLUG and PTEN was mediated by shRNAs-expressing lentiviruses. Expression level of SLUG and PTEN was examined by Western blot, RT-PCR, and qPCR analyses. PTEN promoter activity was measured by luciferase reporter assay. ChIP assay was used to measure the binding between SLUG and the PTEN promoter in vivo. RESULT We showed that overexpression of SLUG decreased expression of PTEN tumor repressor in prostate cancer cell lines 22RV1 and DU145; conversely, knockdown of SLUG expression elevated PTEN expresson at both protein and RNA level in these cells. We demonstrated that SLUG overexpression inhibits PTEN promoter activity through the proximal promoter region in prostate cancer cells. By ChIP assay, we confirmed that SLUG directly binds to the PTEN promoter region covering the E-box sites. We also showed that Slug deficiency leads to an increased expression of PTEN in mouse embryo fibroblasts and prostate tissues. Importantly, we found that overexpression of SLUG increases drug resistance of DU145 prostate cancer cell line and knockdown of SLUG by shRNA sensitizes DU145 cell line to chemotherapeutic drugs. We further demonstrated that PTEN knockdown converts drug sensitivity of DU145 cells expressing SLUG shRNA to anticancer drugs. CONCLUSION We provide compelling evidence showing that PTEN is a direct functional target of SLUG. Our findings offer new insight in the regulation of the PTEN/AKT pathway and provide a molecular basis for potential targeted therapies of prostate cancer Prostate 75:907-916, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- HEK293 Cells
- Humans
- Immunohistochemistry
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- PTEN Phosphohydrolase/antagonists & inhibitors
- PTEN Phosphohydrolase/biosynthesis
- PTEN Phosphohydrolase/genetics
- Promoter Regions, Genetic
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- RNA, Neoplasm/chemistry
- RNA, Neoplasm/genetics
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Snail Family Transcription Factors
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Berna Uygur
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
- Program in Biochemistry and Molecular Biology, University of Maine, Orono, Maine, USA
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Katrina Abramo
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Evgenia Leikina
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Calvin Vary
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Lucy Liaw
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Wen-Shu Wu
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
- Department of Medicine, University of Illinois College of Medicine at Chicago, Chicago, Illinois, USA
- Correspondence to: Wen-Shu Wu, Department of Medicine, University of Illinois College of Medicine at Chicago, Chicago, IL 60612, USA. Tel: 312-996-2586; Fax: 011-312-413-4131;
| |
Collapse
|
12
|
Lim ML, Ooi BNS, Jungebluth P, Sjöqvist S, Hultman I, Lemon G, Gustafsson Y, Asmundsson J, Baiguera S, Douagi I, Gilevich I, Popova A, Haag JC, Rodríguez AB, Lim J, Liedén A, Nordenskjöld M, Alici E, Baker D, Unger C, Luedde T, Vassiliev I, Inzunza J, Ährlund-Richter L, Macchiarini P. Characterization of stem-like cells in mucoepidermoid tracheal paediatric tumor. PLoS One 2014; 9:e107712. [PMID: 25229469 PMCID: PMC4167860 DOI: 10.1371/journal.pone.0107712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 08/14/2014] [Indexed: 02/06/2023] Open
Abstract
Stem cells contribute to regeneration of tissues and organs. Cells with stem cell-like properties have been identified in tumors from a variety of origins, but to our knowledge there are yet no reports on tumor-related stem cells in the human upper respiratory tract. In the present study, we show that a tracheal mucoepidermoid tumor biopsy obtained from a 6 year-old patient contained a subpopulation of cells with morphology, clonogenicity and surface markers that overlapped with bone marrow mesenchymal stromal cells (BM-MSCs). These cells, designated as MEi (mesenchymal stem cell-like mucoepidermoid tumor) cells, could be differentiated towards mesenchymal lineages both with and without induction, and formed spheroids in vitro. The MEi cells shared several multipotent characteristics with BM-MSCs. However, they displayed differences to BM-MSCs in growth kinectics and gene expression profiles relating to cancer pathways and tube development. Despite this, the MEi cells did not possess in vivo tumor-initiating capacity, as proven by the absence of growth in situ after localized injection in immunocompromised mice. Our results provide an initial characterization of benign tracheal cancer-derived niche cells. We believe that this report could be of importance to further understand tracheal cancer initiation and progression as well as therapeutic development.
Collapse
Affiliation(s)
- Mei Ling Lim
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | | | - Philipp Jungebluth
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Sjöqvist
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Isabell Hultman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Greg Lemon
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Ylva Gustafsson
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Jurate Asmundsson
- Department of Oncology and Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Silvia Baiguera
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Iyadh Douagi
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Irina Gilevich
- International Scientific-Research Clinical and Educational Center of Regenerative Medicine, Kuban State Medical University, Krasnodar, Russian Federation
| | - Alina Popova
- International Scientific-Research Clinical and Educational Center of Regenerative Medicine, Kuban State Medical University, Krasnodar, Russian Federation
| | - Johannes Cornelius Haag
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Beltrán Rodríguez
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Jianri Lim
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| | - Agne Liedén
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Nordenskjöld
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Duncan Baker
- Department of Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Christian Unger
- Department of Biomedical Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Tom Luedde
- Department of Medicine III, University Hospital RWTH Aachen, Germany
| | - Ivan Vassiliev
- Robinson Institute, Center for Stem Cell Research, The University of Adelaide, Adelaide, Australia
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Lars Ährlund-Richter
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Macchiarini
- Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Division of Ear, Nose, Throat, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Geary LA, Nash KA, Adisetiyo H, Liang M, Liao CP, Jeong JH, Zandi E, Roy-Burman P. CAF-secreted annexin A1 induces prostate cancer cells to gain stem cell-like features. Mol Cancer Res 2014; 12:607-21. [PMID: 24464914 DOI: 10.1158/1541-7786.mcr-13-0469] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Annexin A1 (AnxA1), a phospholipid-binding protein and regulator of glucocorticoid-induced inflammatory signaling, has implications in cancer. Here, a role for AnxA1 in prostate adenocarcinoma was determined using primary cultures and a tumor cell line (cE1), all derived from the conditional Pten deletion mouse model of prostate cancer. AnxA1 secretion by prostate-derived cancer-associated fibroblasts (CAF) was significantly higher than by normal prostate fibroblasts (NPF). Prostate tumor cells were sorted to enrich for epithelial subpopulations based on nonhematopoietic lineage, high SCA-1, and high or medium levels of CD49f. Compared with controls, AnxA1 enhanced stem cell-like properties in high- and medium-expression subpopulations of sorted cE1 and primary cells, in vitro, through formation of greater number of spheroids with increased complexity, and in vivo, through generation of more, larger, and histologically complex glandular structures, along with increased expression of p63, a basal/progenitor marker. The differentiated medium-expression subpopulations from cE1 and primary cells were most susceptible to gain stem cell-like properties as shown by increased spheroid and glandular formation. Further supporting this increased plasticity, AnxA1 was shown to regulate epithelial-to-mesenchymal transition in cE1 cells. These results suggest that CAF-secreted AnxA1 contributes to tumor stem cell dynamics via two separate but complementary pathways: induction of a dedifferentiation process leading to generation of stem-like cells from a subpopulation of cancer epithelial cells and stimulation of proliferation and differentiation of the cancer stem-like cells. IMPLICATIONS AnxA1 participates in a paradigm in which malignant prostate epithelial cells that are not cancer stem cells are induced to gain cancer stem cell-like properties.
Collapse
Affiliation(s)
- Lauren A Geary
- Department of Pathology, University of Southern California, Keck School of Medicine, 2011 Zonal Avenue, HMR 210B, Los Angeles, CA 90033.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Mouse models of prostate cancer (PCa) are critical for understanding the biology of PCa initiation, progression, and treatment modalities. Here, we summarize recent advances in PCa mouse models that led to new insights into specific gene functions in PCa. For example, the study of transgenic mice with TMPRSS2/ERG, an androgen-regulated fusion protein, revealed its role in developing PCa precursor lesions, prostate intraepithelial neoplasia; however, it is not sufficient for PCa development. Double deficiency of Pten and Smad4 leads to a high incidence of metastatic PCa. Targeted deletion of Pten in castration-resistant Nkx3-1-expressing cells results in rapid carcinoma formation after androgen-mediated regeneration, indicating that progenitor cells with luminal characteristics can play a role in initiation of PCa. Transgenic mice with activated oncogenes, growth factors, and steroid hormone receptors or inactivated tumor suppressors continue to provide insights into disease progression from initiation to metastasis. Further development of new PCa models with spatial and temporal regulation of candidate gene expression will probably enhance our understanding of the complex events that lead to PCa initiation and progression, thereby invoking novel strategies to combat this common disease in men.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Pathology, New York University School of Medicine, New York, NY
| | | | - Pradip Roy-Burman
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Peng Lee
- Department of Pathology, New York University School of Medicine, New York, NY
- Department of Urology, New York University School of Medicine, New York, NY
- NYU Cancer Institute, New York University School of Medicine, New York, NY
- New York Harbor Healthcare System, New York, NY
| | - Zoran Culig
- Department of Urology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
15
|
The Functional Role of DAB2IP, a Homeostatic Factor, in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
16
|
Xu X, Liu RF, Zhang X, Huang LY, Chen F, Fei QL, Han ZG. DLK1 as a potential target against cancer stem/progenitor cells of hepatocellular carcinoma. Mol Cancer Ther 2012; 11:629-38. [PMID: 22238367 DOI: 10.1158/1535-7163.mct-11-0531] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Delta-like 1 homolog (DLK1; Drosophila) is a hepatic stem/progenitor cell marker in fetal livers that plays a vital role in oncogenesis of hepatocellular carcinoma (HCC). The aim of this study is to investigate whether DLK1 could serve as a potential therapeutic target against cancer stem/progenitor cells of HCC. DLK1(+) and DLK1(-) cells were sorted by fluorescence-activated cell sorting and magnetic-activated cell sorting, respectively, and then were evaluated by flow cytometry. The biological behaviors of these isolated cells and those with DLK1 knockdown were assessed by growth curve, colony formation assay, spheroid colony formation, chemoresistance, and in vivo tumorigenicity. Adenovirus-mediated RNA interference was used to knockdown the endogenous DLK1. We found that DLK1(+) population was less than 10% in almost all 17 HCC cell lines examined. DLK1(+) HCC cells showed stronger ability of chemoresistance, colony formation, spheroid colony formation, and in vivo tumorigenicity compared with DLK1(-) cells. The DLK1(+) HCC cells could generate the progeny without DLK1 expression. Furthermore, DLK1 knockdown could suppress the ability of proliferation, colony formation, spheroid colony formation, and in vivo tumorigenicity of Hep3B and Huh-7 HCC cells. Our data suggested that DLK1(+) HCC cells have characteristics similar to those of cancer stem/progenitor cells. RNA interference against DLK1 can suppress the malignant behaviors of HCC cells, possibly through directly disrupting cancer stem/progenitor cells, which suggested that DLK1 could be a potential therapeutic target against the HCC stem/progenitor cells.
Collapse
Affiliation(s)
- Xiao Xu
- National Human Genome Center of Rui-Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Isolation of prostate epithelial cells with stem/progenitor characteristics may enable further evaluation of the hierarchy of prostate glandular development and malignant transformation. Prostate epithelial cells capable of sphere formation in semisolid cultures possess stem/progenitor cell characteristics. This is demonstrated by self-renewal (via indefinite passaging) and in vivo differentiation into prostate tubules with discreet basal and luminal layers. Here, we describe a method for isolating prostate stem/progenitor cells from human tissues via in vitro prostasphere formation. Prostate tissue regeneration using human prostaspheres is also described, enabling the differentiation potential of sphere-forming cells to be observed.
Collapse
|
18
|
Liao CP, Adisetiyo H, Liang M, Roy-Burman P. Cancer stem cells and microenvironment in prostate cancer progression. Discov Oncol 2011; 1:297-305. [PMID: 21761361 DOI: 10.1007/s12672-010-0051-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
For a study of interactions between the cancer-associated fibroblasts (CAFs) and the putative prostate cancer stem cells (CSCs), we used a conditional Pten deletion mouse model of prostatic adenocarcinoma to isolate both CAF cultures and CSC-enriched cell fractions from the primary tumors. The CSC subpopulation exhibited a collective phenotype of Lin(-)/SCA-1(hi)/CD49f(hi)/p63(hi)/CK5(hi)/AR(lo)/CK18(lo)/Survivin(hi)/Runx2(hi) and contained cells with the ability to both self-renew and differentiate into basal and luminal cells in vitro. The spheroids generated from the CSC-enriched subpopulation mimicked the glandular structures that could be produced from a similarly isolated cell fraction from the normal mouse prostate. The efficiency of spheroid formation was found to be influenced differentially by the nature of the fibroblasts that were co-cultured in the 3-D system. The growth and differentiation properties of the CSCs were significantly more enhanced by factors released from CAFs relative to normal prostate fibroblasts (NPFs). Additionally, increased commitment to differentiation to the luminal cell lineage was noted when CAFs were present. When CSCs admixed with either CAFs or NPFs were examined for formation of prostatic glandular structures in renal grafts in vivo, the lesions formed were generally more in numbers in the presence of CAFs than NPFs. Furthermore, lesions formed with CAFs often displayed tumor-like complex histopathology and contained increased numbers of proliferating cells. Taken together, the results suggested that the CAFs in the prostate tumor microenvironment can contribute to the biologic properties of the CSCs and by this account may play a major role in prostate tumorigenesis and progression. Thus, it would be important now to identify the paracrine and/or juxtacrine factors that are responsible for the stimulation of the cancer stem cells.
Collapse
Affiliation(s)
- Chun-Peng Liao
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, 2011 Zonal Avenue, Los Angeles, CA 90033, USA
| | | | | | | |
Collapse
|
19
|
Bae KM, Su Z, Frye C, McClellan S, Allan RW, Andrejewski JT, Kelley V, Jorgensen M, Steindler DA, Vieweg J, Siemann DW. Expression of pluripotent stem cell reprogramming factors by prostate tumor initiating cells. J Urol 2010; 183:2045-53. [PMID: 20303530 DOI: 10.1016/j.juro.2009.12.092] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Indexed: 10/19/2022]
Abstract
PURPOSE We identified a discrete population of stem cell-like tumor cells expressing 5 essential transcription factors required to reprogram pluripotency in prostate tumor cell lines and primary prostate cancer tissue. MATERIALS AND METHODS DU145 and PC3 human prostate cancer cell lines (ATCC), tumor tissue from patients with prostate cancer and normal prostate tissue were evaluated for the reprogramming factors OCT3/4 (Cell Signaling Technology), SOX2, Klf4 (Santa Cruz Biotechnology, Santa Cruz, California), Nanog (BioLegend) and c-Myc (Cell Signaling) by semiquantitative reverse transcriptase-polymerase chain reaction, histological and immunohistochemical analysis. Stem cell-like tumor cells were enriched by flow cytometric cell sorting using E-cadherin (R&D Systems) as a surface marker, and soft agar, spheroid and tumorigenicity assays to confirm cancer stem cell-like characteristics. RESULTS mRNA expression of transcription factors OCT3/4 and SOX2 highly correlated in primary prostate tumor tissue samples. The number of OCT3/4 or SOX2 expressing cells was significantly increased in prostate cancer tissue compared to that in normal prostate or benign prostate hyperplasia tissue (p <0.05). When isolated from the DU145 and PC3 prostate cancer cell lines by flow cytometry, stem cell-like tumor cells expressing high OCT3/4 and SOX2 levels showed high tumorigenicity in immunodeficient mice. In vivo growth of the parental DU145 and PC3 prostate cancer cell lines was inhibited by short hairpin RNA knockdown of OCT3/4 or SOX2. CONCLUSIONS Data suggest that prostate tumor cells expressing pluripotent stem cell transcription factors are highly tumorigenic. Identifying such cells and their importance in prostate cancer growth could provide opportunities for novel targeting strategies for prostate cancer therapy.
Collapse
Affiliation(s)
- Kyung-Mi Bae
- Department of Urology (Prostate Disease Center), College of Medicine and Flow Cytometry Core Facility, University of Florida, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Li X, Wang Y, Sharif-Afshar AR, Uwamariya C, Yi A, Ishii K, Hayward SW, Matusik RJ, Bhowmick NA. Urothelial transdifferentiation to prostate epithelia is mediated by paracrine TGF-beta signaling. Differentiation 2008; 77:95-102. [PMID: 19281768 DOI: 10.1016/j.diff.2008.09.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 07/18/2008] [Accepted: 07/23/2008] [Indexed: 02/08/2023]
Abstract
The embryonic urogenital sinus mesenchyme (UGM) induces prostate epithelial morphogenesis in development. The molecular signals that drive UGM-mediated prostatic induction have not been defined. We hypothesized that the TGF-beta signaling directed the prostatic induction. UGM from TGF-beta type II receptor stromal conditional knockout mice (Tgfbr2(fspKO)) or control mice (Tgfbr2(floxE2/floxE2)) was recombined with wild-type adult mice bladder urothelial cells. The resulting urothelium associated with Tgfbr2(floxE2/floxE2) UGM was instructively differentiated into prostatic epithelium, as expected. In contrast, the urothelium associated with Tgfbr2(fspKO) UGM permissively maintained the phenotype of bladder epithelial cells. Microarray analysis of UGM tissues suggested the down-regulation of multiple Wnt ligands and the up-regulation of the Wnt antagonist, Wif 1, by the Tgfbr2(fspKO) UGM compared with Tgfbr2(floxE2/floxE2) UGM. The overexpression of Wif-1 by wild-type UGM resulted in the inhibition of prostatic induction. These data suggest that the stromal TGF-beta activity mediated by paracrine Wnt is necessary for the induction of prostatic differentiation. As Wnt ligands mediate differentiation and maintain the stem cell phenotype, the contribution of mouse stem cells and somatic cells to prostatic epithelium in the tissue recombination models was tested. The directed differentiation of mouse embryonic stem cells by UGM is suggested by a threshold number of mouse stem cells required in prostatic differentiation. To determine the contribution of somatic cells, the adult bladder epithelial compartment was labeled with green-fluorescent vital dye (CMFDA) and the stem-like cells marked by bromodeoxyuridine (BrdU) label-retention. The resulting prostatic epithelia of the tissue recombinants maintained the CMFDA dye, suggesting minimal cell division. Thus, the UGM can induce endoderm-derived epithelia and stem cells to form prostate through a transdifferentiation mechanism that requires stromal TGF-beta signaling to mediate epithelial Wnt activity.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Urologic Surgery, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232-2765, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Klonisch T, Wiechec E, Hombach-Klonisch S, Ande SR, Wesselborg S, Schulze-Osthoff K, Los M. Cancer stem cell markers in common cancers - therapeutic implications. Trends Mol Med 2008; 14:450-60. [PMID: 18775674 DOI: 10.1016/j.molmed.2008.08.003] [Citation(s) in RCA: 288] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 08/01/2008] [Accepted: 08/01/2008] [Indexed: 12/17/2022]
Abstract
Rapid advances in the cancer stem cell (CSC) field have provided cause for optimism for the development of more reliable cancer therapies in the future. Strategies aimed at efficient targeting of CSCs are becoming important for monitoring the progress of cancer therapy and for evaluating new therapeutic approaches. Here, we characterize and compare the specific markers that have been found to be present on stem cells, cancer cells and CSCs in selected tissues (colon, breast, liver, pancreas and prostate). We then discuss future directions of this intriguing new research field in the context of new diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, R3E 0W3, MB, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
Korkaya H, Wicha MS. Selective targeting of cancer stem cells: a new concept in cancer therapeutics. BioDrugs 2008; 21:299-310. [PMID: 17896836 DOI: 10.2165/00063030-200721050-00002] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Although the concept of 'cancer stem cell' was first proposed more then a century ago, it has attracted a great deal of attention recently due to advances in stem cell biology, leading to the identification of these cells in a wide variety of human cancers. There is accumulating evidence that the resistance of cancer stem cells to many conventional therapies may account for the inability of these therapies to cure most metastatic cancers. The recent identification of stem cell markers and advances in stem cell biology have facilitated research in multiple aspects of cancer stem cell behavior. Stem cell subcomponents have now been identified in a number of human malignancies, including hematologic malignancies and tumors of the breast, prostate, brain, pancreas, head and neck, and colon. Furthermore, pathways that regulate self-renewal and cell fate in these systems are beginning to be elucidated. In addition to pathways such as Wnt, Notch and Hedgehog, known to regulate self-renewal of normal stem cells, tumor suppressor genes such as PTEN (phosphatase and tensin homolog on chromosome 10) and TP53 (tumor protein p53) have also been implicated in the regulation of cancer stem cell self-renewal. In cancer stem cells, these pathways are believed to be deregulated, leading to uncontrolled self-renewal of cancer stem cells which generate tumors that are resistant to conventional therapies. Current cancer therapeutics based on tumor regression may target and kill differentiated tumor cells, which compose the bulk of the tumor, while sparing the rare cancer stem cell population. The cancer stem cell model suggests that the design of new cancer therapeutics may require the targeting and elimination of cancer stem cells. Therefore, it is imperative to design new strategies based upon a better understanding of the signaling pathways that control aspects of self-renewal and survival in cancer stem cells in order to identify novel therapeutic targets in these cells.
Collapse
Affiliation(s)
- Hasan Korkaya
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA.
| | | |
Collapse
|
23
|
Tumor Stem Cells: How to Define Them and How to Find Them? Stem Cells 2008. [DOI: 10.1007/978-1-4020-8274-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
24
|
Abstract
Peter Nowell and David Hungerford's discovery of the Philadelphia chromosome facilitated many critical studies that have led to a paradigm shift in our understanding of cancer as a disease of stem cells. This Review focuses on the application of these concepts to investigation of the role of stem cells in prostate cancer initiation and progression. Major strides in the development of in vitro and in vivo assays have enabled identification and characterization of prostate stem cells as well as functional evaluation of the tumorigenic effects of prostate cancer-related genetic alterations.
Collapse
Affiliation(s)
- Devon A. Lawson
- Department of Microbiology, Immunology and Molecular Genetics, and
Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, and
Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| | - Owen N. Witte
- Department of Microbiology, Immunology and Molecular Genetics, and
Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, and
Howard Hughes Medical Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|
25
|
Lin VK, Wang SY, Vazquez DV, C Xu C, Zhang S, Tang L. Prostatic stromal cells derived from benign prostatic hyperplasia specimens possess stem cell like property. Prostate 2007; 67:1265-76. [PMID: 17597114 DOI: 10.1002/pros.20599] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The hyper-proliferative activity of stromal smooth muscle (SM) cells is believed to be responsible for the pathogenesis of benign prostatic hyperplasia (BPH). We have observed that those stromal cells can differentiate into unrelated specialized cells. We thus hypothesize that stromal cells derived from adults prostate specimens may contain adult stem cells. To test this hypothesis, human prostate stromal primary cultures were established and used for characterization of their stem cell properties. METHODS Immunoblotting, immunohistochemistry, RT-PCR, and tissue culture techniques were used to characterize the primary cultured human prostate-derived stromal cells for their stem cell and differentiation properties. The plasticity of these stromal cells was analyzed using cell culture and histology techniques. RESULTS Primary cultured prostate stromal cells from BPH patient possess polygonal and elongated fibroblast/myofibroblast cellular morphology. They are positive in CD30, CD34, CD44, NSE, CD133, Flt-1, stem cell factor (SCF), and neuron-specific enolase (NSE), but negative in C-Kit, stem cell antigen (SCA), SH2, CD11b. Expression of SM myogenic markers in these cells may be induced by sodium butyrate (NaBu) treatment. Induction to osteogenic and adipogenic differentiation in these cells is also evident. CONCLUSIONS Our study on primary stromal cells from BPH patients have yielded many interesting findings that these prostate stroma cells possess: (1) mesenchymal stem cell (MSC) markers; (2) strong proliferative potential; and (3) ability to differentiate or transdifferentiate to myogenic, adipogenic, and osteogenic lineages. These cell preparations may serve as a potential tool for studies in prostate adult stem cell research and the regulation of benign prostatic hyperplasia.
Collapse
Affiliation(s)
- Victor K Lin
- Department of Urology, The University of Texas, Southwestern Medical Center, Dallas, Texas 75390-9110, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Chaiswing L, Bourdeau-Heller JM, Zhong W, Oberley TD. Characterization of redox state of two human prostate carcinoma cell lines with different degrees of aggressiveness. Free Radic Biol Med 2007; 43:202-15. [PMID: 17603930 DOI: 10.1016/j.freeradbiomed.2007.03.031] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 03/31/2007] [Indexed: 11/26/2022]
Abstract
We characterized the redox profiles in two different human prostate carcinoma cell lines (LNCaP vs PC3) that are known to exhibit varying degrees of invasiveness/metastatic ability. We confirmed that PC3 cells were more invasive than LNCaP cells through an in vitro analysis. The present study documented higher 8-hydroxy-2'-deoxyguanosine levels in PC3 cells than in LNCaP cells. The levels of lipid peroxidation were higher in LNCaP cells than in PC3 cells. The reduced glutathione (GSH)/glutathione disulfide (GSSG) ratio increased to a greater extent during cell growth in PC3 cells than in LNCaP cells, whereas both reduced GSH and GSSG levels were higher in the medium of PC3 cells than in that of LNCaP cells. The levels of reactive oxygen (ROS) and reactive nitrogen species (RNS), both intracellularly and in the medium, were higher for LNCaP cells than for PC3 cells during cell growth. In addition, our results demonstrated higher ROS/RNS levels in LNCaP cells than in PC3 cells in S and G(2)/M phases of the cell cycle during logarithmic growth. Each cell type showed distinct cytotoxic responses to low-molecular-weight redox-modulating compounds. Our results document that human prostate cancer cell lines of varying degrees of aggressive behavior have distinct redox properties, findings that could lead to novel therapeutic interventions.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Pathology and Laboratory Medicine Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | | | | | | |
Collapse
|
27
|
Mimeault M, Johansson SL, Vankatraman G, Moore E, Henichart JP, Depreux P, Lin MF, Batra SK. Combined targeting of epidermal growth factor receptor and hedgehog signaling by gefitinib and cyclopamine cooperatively improves the cytotoxic effects of docetaxel on metastatic prostate cancer cells. Mol Cancer Ther 2007; 6:967-78. [PMID: 17363490 DOI: 10.1158/1535-7163.mct-06-0648] [Citation(s) in RCA: 319] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The epidermal growth factor receptor (EGFR) and hedgehog cascades provide a critical role in prostate cancer progression and contribute to the resistance to clinical therapies and disease relapse. Therefore, we evaluated, for the first time, the antiproliferative and cytotoxic effects induced by a combination of selective inhibitors of EGFR tyrosine kinase and smoothened hedgehog signaling element, gefitinib and cyclopamine, with a current chemotherapeutic drug used in the clinics, docetaxel, on some metastatic prostate cancer cell lines. Immunohistochemical analyses revealed that sonic hedgehog (SHH) expression was enhanced in 39% of primary prostatic adenocarcinomas (Gleason scores 4-10) compared with the corresponding normal tissues of the same prostate gland from 32 prostate cancer patients. The confocal microscopy and Western blot analyses have also indicated the high expression levels of SHH and EGFR in metastatic LNCaP, DU145, and PC3 cells. Moreover, the results revealed that the drugs, alone or in combination, at lower concentrations inhibited the growth of EGF plus SHH-stimulated and serum-stimulated androgen-responsive LNCaP-C33 and androgen-independent LNCaP-C81, DU145, and PC3 cells. Importantly, the combined docetaxel, gefitinib, and cyclopamine also caused a higher rate of apoptotic death of prostate cancer cells compared with individual agents. The cytotoxic effects induced by these drugs in PC3 cells seem to be mediated in part through the cellular ceramide production and activation of caspase cascades via a mitochondrial pathway and the release of cytochrome c into the cytosol. Additionally, the combined agents were more effective at suppressing the invasiveness of PC3 cells through Matrigel in vitro than the single drugs. These findings indicate that the combined use of inhibitors of EGF-EGFR and hedgehog signaling with docetaxel could represent a more promising strategy for treatment in patients with metastatic and androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, 985870 Nebraska Medical Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Lawson DA, Xin L, Lukacs RU, Cheng D, Witte ON. Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci U S A 2006; 104:181-6. [PMID: 17185413 PMCID: PMC1716155 DOI: 10.1073/pnas.0609684104] [Citation(s) in RCA: 302] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The ability to isolate prostate stem cells is essential to explore their role in prostate development and disease. In vitro prostate colony- and sphere-forming assays were used to quantitatively measure murine prostate stem/progenitor cell enrichment and self-renewal. Cell surface markers were screened for their ability to positively or negatively enrich for cells with enhanced growth potential in these assays. Immunohistochemical and FACS analyses demonstrate that specific cell surface markers can be used to discriminate prostate stromal (CD34(+)), luminal epithelial (CD24(+)CD49f(-)), basal epithelial (CD24(+)CD49f(+)), hematopoietic (CD45(+), Ter119(+)), and endothelial (CD31(+)) lineages. Sorting for cells with a CD45(-)CD31(-)Ter119(-)Sca-1(+)CD49f(+) antigenic profile results in a 60-fold enrichment for colony- and sphere-forming cells. These cells can self-renew and expand to form spheres for many generations and can differentiate to produce prostatic tubule structures containing both basal and luminal cells in vivo. These cells also localize to the basal cell layer within the region of the gland that is proximal to the urethra, which has been identified as the prostate stem cell niche. Prostate stem cells can be isolated to a purity of up to 1 in 35 by using this antigenic profile. The remarkable similarity in cell surface profile between prostate and mammary gland stem cells suggests these markers may be conserved among epithelial stem cell populations.
Collapse
Affiliation(s)
- Devon A. Lawson
- *Departments of Microbiology, Immunology, and Molecular Genetics and
| | - Li Xin
- *Departments of Microbiology, Immunology, and Molecular Genetics and
| | - Rita U. Lukacs
- *Departments of Microbiology, Immunology, and Molecular Genetics and
| | - Donghui Cheng
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095
| | - Owen N. Witte
- *Departments of Microbiology, Immunology, and Molecular Genetics and
- Molecular and Medical Pharmacology, David Geffen School of Medicine, and
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|