1
|
Ou Y, Chu GCY, Lyu J, Yin L, Lim A, Zhai N, Cui X, Lewis MS, Edderkaoui M, Pandol SJ, Wang R, Zhang Y. Overcoming Resistance in Prostate Cancer Therapy Using a DZ-Simvastatin Conjugate. Mol Pharm 2024; 21:873-882. [PMID: 38229228 PMCID: PMC11025579 DOI: 10.1021/acs.molpharmaceut.3c00993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Prostate cancer (PC), particularly its metastatic castration-resistant form (mCRPC), is a leading cause of cancer-related deaths among men in the Western world. Traditional systemic treatments, including hormonal therapy and chemotherapy, offer limited effectiveness due to tumors' inherent resistance to these therapies. Moreover, they often come with significant side effects. We have developed a delivery method using a tumor-cell-specific heptamethine carbocyanine dye (DZ) designed to transport therapeutic agents directly to tumor cells. This research evaluated simvastatin (SIM) as the antitumor payload because of its demonstrated chemopreventive effects on human cancers and its well-documented safety profile. We designed and synthesized a DZ-SIM conjugate for tumor cell targeting. PC cell lines and xenograft tumor models were used to assess tumor-cell targeting specificity and killing activity and to investigate the corresponding mechanisms. DZ-SIM treatment effectively killed PC cells regardless of their androgen receptor status or inherent therapeutic resistance. The conjugate targeted and suppressed xenograft tumor formation without harming normal cells of the host. In cancer cells, DZ-SIM was enriched in subcellular organelles, including mitochondria, where the conjugate formed adducts with multiple proteins and caused the loss of transmembrane potential, promoting cell death. The DZ-SIM specifically targets PC cells and their mitochondria, resulting in a loss of mitochondrial function and cell death. With a unique subcellular targeting strategy, the conjugate holds the potential to outperform existing chemotherapeutic drugs. It presents a novel strategy to circumvent therapeutic resistance, offering a more potent treatment for mCRPC.
Collapse
Affiliation(s)
- Yan Ou
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Gina Chia-Yi Chu
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ji Lyu
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Liyuan Yin
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ning Zhai
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Xiaojiang Cui
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Michael S. Lewis
- Department of Pathology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, Los Angeles, CA 90073, United States
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ruoxiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Yi Zhang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| |
Collapse
|
2
|
Mrdenovic S, Wang Y, Yin L, Chu GCY, Ou Y, Lewis MS, Heffer M, Posadas EM, Zhau HE, Chung LWK, Edderkaoui M, Pandol SJ, Wang R, Zhang Y. A cisplatin conjugate with tumor cell specificity exhibits antitumor effects in renal cancer models. BMC Cancer 2023; 23:499. [PMID: 37268911 PMCID: PMC10236852 DOI: 10.1186/s12885-023-10878-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/24/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer and is notorious for its resistance to both chemotherapy and small-molecule inhibitor targeted therapies. Subcellular targeted cancer therapy may thwart the resistance to produce a substantial effect. METHODS We tested whether the resistance can be circumvented by subcellular targeted cancer therapy with DZ-CIS, which is a chemical conjugate of the tumor-cell specific heptamethine carbocyanine dye (HMCD) with cisplatin (CIS), a chemotherapeutic drug with limited use in ccRCC treatment because of frequent renal toxicity. RESULTS DZ-CIS displayed cytocidal effects on Caki-1, 786-O, ACHN, and SN12C human ccRCC cell lines and mouse Renca cells in a dose-dependent manner and inhibited ACHN and Renca tumor formation in experimental mouse models. Noticeably, in tumor-bearing mice, repeated DZ-CIS use did not cause renal toxicity, in contrast to the CIS-treated control animals. In ccRCC tumors, DZ-CIS treatment inhibited proliferation markers but induced cell death marker levels. In addition, DZ-CIS at half maximal inhibitory concentration (IC50) sensitized Caki-1 cells to small-molecule mTOR inhibitors. Mechanistically, DZ-CIS selectively accumulated in ccRCC cells' subcellular organelles, where it damages the structure and function of mitochondria, leading to cytochrome C release, caspase activation, and apoptotic cancer cell death. CONCLUSIONS Results from this study strongly suggest DZ-CIS be tested as a safe and effective subcellular targeted cancer therapy.
Collapse
Affiliation(s)
- Stefan Mrdenovic
- Division of Hematology, Department of Internal Medicine, University Hospital Osijek, Osijek, Croatia
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Internal Medicine, Family Medicine and History of Medicine, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Yanping Wang
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lijuan Yin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gina Chia-Yi Chu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yan Ou
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael S Lewis
- Departments of Pathology, Cedars-Sinai Medical Center and the VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Marija Heffer
- Department of Medical Biology and Genetics, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Edwin M Posadas
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haiyen E Zhau
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Leland W K Chung
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruoxiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Departments of Pathology, Cedars-Sinai Medical Center and the VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yi Zhang
- Biomedical Imaging Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Davis 3059, 90048, Los Angeles, CA, USA.
| |
Collapse
|
3
|
SMYD Family Members Serve as Potential Prognostic Markers and Correlate with Immune Infiltrates in Gastric Cancer. JOURNAL OF ONCOLOGY 2023; 2023:6032864. [PMID: 36816359 PMCID: PMC9929213 DOI: 10.1155/2023/6032864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 02/10/2023]
Abstract
Background The SMYD family comprises a group of genes encoding lysine methyltransferases, which are closely related to tumorigenesis. However, a systematic understanding of their role in gastric cancer (GC) is lacking. Methods Using databases and tools such as the Cancer Genome Atlas, Human Protein Atlas, Kaplan-Meier Plotter, Gene Expression Profiling Interactive Analysis, and Metascape, we comprehensively analyzed differences in SMYD expression and its prognostic value as well as the association of SMYDs with immune cell infiltration, tumor mutational burden (TMB), and microsatellite instability (MSI). We conducted functional enrichment analysis and explored a competing endogenous RNA mechanism regulating SMYD mRNA and protein levels in patients with GC. Results In GC, the expression of SMYD2/3/4/5 mRNA was significantly upregulated, as opposed to that of SMYD1 mRNA, which was significantly downregulated. The protein levels of SMYDs were consistent with mRNA levels. SMYD1/2/4/5 was negatively correlated with overall survival; SMYD1/2/3/5 was negatively correlated with progression-free survival. Our SMYD-based signature and nomogram model may be useful for inferring the prognosis of GC. All SMYDs were closely associated with the infiltration of six immune cell types: uncharacterized, CD8+ T, CD4+ T, macrophage, endothelial, and B cells. TMB was significantly negatively correlated with SMYD1 expression, while a significant positive correlation was observed with SMYD2/5. Furthermore, MSI was significantly positively correlated with SMYD2/5 expression. Long non-coding RNAs, such as chr22-38_28785274-29006793.1, XLOC_002309, and CTD-2008N3.1, were suggested to regulate SMYD expression by sponging multiple microRNAs. Conclusion SMYDs are differentially expressed in GC and are thus potential prognostic markers. SMYD expression is closely related to immune infiltration, TMB, and MSI, all of which are closely related to the response to targeted immune therapy.
Collapse
|
4
|
Yang Y, Qiu R, Zhao S, Shen L, Tang B, Weng Q, Xu Z, Zheng L, Chen W, Shu G, Wang Y, Zhao Z, Chen M, Ji J. SMYD3 associates with the NuRD (MTA1/2) complex to regulate transcription and promote proliferation and invasiveness in hepatocellular carcinoma cells. BMC Biol 2022; 20:294. [PMID: 36575438 PMCID: PMC9795622 DOI: 10.1186/s12915-022-01499-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 12/07/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND SMYD3, a member of the SET and MYND domain-containing (SMYD) family, is a histone methyltransferase (HMT) and transcription factor that plays an important role in transcriptional regulation in human carcinogenesis. RESULTS Using affinity purification and mass spectrometry assays to identify SMYD3-associated proteins in hepatocellular carcinoma (HCC) cells, we found several previously undiscovered SMYD3-interacting proteins, including the NuRD (MTA1/2) complex, the METTL family, and the CRL4B complex. Transcriptomic analysis of the consequences of knocking down SMYD3, MTA1, or MTA2 in HCC cells showed that SMYD3/NuRD complex targets a cohort of genes, some of which are critically involved in cell growth and migration. qChIP analyses showed that SMYD3 knockdown led to a significant reduction in the binding of MTA1 or MTA2 to the promoters of IGFBP4 and led to a significant decrease in H4K20me3 and a marked increase in H4Ac at the IGFBP4 promoter. In addition, we demonstrated that SMYD3 promotes cell proliferation, invasion, and tumorigenesis in vivo and in vitro and found that its expression is markedly upregulated in human liver cancer. Knockdown of MTA1 or MTA2 had the same effect as knockdown of SMYD3 on proliferation and invasion of hepatocellular carcinoma cells. Catalytic mutant SMYD3 could not rescue the phenotypic effects caused by knockdown of SMYD3. Inhibitors of SMYD3 effectively inhibited the proliferation and invasiveness of HCC cells. CONCLUSIONS These findings revealed that SMYD3 could transcriptionally repress a cohort of target genes expression by associating with the NuRD (MTA1/2) complex, thereby promoting the proliferation and invasiveness of HCC cells. Our results support the case for pursuing SMYD3 as a practical prognostic marker or therapeutic target against HCC.
Collapse
Affiliation(s)
- Yang Yang
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Rongfang Qiu
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Siyu Zhao
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Lin Shen
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Bufu Tang
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Qiaoyou Weng
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Ziwei Xu
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Liyun Zheng
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Weiqian Chen
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Gaofeng Shu
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Yajie Wang
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Zhongwei Zhao
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Minjiang Chen
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| | - Jiansong Ji
- grid.268099.c0000 0001 0348 3990Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnosis and Interventional Minimally Invasive Institute, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000 China ,grid.440824.e0000 0004 1757 6428Department of Interventional Radiology, Clinical College of The Affiliated Central Hospital, Lishui University, Lishui, 323000 China
| |
Collapse
|
5
|
Yin L, Li Q, Mrdenovic S, Chu GCY, Wu BJ, Bu H, Duan P, Kim J, You S, Lewis MS, Liang G, Wang R, Zhau HE, Chung LWK. KRT13 promotes stemness and drives metastasis in breast cancer through a plakoglobin/c-Myc signaling pathway. Breast Cancer Res 2022; 24:7. [PMID: 35078507 PMCID: PMC8788068 DOI: 10.1186/s13058-022-01502-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/13/2022] [Indexed: 02/08/2023] Open
Abstract
Background Keratins (KRTs) are intermediate filament proteins that interact with multiple regulatory proteins to initiate signaling cascades. Keratin 13 (KRT13) plays an important role in breast cancer progression and metastasis. The objective of this study is to elucidate the mechanism by which KRT13 promotes breast cancer growth and metastasis.
Methods The function and mechanisms of KRT13 in breast cancer progression and metastasis were assessed by overexpression and knockdown followed by examination of altered behaviors in breast cancer cells and in xenograft tumor formation in mouse mammary fat pad. Human breast cancer specimens were examined by immunohistochemistry and multiplexed quantum dot labeling analysis to correlate KRT13 expression to breast cancer progression and metastasis. Results KRT13-overexpressing MCF7 cells displayed increased proliferation, invasion, migration and in vivo tumor growth and metastasis to bone and lung. Conversely, KRT13 knockdown inhibited the aggressive behaviors of HCC1954 cells. At the molecular level, KRT13 directly interacted with plakoglobin (PG, γ-catenin) to form complexes with desmoplakin (DSP). This complex interfered with PG expression and nuclear translocation and abrogated PG-mediated suppression of c-Myc expression, while the KRT13/PG/c-Myc signaling pathway increased epithelial to mesenchymal transition and stem cell-like phenotype. KRT13 expression in 58 human breast cancer tissues was up-regulated especially at the invasive front and in metastatic specimens (12/18) (p < 0.05). KRT13 up-regulation in primary breast cancer was associated with decreased overall patient survival. Conclusions This study reveals that KRT13 promotes breast cancer cell growth and metastasis via a plakoglobin/c-Myc pathway. Our findings reveal a potential novel pathway for therapeutic targeting of breast cancer progression and metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-022-01502-6.
Collapse
Affiliation(s)
- Lijuan Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Stefan Mrdenovic
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Gina Chia-Yi Chu
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Boyang Jason Wu
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peng Duan
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Jayoung Kim
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael S Lewis
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Gangning Liang
- Department of Urology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Ruoxiang Wang
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA.
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| |
Collapse
|
6
|
Yin L, Zhang Y, Yin L, Ou Y, Lewis MS, Wang R, Zhau HE, Zhou Q, Chung LWK. Novel Mitochondria-Based Targeting Restores Responsiveness in Therapeutically Resistant Human Lung Cancer Cells. Mol Cancer Ther 2021; 20:2527-2538. [PMID: 34583981 PMCID: PMC9559783 DOI: 10.1158/1535-7163.mct-20-1095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/08/2021] [Accepted: 09/24/2021] [Indexed: 02/05/2023]
Abstract
Cisplatin and tyrosine kinase inhibitors (TKI) are recommended to treat non-small cell lung cancer (NSCLC). However, ubiquitously acquired drug resistance in patients with NSCLC diminishes their therapeutic efficacy. Strategies for overcoming cisplatin and TKI resistance are an unmet medical need. We previously described a group of near-infrared heptamethine carbocyanine fluorescent dyes, referred to as DZ, with tumor-homing properties via differentially expressed organic anion-transporting polypeptides on cancer cells. This group of organic dyes can deliver therapeutic payloads specifically to tumor cells in the form of a chemical conjugate. We synthesized DZ-simvastatin (DZ-SIM) initially to target cholesterol biosynthesis in lung cancer cells. DZ-SIM killed both cisplatin-sensitive and cisplatin-resistant as well as EGFR-TKI-sensitive and EGFR-TKI-resistant lung cancer cells. This conjugate specifically accumulated in and effectively inhibited the growth of xenograft tumors formed by NSCLC cells resistant to first-generation (H1650) and third-generation (PC9AR) EGFR TKIs. DZ-SIM induced cell death by targeting mitochondrial structure and function. We concluded that DZ-SIM could be a promising novel therapy for overcoming drug resistance in patients with NSCLC.
Collapse
Affiliation(s)
- Liyuan Yin
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yi Zhang
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lijuan Yin
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yan Ou
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Ruoxiang Wang
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA,Corresponding authors: Leland W. K. Chung, Ph.D., Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 103, Los Angeles, CA, 90048, USA, Tel.: (310) 423-7622, FAX: (310) 423-8543, ; Qinghua Zhou, M.D., Ph.D., No. 37, Guoxue Alley, Chengdu, 610041, China, , Ruoxiang Wang, M.D., Ph.D., Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA, Tel.: (310) 423-9541, FAX: (310) 423-8543,
| | - Haiyen E. Zhau
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Qinghua Zhou
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Corresponding authors: Leland W. K. Chung, Ph.D., Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 103, Los Angeles, CA, 90048, USA, Tel.: (310) 423-7622, FAX: (310) 423-8543, ; Qinghua Zhou, M.D., Ph.D., No. 37, Guoxue Alley, Chengdu, 610041, China, , Ruoxiang Wang, M.D., Ph.D., Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA, Tel.: (310) 423-9541, FAX: (310) 423-8543,
| | - Leland W. K. Chung
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA,Corresponding authors: Leland W. K. Chung, Ph.D., Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 103, Los Angeles, CA, 90048, USA, Tel.: (310) 423-7622, FAX: (310) 423-8543, ; Qinghua Zhou, M.D., Ph.D., No. 37, Guoxue Alley, Chengdu, 610041, China, , Ruoxiang Wang, M.D., Ph.D., Uro-Oncology Research, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA, Tel.: (310) 423-9541, FAX: (310) 423-8543,
| |
Collapse
|
7
|
Wei J, Yin L, Li J, Wang J, Pu T, Duan P, Lin TP, Gao AC, Wu BJ. Bidirectional Cross-talk between MAOA and AR Promotes Hormone-Dependent and Castration-Resistant Prostate Cancer. Cancer Res 2021; 81:4275-4289. [PMID: 34167949 DOI: 10.1158/0008-5472.can-21-0198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/12/2021] [Accepted: 06/21/2021] [Indexed: 01/03/2023]
Abstract
Androgen receptor (AR) is the primary oncogenic driver of prostate cancer, including aggressive castration-resistant prostate cancer (CRPC). The molecular mechanisms controlling AR activation in general and AR reactivation in CRPC remain elusive. Here we report that monoamine oxidase A (MAOA), a mitochondrial enzyme that degrades monoamine neurotransmitters and dietary amines, reciprocally interacts with AR in prostate cancer. MAOA was induced by androgens through direct AR binding to a novel intronic androgen response element of the MAOA gene, which in turn promoted AR transcriptional activity via upregulation of Shh/Gli-YAP1 signaling to enhance nuclear YAP1-AR interactions. Silencing MAOA suppressed AR-mediated prostate cancer development and growth, including CRPC, in mice. MAOA expression was elevated and positively associated with AR and YAP1 in human CRPC. Finally, genetic or pharmacologic targeting of MAOA enhanced the growth-inhibition efficacy of enzalutamide, darolutamide, and apalutamide in both androgen-dependent and CRPC cells. Collectively, these findings identify and characterize an MAOA-AR reciprocal regulatory circuit with coamplified effects in prostate cancer. Moreover, they suggest that cotargeting this complex may be a viable therapeutic strategy to treat prostate cancer and CRPC. SIGNIFICANCE: MAOA and AR comprise a positive feedback loop in androgen-dependent and CRPC, providing a mechanistic rationale for combining MAOA inhibition with AR-targeted therapies for prostate cancer treatment.
Collapse
Affiliation(s)
- Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Lijuan Yin
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Jing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Tianjie Pu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Peng Duan
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Tzu-Ping Lin
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China.,Department of Urology, School of Medicine, Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, Sacramento, California
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington.
| |
Collapse
|
8
|
Bernard BJ, Nigam N, Burkitt K, Saloura V. SMYD3: a regulator of epigenetic and signaling pathways in cancer. Clin Epigenetics 2021; 13:45. [PMID: 33637115 PMCID: PMC7912509 DOI: 10.1186/s13148-021-01021-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/02/2021] [Indexed: 12/16/2022] Open
Abstract
Chromatin modifiers and their implications in oncogenesis have been an exciting area of cancer research. These are enzymes that modify chromatin via post-translational modifications such as methylation, acetylation, sumoylation, phosphorylation, in addition to others. Depending on the modification, chromatin modifiers can either promote or repress transcription. SET and MYN-domain containing 3 (SMYD3) is a chromatin modifier that has been implicated in the development and progression of various cancer types. It was first reported to tri-methylate Histone 3 Lysine 4 (H3K4), a methylation mark known to promote transcription. However, since this discovery, other histone (H4K5 and H4K20, for example) and non-histone (VEGFR, HER2, MAP3K2, ER, and others) substrates of SMYD3 have been described, primarily in the context of cancer. This review aims to provide a background on basic characteristics of SMYD3, such as its protein structure and tissue expression profiles, discuss reported histone and non-histone substrates of SMYD3, and underscore prognostic and functional implications of SMYD3 in cancer. Finally, we briefly discuss ongoing efforts to develop inhibitors of SMYD3 for future therapeutic use. It is our hope that this review will help synthesize existing research on SMYD3 in an effort to propel future discovery.
Collapse
Affiliation(s)
- Benjamin J Bernard
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, 41 Medlars Drive, Bethesda, MD, 20852, USA
| | - Nupur Nigam
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, 41 Medlars Drive, Bethesda, MD, 20852, USA
| | | | - Vassiliki Saloura
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, 41 Medlars Drive, Bethesda, MD, 20852, USA.
| |
Collapse
|
9
|
Tosat-Bitrián C, Palomo V. CdSe quantum dots evaluation in primary cellular models or tissues derived from patients. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 30:102299. [PMID: 32931928 DOI: 10.1016/j.nano.2020.102299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022]
Abstract
In recent years quantum dots (QDs) have risen as useful luminescent nanoparticles with multiple applications ranging from laser, image displays and biomedical applications. Here we review and discuss the studies of these nanoparticles in patient derived cellular samples or tissues, including cellular models from iPSCs from patients, biopsied and post-mortem tissue. QD-based multiplexed imaging has been proved to overcome most of the major drawbacks of conventional techniques, exhibiting higher sensitivity, reliability, accuracy and simultaneous labeling of key biomarkers. In this sense, QDs are very promising tools to be further used in clinical applications including diagnosis and therapy approaches. Analyzing the possibilities of these materials in these biological samples gives an overview of the future applications of the nanoparticles in models closer to patients and their specific disease.
Collapse
Affiliation(s)
| | - Valle Palomo
- Centro de Investigaciones Biológicas Margarita Salas CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Elshafae SM, Dirksen WP, Alasonyalilar-Demirer A, Breitbach J, Yuan S, Kantake N, Supsavhad W, Hassan BB, Attia Z, Rosol TJ. Canine prostatic cancer cell line (LuMa) with osteoblastic bone metastasis. Prostate 2020; 80:698-714. [PMID: 32348616 PMCID: PMC7291846 DOI: 10.1002/pros.23983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Osteoblastic bone metastasis represents the most common complication in men with prostate cancer (PCa). During progression and bone metastasis, PCa cells acquire properties similar to bone cells in a phenomenon called osteomimicry, which promotes their ability to metastasize, proliferate, and survive in the bone microenvironment. The mechanism of osteomimicry resulting in osteoblastic bone metastasis is unclear. METHODS We developed and characterized a novel canine prostatic cancer cell line (LuMa) that will be useful to investigate the relationship between osteoblastic bone metastasis and osteomimicry in PCa. The LuMa cell line was established from a primary prostate carcinoma of a 13-year old mixed breed castrated male dog. Cell proliferation and gene expression of LuMa were measured and compared to three other canine prostatic cancer cell lines (Probasco, Ace-1, and Leo) in vitro. The effect of LuMa cells on calvaria and murine preosteoblastic (MC3T3-E1) cells was measured by quantitative reverse-transcription polymerase chain reaction and alkaline phosphatase assay. LuMa cells were transduced with luciferase for monitoring in vivo tumor growth and metastasis using different inoculation routes (subcutaneous, intratibial [IT], and intracardiac [IC]). Xenograft tumors and metastases were evaluated using radiography and histopathology. RESULTS After left ventricular injection, LuMa cells metastasized to bone, brain, and adrenal glands. IT injections induced tumors with intramedullary new bone formation. LuMa cells had the highest messenger RNA levels of osteomimicry genes (RUNX2, RANKL, and Osteopontin [OPN]), CD44, E-cadherin, and MYOF compared to Ace-1, Probasco, and Leo cells. LuMa cells induced growth in calvaria defects and modulated gene expression in MC3T3-E1 cells. CONCLUSIONS LuMa is a novel canine PCa cell line with osteomimicry and stemness properties. LuMa cells induced osteoblastic bone formation in vitro and in vivo. LuMa PCa cells will serve as an excellent model for studying the mechanisms of osteomimicry and osteoblastic bone and brain metastasis in prostate cancer.
Collapse
Affiliation(s)
- Said M. Elshafae
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Pathology, Faculty of Veterinary medicine, Benha University, Benha, Egypt
- Dept. of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Wessel P. Dirksen
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Aylin Alasonyalilar-Demirer
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Faculty of Veterinary Medicine, Bursa Uludag University, Turkey
| | - Justin Breitbach
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Shiyu Yuan
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Noriko Kantake
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Wachiraphan Supsavhad
- Dept. of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Bardes B. Hassan
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Zayed Attia
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Sadat City University, Sadat City, Egypt
| | - Thomas J. Rosol
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Correspondence to: Dr. Thomas Rosol, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, 225 Irvine Hall, Athens, OH 45701, USA. , Phone: 740.593.2405
| |
Collapse
|
11
|
Wang L, Xu ML, Wang C, Dong QQ, Miao Z, Chen XY, Wang N, He HP, Zhang TC, Luo XG. SET and MYND domain-containing protein 3 inhibits tumor cell sensitivity to cisplatin. Oncol Lett 2020; 19:3469-3476. [PMID: 32269620 PMCID: PMC7114722 DOI: 10.3892/ol.2020.11465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/24/2020] [Indexed: 12/22/2022] Open
Abstract
Cisplatin resistance has been a major factor limiting its clinical use as a chemotherapy drug. The present study aimed to investigate whether SET and MYND domain-containing protein 3 (SMYD3), a histone methyltransferase closely associated with tumors can affect the sensitivity of tumors to cisplatin chemotherapy. Real time-qPCR, western blotting, the luciferase reporter, MTT and clonogenic assays were performed to detect the effects of SMYD3 on the chemotherapy capacity of cisplatin. In the present study, SMYD3 exhibited different expression patterns in MCF-7 and T47D breast cancer cells. In addition, this differential expression was associated with tumor cell resistance to cisplatin. Furthermore, SMYD3 knockdown following small interfering RNA transfection increased cisplatin sensitivity, whereas SMYD3 overexpression decreased cisplatin sensitivity. In addition, SMYD3 knockdown synergistically enhanced cisplatin-induced cell apoptosis. SMYD3 expression was downregulated during cisplatin treatment. In addition, transcriptional regulatory activities of SMYD3 3'-untranslated region were also downregulated. These results suggested that SMYD3 may affect cell sensitivity to cisplatin and participate in the development of cisplatin resistance, which is a process that may involve microRNA-124-mediated regulation.
Collapse
Affiliation(s)
- Lei Wang
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Man-Li Xu
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Chang Wang
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Qing-Qing Dong
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Zhi Miao
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Xiao-Ying Chen
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Nan Wang
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Hong-Peng He
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Xue-Gang Luo
- Key Laboratory of Industrial Fermentation Microbiology, Tianjin University of Science and Technology, Ministry of Education, Tianjin 300457, P.R. China
- Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| |
Collapse
|
12
|
Mrdenovic S, Zhang Y, Wang R, Yin L, Chu GCY, Yin L, Lewis M, Heffer M, Zhau HE, Chung LWK. Targeting Burkitt lymphoma with a tumor cell-specific heptamethine carbocyanine-cisplatin conjugate. Cancer 2019; 125:2222-2232. [PMID: 30840322 PMCID: PMC6618854 DOI: 10.1002/cncr.32033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/16/2019] [Accepted: 02/04/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND Burkitt lymphoma is a fast-growing mature B cell malignancy, whose genetic hallmark is translocation and activation of the c-myc gene. Prompt multiagent immunochemotherapy regimens can have favorable outcomes, but prognosis is poor in refractory or relapsed disease. We previously identified a novel family of near-infrared heptamethine carbocyanine fluorescent dyes (HMCD or DZ) with tumor-homing properties via organic anion-transporting peptides. These membrane carriers have uptake in tumor cells but not normal cells in cell culture, mouse and dog tumor models, patient-derived xenografts, and perfused kidney cancers in human patients. METHODS Here we report the cytotoxic effects of a synthesized conjugate of DZ with cisplatin (CIS) on B cell lymphoma CA46, Daudi, Namalwa, Raji, and Ramos cell lines in cell culture and in xenograft tumor formation. Impaired mitochondrial membrane permeability was examined as the mechanism of DZ-CIS-induced lymphoma cell death. RESULTS The new conjugate, DZ-CIS, is cytotoxic against Burkitt lymphoma cell lines and tumor models. DZ-CIS retains tumor-homing properties to mitochondrial and lysosomal compartments, does not accumulate in normal cells and tissues, and has no nephrotoxicity in mice. DZ-CIS accumulated in Burkitt lymphoma cells and tumors induces apoptosis and retards tumor cell growth in culture and xenograft tumor growth in mice. CONCLUSION DZ-CIS downregulated c-myc and overcame CIS resistance in myc-driven TP53-mutated aggressive B cell Burkitt lymphoma. We propose that DZ-CIS could be used to treat relapsed/refractory aggressive Burkitt lymphomas.
Collapse
Affiliation(s)
- Stefan Mrdenovic
- Division of Hematology, Department of Internal Medicine, University Hospital Osijek, Osijek, Croatia.,Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yi Zhang
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ruoxiang Wang
- Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Lijuan Yin
- Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Gina Chia-Yi Chu
- Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Liyuan Yin
- Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michael Lewis
- Department of Pathology, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Marija Heffer
- Faculty of Medicine, Department of Medical Biology and Genetics, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Haiyen E Zhau
- Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Leland W K Chung
- Department of Medicine, Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
13
|
Mi J, Hooker E, Balog S, Zeng H, Johnson DT, He Y, Yu EJ, Wu H, Le V, Lee DH, Aldahl J, Gonzalgo ML, Sun Z. Activation of hepatocyte growth factor/MET signaling initiates oncogenic transformation and enhances tumor aggressiveness in the murine prostate. J Biol Chem 2018; 293:20123-20136. [PMID: 30401749 DOI: 10.1074/jbc.ra118.005395] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/04/2018] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence has shown that the hepatocyte growth factor (HGF) and its receptor, MET proto-oncogene, receptor tyrosine kinase (MET), promote cell proliferation, motility, morphogenesis, and angiogenesis. Whereas up-regulation of MET expression has been observed in aggressive and metastatic prostate cancer, a clear understanding of MET function in prostate tumorigenesis remains elusive. Here, we developed a conditional Met transgenic mouse strain, H11 Met/+ :PB-Cre4, to mimic human prostate cancer cells with increased MET expression in the prostatic luminal epithelium. We found that these mice develop prostatic intraepithelial neoplasia after HGF administration. To further assess the biological role of MET in prostate cancer progression, we bred H11 Met/+ /PtenLoxP/LoxP:PBCre4 compound mice, in which transgenic Met expression and deletion of the tumor suppressor gene Pten occurred simultaneously only in prostatic epithelial cells. These compound mice exhibited accelerated prostate tumor formation and invasion as well as increased metastasis compared with PtenLoxP/LoxP:PB-Cre4 mice. Moreover, prostatic sarcomatoid carcinomas and lesions resembling the epithelial-to-mesenchymal transition developed in tumor lesions of the compound mice. RNA-Seq and qRT-PCR analyses revealed a robust enrichment of known tumor progression and metastasis-promoting genes in samples isolated from H11 Met/+ /PtenLoxP/LoxP:PB-Cre4 compound mice compared with those from PtenLoxP/LoxP:PB-Cre4 littermate controls. HGF-induced cell proliferation and migration also increased in mouse embryonic fibroblasts (MEFs) from animals with both Met transgene expression and Pten deletion compared with Pten-null MEFs. The results from these newly developed mouse models indicate a role for MET in hastening tumorigenesis and metastasis when combined with the loss of tumor suppressors.
Collapse
Affiliation(s)
- Jiaqi Mi
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - Erika Hooker
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010; the Department of Urology and Stanford University School of Medicine, Stanford, California 94305
| | - Steven Balog
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - Hong Zeng
- the Transgenic, Knockout and Tumor Model Center, Stanford University School of Medicine, Stanford, California 94305, and
| | - Daniel T Johnson
- the Department of Urology and Stanford University School of Medicine, Stanford, California 94305
| | - Yongfeng He
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010; the Department of Urology and Stanford University School of Medicine, Stanford, California 94305
| | - Eun-Jeong Yu
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010; the Department of Urology and Stanford University School of Medicine, Stanford, California 94305
| | - Huiqing Wu
- Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - Vien Le
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - Dong-Hoon Lee
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - Joseph Aldahl
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010
| | - Mark L Gonzalgo
- the Department of Urology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Zijie Sun
- From the Departments of Cancer Biology and Pathology, Beckman Research Institute, City of Hope, Duarte, California 91010; the Department of Urology and Stanford University School of Medicine, Stanford, California 94305.
| |
Collapse
|
14
|
Lin TP, Li J, Li Q, Li X, Liu C, Zeng N, Huang JM, Chu GCY, Lin CH, Zhau HE, Chung LWK, Wu BJ, Shih JC. R1 Regulates Prostate Tumor Growth and Progression By Transcriptional Suppression of the E3 Ligase HUWE1 to Stabilize c-Myc. Mol Cancer Res 2018; 16:1940-1951. [PMID: 30042175 DOI: 10.1158/1541-7786.mcr-16-0346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/22/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
Abstract
Prostate cancer is a prevalent public health problem, especially because noncutaneous advanced malignant forms significantly affect the lifespan and quality of life of men worldwide. New therapeutic targets and approaches are urgently needed. The current study reports elevated expression of R1 (CDCA7L/RAM2/JPO2), a c-Myc-interacting protein and transcription factor, in human prostate cancer tissue specimens. In a clinical cohort, high R1 expression is associated with disease recurrence and decreased patient survival. Overexpression and knockdown of R1 in human prostate cancer cells indicate that R1 induces cell proliferation and colony formation. Moreover, silencing R1 dramatically reduces the growth of prostate tumor xenografts in mice. Mechanistically, R1 increases c-Myc protein stability by inhibiting ubiquitination and proteolysis through transcriptional suppression of HUWE1, a c-Myc-targeting E3 ligase, via direct interaction with a binding element in the promoter. Moreover, transcriptional repression is supported by a negative coexpression correlation between R1 and HUWE1 in a prostate cancer clinical dataset. Collectively, these findings, for the first time, characterize the contribution of R1 to prostate cancer pathogenesis. IMPLICATIONS: These findings provide evidence that R1 is a novel regulator of prostate tumor growth by stabilizing c-Myc protein, meriting further investigation of its therapeutic and prognostic potential.
Collapse
Affiliation(s)
- Tzu-Ping Lin
- Depatment of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
- USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles, California
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Urology, School of Medicine and Shu-Tien Urological Research Center, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Qinlong Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Pathology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiangyan Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chunyan Liu
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ni Zeng
- Depatment of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jen-Ming Huang
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Gina Chia-Yi Chu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Chi-Hung Lin
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Leland W K Chung
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington.
| | - Jean C Shih
- Depatment of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California.
- USC-Taiwan Center for Translational Research, University of Southern California, Los Angeles, California
- Depatment of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
15
|
Stewart PA, Khamis ZI, Zhau HE, Duan P, Li Q, Chung LWK, Sang QXA. Upregulation of minichromosome maintenance complex component 3 during epithelial-to-mesenchymal transition in human prostate cancer. Oncotarget 2018; 8:39209-39217. [PMID: 28424404 PMCID: PMC5503607 DOI: 10.18632/oncotarget.16835] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/08/2017] [Indexed: 01/13/2023] Open
Abstract
Metastasis is often associated with epithelial-to-mesenchymal transition (EMT). To understand the molecular mechanisms of this process, we conducted proteomic analysis of androgen-repressed cancer of the prostate (ARCaP), an experimental model of metastatic human prostate cancer. The protein signatures of epithelial (ARCaPE) and mesenchymal (ARCaPM) cells were consistent with their phenotypes. Importantly, the expression of mini-chromosome maintenance 3 (MCM3) protein, a crucial subunit of DNA helicase, was significantly higher in ARCaPM cells than that of ARCaPE cells. This increased MCM3 protein expression level was verified using Western blot analysis of the ARCaP cell lineages. Furthermore, immunohistochemical analysis of MCM3 protein levels in human prostate tissue specimens showed elevated expression in bone metastasis and advanced human prostate cancer tissue samples. Subcutaneous injection experiments using ARCaPE and ARCaPM cells in a mouse model also revealed increased MCM3 protein levels in mesenchymal-derived tumors. This study identifies MCM3 as an upregulated molecule in mesenchymal phenotype of human prostate cancer cells and advanced human prostate cancer specimens, suggesting MCM3 may be a new potential drug target for prostate cancer treatment.
Collapse
Affiliation(s)
- Paul A Stewart
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, United States of America
| | - Zahraa I Khamis
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, United States of America.,Laboratory of Cancer Biology and Molecular Immunology, Department of Biochemistry, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Haiyen E Zhau
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Peng Duan
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Quanlin Li
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Leland W K Chung
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Qing-Xiang Amy Sang
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, United States of America.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, United States of America
| |
Collapse
|
16
|
Grindel BJ, Martinez JR, Tellman TV, Harrington DA, Zafar H, Nakhleh L, Chung LW, Farach-Carson MC. Matrilysin/MMP-7 Cleavage of Perlecan/HSPG2 Complexed with Semaphorin 3A Supports FAK-Mediated Stromal Invasion by Prostate Cancer Cells. Sci Rep 2018; 8:7262. [PMID: 29740048 PMCID: PMC5940808 DOI: 10.1038/s41598-018-25435-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Interrupting the interplay between cancer cells and extracellular matrix (ECM) is a strategy to halt tumor progression and stromal invasion. Perlecan/heparan sulfate proteoglycan 2 (HSPG2) is an extracellular proteoglycan that orchestrates tumor angiogenesis, proliferation, differentiation and invasion. Metastatic prostate cancer (PCa) cells degrade perlecan-rich tissue borders to reach bone, including the basement membrane, vasculature, reactive stromal matrix and bone marrow. Domain IV-3, perlecan’s last 7 immunoglobulin repeats, mimics native proteoglycan by promoting tumoroid formation. This is reversed by matrilysin/matrix metalloproteinase-7 (MMP-7) cleavage to favor cell dispersion and tumoroid dyscohesion. Both perlecan and Domain IV-3 induced a strong focal adhesion kinase (FAK) dephosphorylation/deactivation. MMP-7 cleavage of perlecan reversed this, with FAK in dispersed tumoroids becoming phosphorylated/activated with metastatic phenotype. We demonstrated Domain IV-3 interacts with the axon guidance protein semaphorin 3A (Sema3A) on PCa cells to deactivate pro-metastatic FAK. Sema3A antibody mimicked the Domain IV-3 clustering activity. Direct binding experiments showed Domain IV-3 binds Sema3A. Knockdown of Sema3A prevented Domain IV-3-induced tumoroid formation and Sema3A was sensitive to MMP-7 proteolysis. The perlecan-Sema3A complex abrogates FAK activity and stabilizes PCa cell interactions. MMP-7 expressing cells destroy the complex to initiate metastasis, destroy perlecan-rich borders, and favor invasion and progression to lethal bone disease.
Collapse
Affiliation(s)
- Brian J Grindel
- Department of BioSciences, Rice University, Houston, TX, 77005, USA.,Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA.,Department of Cancer Systems Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jerahme R Martinez
- Department of BioSciences, Rice University, Houston, TX, 77005, USA.,Department of Mechanical Engineering, University of Delaware, Newark, DE, 19706, USA
| | - Tristen V Tellman
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA
| | - Daniel A Harrington
- Department of BioSciences, Rice University, Houston, TX, 77005, USA.,Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA
| | - Hamim Zafar
- Department of Computer Science, Rice University, Houston, TX, 77005, USA
| | - Luay Nakhleh
- Department of Computer Science, Rice University, Houston, TX, 77005, USA
| | - Leland W Chung
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Mary C Farach-Carson
- Department of BioSciences, Rice University, Houston, TX, 77005, USA. .,Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, 77054, USA.
| |
Collapse
|
17
|
Yin L, Li J, Liao CP, Jason Wu B. Monoamine Oxidase Deficiency Causes Prostate Atrophy and Reduces Prostate Progenitor Cell Activity. Stem Cells 2018; 36:1249-1258. [PMID: 29637670 DOI: 10.1002/stem.2831] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 03/01/2018] [Accepted: 03/26/2018] [Indexed: 12/23/2022]
Abstract
Monoamine oxidases (MAOs) degrade a number of biogenic and dietary amines, including monoamine neurotransmitters, and play an essential role in many biological processes. Neurotransmitters and related neural events have been shown to participate in the development, differentiation, and maintenance of diverse tissues and organs by regulating the specialized cellular function and morphological structures of innervated organs such as the prostate. Here we show that mice lacking both MAO isoforms, MAOA and MAOB, exhibit smaller prostate mass and develop epithelial atrophy in the ventral and dorsolateral prostates. The cellular composition of prostate epithelium showed reduced CK5+ or p63+ basal cells, accompanied by lower Sca-1 expression in p63+ basal cells, but intact differentiated CK8+ luminal cells in MAOA/B-deficient mouse prostates. MAOA/B ablation also decreased epithelial cell proliferation without affecting cell apoptosis in mouse prostates. Using a human prostate epithelial cell line, we found that stable knockdown of MAOA and MAOB impaired the capacity of prostate stem cells to form spheres, coinciding with a reduced CD133+ /CD44+ /CD24- stem cell population and less expression of CK5 and select stem cell markers, including ALDH1A1, TROP2, and CD166. Alternative pharmacological inhibition of MAOs also repressed prostate cell stemness. In addition, we found elevated expression of MAOA and MAOB in epithelial and/or stromal components of human prostate hyperplasia samples compared with normal prostate tissues. Taken together, our findings reveal critical roles for MAOs in the regulation of prostate basal progenitor cells and prostate maintenance. Stem Cells 2018;36:1249-1258.
Collapse
Affiliation(s)
- Lijuan Yin
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Chun-Peng Liao
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| |
Collapse
|
18
|
Zhao N, Zhang C, Zhao Y, Bai B, An J, Zhang H, Wu JB, Shi C. Optical imaging of gastric cancer with near-infrared heptamethine carbocyanine fluorescence dyes. Oncotarget 2018; 7:57277-57289. [PMID: 27329598 PMCID: PMC5302988 DOI: 10.18632/oncotarget.10031] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/26/2016] [Indexed: 12/17/2022] Open
Abstract
Near-infrared fluorescence (NIRF) imaging agents are promising tools for noninvasive cancer imaging. Here, we explored the tumor-specific targeting ability of NIRF heptamethine carbocyanine MHI-148 dye in cultured gastric cancer cells, gastric cancer cell-derived and patient-derived tumor xenograft (PDX) models. We show that the NIRF dye specifically accumulated in tumor regions of both xenograft models, suggesting the potential utility of the dye for tumor-specific imaging and targeting in gastric cancer. We also demonstrated significant correlations between NIRF signal intensity and tumor volume in PDX models. Mechanistically, the higher cellular uptake of MHI-148 in gastric cancer cells than in normal cells was stimulated by hypoxia and activation of a group of organic anion-transporting polypeptide (OATP) genes. Importantly, this NIRF dye was not retained in inflammatory stomach tissues induced by gastric ulcer in mice. In addition, fresh clinical gastric tumor specimens, when perfused with NIR dye, exhibited increased uptake of NIR dye in situ. Together, these results show the possibility of using NIRF dyes as novel candidate agents for clinical imaging and detection of gastric cancer.
Collapse
Affiliation(s)
- Ningning Zhao
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Caiqin Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yong Zhao
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Bing Bai
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jiaze An
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hai Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jason Boyang Wu
- Urologic Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Changhong Shi
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
19
|
Li Q, Yin L, Jones LW, Chu GCY, Wu JBY, Huang JM, Li Q, You S, Kim J, Lu YT, Mrdenovic S, Wang R, Freeman MR, Garraway I, Lewis MS, Chung LWK, Zhau HE. Keratin 13 expression reprograms bone and brain metastases of human prostate cancer cells. Oncotarget 2018; 7:84645-84657. [PMID: 27835867 PMCID: PMC5356688 DOI: 10.18632/oncotarget.13175] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/28/2016] [Indexed: 12/14/2022] Open
Abstract
Lethal progression of prostate cancer metastasis can be improved by developing animal models that recapitulate the clinical conditions. We report here that cytokeratin 13 (KRT13), an intermediate filament protein, plays a directive role in prostate cancer bone, brain, and soft tissue metastases. KRT13 expression was elevated in bone, brain, and soft tissue metastatic prostate cancer cell lines and in primary and metastatic clinical prostate, lung, and breast cancer specimens. When KRT13 expression was determined at a single cell level in primary tumor tissues of 44 prostate cancer cases, KRT13 level predicted bone metastasis and the overall survival of prostate cancer patients. Genetically enforced KRT13 expression in human prostate cancer cell lines drove metastases toward mouse bone, brain and soft tissues through a RANKL-independent mechanism, as KRT13 altered the expression of genes associated with EMT, stemness, neuroendocrine/neuromimicry, osteomimicry, development, and extracellular matrices, but not receptor activator NF-κB ligand (RANKL) signaling networks in prostate cancer cells. Our results suggest new inhibitors targeting RANKL-independent pathways should be developed for the treatment of prostate cancer bone and soft tissue metastases.
Collapse
Affiliation(s)
- Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Current address: Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lijuan Yin
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lawrence W Jones
- Urological Research, Huntington Medical Research Institutes, Huntington Memorial Hospital, Pasadena, CA, USA
| | - Gina C-Y Chu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jason B-Y Wu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jen-Ming Huang
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Quanlin Li
- Biostatistics and Bioinformatics, Department of Medicine, Los Angeles, CA, USA
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jayoung Kim
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yi-Tsung Lu
- John H. Stroger, Jr. Hospital of Cook County, Chicago, IL, USA
| | - Stefan Mrdenovic
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruoxiang Wang
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael R Freeman
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Isla Garraway
- Department of Urology and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA and Division of Urology, Greater Los Angeles Veteran's Affairs Healthcare System, Los Angeles, CA, USA
| | - Michael S Lewis
- Sepulveda Research Corporation VA Medical Center, Los Angeles, CA, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
20
|
Medically translatable quantum dots for biosensing and imaging. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY C-PHOTOCHEMISTRY REVIEWS 2017. [DOI: 10.1016/j.jphotochemrev.2017.01.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
21
|
Josson S, Gururajan M, W.K.Chung L. In situ Hybridization (ISH) and Quantum Dots (QD) of miRNAs. Bio Protoc 2017; 7:e2138. [PMID: 34458458 PMCID: PMC8376534 DOI: 10.21769/bioprotoc.2138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/22/2016] [Accepted: 01/21/2017] [Indexed: 11/02/2022] Open
Abstract
miRNA are short non-coding RNA which inhibit translation of mRNA. miRNA regulate several cellular processes. Certain miRNA are known to induce oncogenesis. miRNA can be measured by real-time PCR and be imaged using a combination of in situ hybridization (ISH) and quantum dots (QD). The advantage of using quantum dots is that several miRNA can be simultaneously measured using multiplexed QD. Additionally, miRNA can be visualized in different regions of the tissue. Since miRNA are biomarkers of various disease states, miRNA can be visualized and quantitated in tissue sections for diagnostic and prognostic purposes. Here we describe ISH-QD analysis of tissue sections. Tissue sections from xenografts or clinical specimens are used. These are deparaffinized, treated with Proteinase K and hybridized with a biotin-probe to specific to the miRNA. The in situ hybridization is performed by labeling the biotin-probes and followed by labeling with streptavidin tagged quantum dots. Image acquisition of the quantum dots is performed and analyzed for the miRNA expression levels. Combining ISH and QD gives a powerful tool to detect miRNA in different cells of the tissue.
Collapse
Affiliation(s)
- Sajni Josson
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, USA
- Present address: Neostrata Inc, Princeton, USA
| | - Murali Gururajan
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, USA
- Present address: Bristol-Myers Squibb Inc, Princeton, USA
| | - Leland W.K.Chung
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| |
Collapse
|
22
|
Zhang T, Boominathan R, Foulk B, Rao C, Kemeny G, Strickler JH, Abbruzzese JL, Harrison MR, Hsu DS, Healy P, Li J, Pi C, Prendergast KM, Hobbs C, Gemberling S, George DJ, Hurwitz HI, Connelly M, Garcia-Blanco MA, Armstrong AJ. Development of a Novel c-MET-Based CTC Detection Platform. Mol Cancer Res 2016; 14:539-47. [PMID: 26951228 DOI: 10.1158/1541-7786.mcr-16-0011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 02/25/2016] [Indexed: 11/16/2022]
Abstract
UNLABELLED Amplification of the MET oncogene is associated with poor prognosis, metastatic dissemination, and drug resistance in many malignancies. We developed a method to capture and characterize circulating tumor cells (CTC) expressing c-MET using a ferromagnetic antibody. Immunofluorescence was used to characterize cells for c-MET, DAPI, and pan-CK, excluding CD45(+) leukocytes. The assay was validated using appropriate cell line controls spiked into peripheral blood collected from healthy volunteers (HV). In addition, peripheral blood was analyzed from patients with metastatic gastric, pancreatic, colorectal, bladder, renal, or prostate cancers. CTCs captured by c-MET were enumerated, and DNA FISH for MET amplification was performed. The approach was highly sensitive (80%) for MET-amplified cells, sensitive (40%-80%) for c-MET-overexpressed cells, and specific (100%) for both c-MET-negative cells and in 20 HVs. Of 52 patients with metastatic carcinomas tested, c-MET CTCs were captured in replicate samples from 3 patients [gastric, colorectal, and renal cell carcinoma (RCC)] with 6% prevalence. CTC FISH demonstrated that MET amplification in both gastric and colorectal cancer patients and trisomy 7 with gain of MET gene copies in the RCC patient. The c-MET CTC assay is a rapid, noninvasive, sensitive, and specific method for detecting MET-amplified tumor cells. CTCs with MET amplification can be detected in patients with gastric, colorectal, and renal cancers. IMPLICATIONS This study developed a novel c-MET CTC assay for detecting c-MET CTCs in patients with MET amplification and warrants further investigation to determine its clinical applicability. Mol Cancer Res; 14(6); 539-47. ©2016 AACR.
Collapse
Affiliation(s)
- Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Rengasamy Boominathan
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Brad Foulk
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Chandra Rao
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Gabor Kemeny
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - John H Strickler
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - James L Abbruzzese
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Michael R Harrison
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - David S Hsu
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Patrick Healy
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Jing Li
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Cinthia Pi
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Katherine M Prendergast
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Carey Hobbs
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Sarah Gemberling
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Daniel J George
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Herbert I Hurwitz
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Mark Connelly
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Mariano A Garcia-Blanco
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Andrew J Armstrong
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina. Department of Pharmacology and Cancer Biology, Duke University, Durham, NC.
| |
Collapse
|
23
|
Ziaee S, Chu GCY, Huang JM, Sieh S, Chung LWK. Prostate cancer metastasis: roles of recruitment and reprogramming, cell signal network and three-dimensional growth characteristics. Transl Androl Urol 2016; 4:438-54. [PMID: 26816842 PMCID: PMC4708593 DOI: 10.3978/j.issn.2223-4683.2015.04.10] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa) metastasizes to bone and soft tissues, greatly decreasing quality of life, causing bone pain, skeletal complications, and mortality in PCa patients. While new treatment strategies are being developed, the molecular and cellular basis of PCa metastasis and the “cross-talk” between cancer cells and their microenvironment and crucial cell signaling pathways need to be successfully dissected for intervention. In this review, we introduce a new concept of the mechanism of PCa metastasis, the recruitment and reprogramming of bystander and dormant cells (DCs) by a population of metastasis-initiating cells (MICs). We provide evidence that recruited and reprogrammed DCs gain MICs phenotypes and can subsequently metastasize to bone and soft tissues. We show that MICs can also recruit and reprogram circulating tumor cells (CTCs) and this could contribute to cancer cell evolution and the acquisition of therapeutic resistance. We summarize relevant molecular signaling pathways, including androgen receptors (ARs) and their variants and growth factors (GFs) and cytokines that could contribute to the predilection of PCa for homing to bone and soft tissues. To understand the etiology and the biology of PCa and the effectiveness of therapeutic targeting, we briefly summarize the animal and cell models that have been employed. We also report our experience in the use of three-dimensional (3-D) culture and co-culture models to understand cell signaling networks and the use of these attractive tools to conduct drug screening exercises against already-identified molecular targets. Further research into PCa growth and metastasis will improve our ability to target cancer metastasis more effectively and provide better rationales for personalized oncology.
Collapse
Affiliation(s)
- Shabnam Ziaee
- 1 Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; 2 Australian Prostate Cancer Research Centre, Brisbane, Queensland 4102, Australia ; 3 Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gina Chia-Yi Chu
- 1 Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; 2 Australian Prostate Cancer Research Centre, Brisbane, Queensland 4102, Australia ; 3 Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jen-Ming Huang
- 1 Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; 2 Australian Prostate Cancer Research Centre, Brisbane, Queensland 4102, Australia ; 3 Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Shirly Sieh
- 1 Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; 2 Australian Prostate Cancer Research Centre, Brisbane, Queensland 4102, Australia ; 3 Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leland W K Chung
- 1 Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA ; 2 Australian Prostate Cancer Research Centre, Brisbane, Queensland 4102, Australia ; 3 Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
24
|
Li Q, Li Q, Nuccio J, Liu C, Duan P, Wang R, Jones LW, Chung LWK, Zhau HE. Metastasis initiating cells in primary prostate cancer tissues from transurethral resection of the prostate (TURP) predicts castration-resistant progression and survival of prostate cancer patients. Prostate 2015; 75:1312-21. [PMID: 25990623 PMCID: PMC4736544 DOI: 10.1002/pros.23011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/10/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND We previously reported that the activation of RANK and c-Met signaling components in both experimental mouse models and human prostate cancer (PC) specimens predicts bone metastatic potential and PC patient survival. This study addresses whether a population of metastasis-initiating cells (MICs) known to express a stronger RANKL, phosphorylated c-Met (p-c-Met), and neuropilin-1 (NRP1) signaling network than bystander or dormant cells (BDCs) can be detected in PC tissues from patients subjected to transurethral resection of the prostate (TURP) for urinary obstruction prior to the diagnosis of PC with or without prior hormonal manipulation, and whether the relative abundance of MICs over BDCs could predict castration-resistant progression and PC patient survival. METHODS We employed a multiplexed quantum-dot labeling (mQDL) protocol to detect and quantify MICs and BDCs at the single cell level in TURP tissues obtained from 44 PC patients with documented overall survival and castration resistance status. RESULTS PC tissues with a higher number of MICs and an activated RANK signaling network, including increased expression of RANKL, p-c-Met, and NRP1 compared to BDCs, were found to correlate with the development of castration resistance and overall survival. CONCLUSIONS The assessment of PC cells with MIC and BDC phenotypes in primary PC tissues from hormone-naïve patients can predict the progression to castration resistance and the overall survival of PC patients.
Collapse
Affiliation(s)
- Qinlong Li
- Department of Medicine, Uro-Oncology Research Program, Los Angeles, California
| | - Quanlin Li
- Biostatistics and Bioinformatics, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jill Nuccio
- Urological Research, Huntington Medical Research Institutes, Huntington Memorial Hospital, Pasadena, California
| | - Chunyan Liu
- Department of Medicine, Uro-Oncology Research Program, Los Angeles, California
| | - Peng Duan
- Department of Medicine, Uro-Oncology Research Program, Los Angeles, California
| | - Ruoxiang Wang
- Department of Medicine, Uro-Oncology Research Program, Los Angeles, California
| | - Lawrence W. Jones
- Urological Research, Huntington Medical Research Institutes, Huntington Memorial Hospital, Pasadena, California
- Correspondence to: Lawrence W. Jones, MD, Urological Research, Huntington Medical Research Institutes, Huntington Memorial Hospital, 99 North El Molino Avenue, Pasadena, CA 91101.
| | - Leland W. K. Chung
- Department of Medicine, Uro-Oncology Research Program, Los Angeles, California
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California
- Correspondence to: Haiyen E. Zhau, PhD, and Leland W. K. Chung, PhD, Department of Medicine, Uro-Oncology Program, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 103, Los Angeles, CA 90048. (HEZ); (LWKC)
| | - Haiyen E. Zhau
- Department of Medicine, Uro-Oncology Research Program, Los Angeles, California
- Correspondence to: Haiyen E. Zhau, PhD, and Leland W. K. Chung, PhD, Department of Medicine, Uro-Oncology Program, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 103, Los Angeles, CA 90048. (HEZ); (LWKC)
| |
Collapse
|
25
|
Abstract
Cancer metastasis is highly inefficient and complex. Common features of metastatic cancer cells have been observed using cancer cell lines and genetically reconstituted mouse and human tumor xenograft models. These include cancer cell interaction with the tumor microenvironment and the ability of cancer cells to sense extracellular stimuli and adapt to adverse growth conditions. This review summarizes the coordinated response of cancer cells to soluble growth factors, such as RANKL, by a unique feed forward mechanism employing coordinated upregulation of RANKL and c-Met with downregulation of androgen receptor. The RANK-mediated signal network was found to drive epithelial to mesenchymal transition in prostate cancer cells, promote osteomimicry and the ability of prostate cancer cells to assume stem cell and neuroendocrine phenotypes, and confer the ability of prostate cancer cells to home to bone. Prostate cancer cells with activated RANK-mediated signal network were observed to recruit and even transform the non-tumorigenic prostate cancer cells to participate in bone and soft tissue colonization. The coordinated regulation of cancer cell invasion and metastasis by the feed forward mechanism involving RANKL, c-Met, transcription factors, and VEGF-neuropilin could offer new therapeutic opportunities to target prostate cancer bone and soft tissue metastases.
Collapse
Affiliation(s)
- Gina Chia-Yi Chu
- Departments of Medicine and Surgery, Samuel Orchin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA,
| | | |
Collapse
|
26
|
Toge M, Yokoyama S, Kato S, Sakurai H, Senda K, Doki Y, Hayakawa Y, Yoshimura N, Saiki I. Critical contribution of MCL-1 in EMT-associated chemo-resistance in A549 non-small cell lung cancer. Int J Oncol 2015; 46:1844-8. [PMID: 25647738 DOI: 10.3892/ijo.2015.2861] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/23/2014] [Indexed: 11/06/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of death in all lung cancer patients due to its metastatic spread. Even though cisplatin treatment after surgical resection of the primary tumor has been established as a standard chemotherapy for residual disease including metastatic spread, NSCLC often acquires a resistance against chemotherapy, and metastatic disease is often observed. Amongst many potential mechanisms, epithelial-to-mesenchymal transition (EMT) has been considered as an important process in acquiring both metastatic spread and chemo-resistance of NSCLC. In this study, we identified MCL-1 as a critical molecule for chemo-resistance in A549 cells associated with TGF-β-induced EMT. Importantly, downregulation of MCL-1 by siRNA or inhibition of MCL-1 with pan-BCL2 inhibitor to inhibit MCL-1 was able to overcome the EMT-associated chemo-resistance in A549 cells. Collectively, MCL-1 can be a new therapeutic target for overcoming EMT-associated chemo-resistance in NSCLC patients in the context of post-operative chemotherapies.
Collapse
Affiliation(s)
- Masayoshi Toge
- Department of Thoracic and Cardiovascular Surgery, University of Toyama, Toyama 930-0194, Japan
| | - Satoru Yokoyama
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Shinichiro Kato
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Hiroaki Sakurai
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Kazutaka Senda
- Department of Thoracic and Cardiovascular Surgery, University of Toyama, Toyama 930-0194, Japan
| | - Yoshinori Doki
- Department of Thoracic and Cardiovascular Surgery, University of Toyama, Toyama 930-0194, Japan
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Naoki Yoshimura
- Department of Thoracic and Cardiovascular Surgery, University of Toyama, Toyama 930-0194, Japan
| | - Ikuo Saiki
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
27
|
Gururajan M, Josson S, Chu GCY, Lu CL, Lu YT, Haga CL, Zhau HE, Liu C, Lichterman J, Duan P, Posadas EM, Chung LWK. miR-154* and miR-379 in the DLK1-DIO3 microRNA mega-cluster regulate epithelial to mesenchymal transition and bone metastasis of prostate cancer. Clin Cancer Res 2014; 20:6559-69. [PMID: 25324143 DOI: 10.1158/1078-0432.ccr-14-1784] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE MicroRNAs in the delta-like 1 homolog-deiodinase, iodothyronine 3 (DLK1-DIO3) cluster have been shown to be critical for embryonic development and epithelial to mesenchymal transition (EMT). DLK1-DIO3 cluster miRNAs are elevated in the serum of patients with metastatic cancer. However, the biologic functions of these miRNAs in the EMT and metastasis of cancer cells are poorly understood. We previously demonstrated the oncogenic and metastatic role of miR-409-3p/5p, a member of this cluster, in prostate cancer. In this study, we defined the role of miR-154* and miR-379, two key members of this cluster, in prostate cancer progression and bone metastasis in both cell line models and clinical specimens. EXPERIMENTAL DESIGN Genetic manipulation of miR-154* and miR-379 was performed to determine their role in tumor growth, EMT, and bone metastasis in mouse models. We determined the expression of miR-154* in prostate cancer clinical samples and bone metastasis samples using in situ hybridization and quantum dot labeling. RESULTS Elevated expression of miR-154* and miR-379 was observed in bone metastatic prostate cancer cell lines and tissues, and miR-379 expression correlated with progression-free survival of patients with prostate cancer. Intracardiac inoculation (to mimic systemic dissemination) of miR-154* inhibitor-treated bone metastatic ARCaPM prostate cancer cells in mice led to decreased bone metastasis and increased survival. CONCLUSION miR-154* and miR-379 play important roles in prostate cancer biology by facilitating tumor growth, EMT, and bone metastasis. This finding has particular translational importance because miRNAs in the DLK1-DIO3 cluster can be attractive biomarkers and possible therapeutic targets to treat bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Murali Gururajan
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Sajni Josson
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Gina Chia-Yi Chu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chia-Lun Lu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yi-Tsung Lu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chunyan Liu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jake Lichterman
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Peng Duan
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Edwin M Posadas
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
28
|
Stack EC, Wang C, Roman KA, Hoyt CC. Multiplexed immunohistochemistry, imaging, and quantitation: a review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods 2014; 70:46-58. [PMID: 25242720 DOI: 10.1016/j.ymeth.2014.08.016] [Citation(s) in RCA: 551] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/12/2014] [Accepted: 08/29/2014] [Indexed: 12/16/2022] Open
Abstract
Tissue sections offer the opportunity to understand a patient's condition, to make better prognostic evaluations and to select optimum treatments, as evidenced by the place pathology holds today in clinical practice. Yet, there is a wealth of information locked up in a tissue section that is only partially accessed, due mainly to the limitations of tools and methods. Often tissues are assessed primarily based on visual analysis of one or two proteins, or 2-3 DNA or RNA molecules. Even while analysis is still based on visual perception, image analysis is starting to address the variability of human perception. This is in contrast to measuring characteristics that are substantially out of reach of human perception, such as parameters revealed through co-expression, spatial relationships, heterogeneity, and low abundance molecules. What is not routinely accessed is the information revealed through simultaneous detection of multiple markers, the spatial relationships among cells and tissue in disease, and the heterogeneity now understood to be critical to developing effective therapeutic strategies. Our purpose here is to review and assess methods for multiplexed, quantitative, image analysis based approaches, using new multicolor immunohistochemistry methods, automated multispectral slide imaging, and advanced trainable pattern recognition software. A key aspect of our approach is presenting imagery in a workflow that engages the pathologist to utilize the strengths of human perception and judgment, while significantly expanding the range of metrics collectable from tissue sections and also provide a level of consistency and precision needed to support the complexities of personalized medicine.
Collapse
|
29
|
Josson S, Gururajan M, Sung SY, Hu P, Shao C, Zhau HE, Liu C, Lichterman J, Duan P, Li Q, Rogatko A, Posadas EM, Haga CL, Chung LWK. Stromal fibroblast-derived miR-409 promotes epithelial-to-mesenchymal transition and prostate tumorigenesis. Oncogene 2014; 34:2690-9. [PMID: 25065597 DOI: 10.1038/onc.2014.212] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 06/10/2014] [Accepted: 06/15/2014] [Indexed: 11/09/2022]
Abstract
Tumor-stromal interaction is a dynamic process that promotes tumor growth and metastasis via cell-cell interaction and extracellular vesicles. Recent studies demonstrate that stromal fibroblast-derived molecular signatures can be used to predict disease progression and drug resistance. To identify the epigenetic role of stromal noncoding RNAs in tumor-stromal interactions in the tumor microenvironment, we performed microRNA profiling of patient cancer-associated prostate stromal fibroblasts isolated by laser capture dissection microscopy and in bone-associated stromal models. We found specific upregulation of miR-409-3p and miR-409-5p located within the embryonically and developmentally regulated DLK1-DIO3 (delta-like 1 homolog-deiodinase, iodothyronine 3) cluster on human chromosome 14. The findings in cell lines were further validated in human prostate cancer tissues. Strikingly, ectopic expression of miR-409 in normal prostate fibroblasts conferred a cancer-associated stroma-like phenotype and led to the release of miR-409 via extracellular vesicles to promote tumor induction and epithelial-to-mesenchymal transition in vitro and in vivo. miR-409 promoted tumorigenesis through repression of tumor suppressor genes such as Ras suppressor 1 and stromal antigen 2. Thus, stromal fibroblasts derived miR-409-induced tumorigenesis, epithelial-to-mesenchymal transition and stemness of the epithelial cancer cells in vivo. Therefore, miR-409 appears to be an attractive therapeutic target to block the vicious cycle of tumor-stromal interactions that plagues prostate cancer patients.
Collapse
Affiliation(s)
- S Josson
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - M Gururajan
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - S Y Sung
- The Ph.D. program for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - P Hu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - C Shao
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - H E Zhau
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - C Liu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - J Lichterman
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - P Duan
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Q Li
- Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - A Rogatko
- Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - E M Posadas
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - C L Haga
- The Scripps Research Institute, Jupiter, FL, USA
| | - L W K Chung
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
30
|
Josson S, Gururajan M, Hu P, Shao C, Chu GY, Zhau HE, Liu C, Lao K, Lu CL, Lu YT, Lichterman J, Nandana S, Li Q, Rogatko A, Berel D, Posadas EM, Fazli L, Sareen D, Chung LWK. miR-409-3p/-5p promotes tumorigenesis, epithelial-to-mesenchymal transition, and bone metastasis of human prostate cancer. Clin Cancer Res 2014; 20:4636-46. [PMID: 24963047 DOI: 10.1158/1078-0432.ccr-14-0305] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE miR-409-3p/-5p is a miRNA expressed by embryonic stem cells, and its role in cancer biology and metastasis is unknown. Our pilot studies demonstrated elevated miR-409-3p/-5p expression in human prostate cancer bone metastatic cell lines; therefore, we defined the biologic impact of manipulation of miR-409-3p/-5p on prostate cancer progression and correlated the levels of its expression with clinical human prostate cancer bone metastatic specimens. EXPERIMENTAL DESIGN miRNA profiling of a prostate cancer bone metastatic epithelial-to-mesenchymal transition (EMT) cell line model was performed. A Gleason score human tissue array was probed for validation of specific miRNAs. In addition, genetic manipulation of miR-409-3p/-5p was performed to determine its role in tumor growth, EMT, and bone metastasis in mouse models. RESULTS Elevated expression of miR-409-3p/-5p was observed in bone metastatic prostate cancer cell lines and human prostate cancer tissues with higher Gleason scores. Elevated miR-409-3p expression levels correlated with progression-free survival of patients with prostate cancer. Orthotopic delivery of miR-409-3p/-5p in the murine prostate gland induced tumors where the tumors expressed EMT and stemness markers. Intracardiac inoculation (to mimic systemic dissemination) of miR-409-5p inhibitor-treated bone metastatic ARCaPM prostate cancer cells in mice led to decreased bone metastasis and increased survival compared with control vehicle-treated cells. CONCLUSION miR-409-3p/-5p plays an important role in prostate cancer biology by facilitating tumor growth, EMT, and bone metastasis. This finding bears particular translational importance as miR-409-3p/-5p appears to be an attractive biomarker and/or possibly a therapeutic target to treat bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Sajni Josson
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California.
| | - Murali Gururajan
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California.
| | - Peizhen Hu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Chen Shao
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - GinaChia-Yi Chu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Chunyan Liu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Kaiqin Lao
- Genetic Systems, Life Technologies Inc., South San Francisco, California
| | - Chia-Lun Lu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Yi-Tsung Lu
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Jake Lichterman
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Srinivas Nandana
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Quanlin Li
- Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Andre Rogatko
- Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Dror Berel
- Biostatistics and Bioinformatics, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Edwin M Posadas
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
| | - Ladan Fazli
- Vancouver Prostate Cancer Center, University of British Columbia, Vancouver, Canada
| | - Dhruv Sareen
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California.
| |
Collapse
|
31
|
Wu JB, Shao C, Li X, Shi C, Li Q, Hu P, Chen YT, Dou X, Sahu D, Li W, Harada H, Zhang Y, Wang R, Zhau HE, Chung LWK. Near-infrared fluorescence imaging of cancer mediated by tumor hypoxia and HIF1α/OATPs signaling axis. Biomaterials 2014; 35:8175-85. [PMID: 24957295 DOI: 10.1016/j.biomaterials.2014.05.073] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/24/2014] [Indexed: 11/26/2022]
Abstract
Near-infrared fluorescence (NIRF) imaging agents are promising tools for noninvasive cancer imaging. Here, we explored the mechanistic properties of a specific group of NIR heptamethine carbocyanines including MHI-148 dye we identified and synthesized, and demonstrated these dyes to achieve cancer-specific imaging and targeting via a hypoxia-mediated mechanism. We found that cancer cells and tumor xenografts exhibited hypoxia-dependent MHI-148 dye uptake in vitro and in vivo, which was directly mediated by hypoxia-inducible factor 1α (HIF1α). Microarray analysis and dye uptake assay further revealed a group of hypoxia-inducible organic anion-transporting polypeptides (OATPs) responsible for dye uptake, and the correlation between OATPs and HIF1α was manifested in progressive clinical cancer specimens. Finally, we demonstrated increased uptake of MHI-148 dye in situ in perfused clinical tumor samples with activated HIF1α/OATPs signaling. Our results establish these NIRF dyes as potential tumor hypoxia-dependent cancer-targeting agents and provide a mechanistic rationale for continued development of NIRF imaging agents for improved cancer detection, prognosis and therapy.
Collapse
Affiliation(s)
- Jason Boyang Wu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Chen Shao
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiangyan Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Changhong Shi
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Peizhen Hu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yi-Ting Chen
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xiaoliang Dou
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Divya Sahu
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Wei Li
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Hiroshi Harada
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Yi Zhang
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ruoxiang Wang
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
32
|
Nomura T, Huang WC, Zhau HE, Josson S, Mimata H, Chung LWK. β2-Microglobulin-mediated signaling as a target for cancer therapy. Anticancer Agents Med Chem 2014; 14:343-52. [PMID: 23848204 PMCID: PMC3931390 DOI: 10.2174/18715206113139990092] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 09/22/2012] [Accepted: 05/19/2013] [Indexed: 01/02/2023]
Abstract
β2-microglobulin (β2-m) has become the focus of intense scrutiny since the discovery of its undesirable roles promoting
osteomimicry and cancer progression. β2-m is a well-known housekeeping protein that forms complexes with the heavy chain of major
histocompatibility complex class I molecules, which are heterodimeric cell surface proteins that present antigenic peptides to cytotoxic T
cells. On recognition of foreign peptide antigens on cell surfaces, T cells actively bind and lyse antigen-presenting cancer cells. In
addition to its roles in tumor immunity, β2-m has two different functions in cancer cells, either tumor promoting or tumor suppressing, in
cancer cell context-dependent manner. Our studies have demonstrated that β2-m is involved extensively in the functional regulation of
growth, survival, apoptosis, and even metastasis of cancer cells. We found that β2-m is a soluble growth factor and a pleiotropic signaling
molecule which interacts with its receptor, hemochromatosis protein, to modulate epithelial-to-mesenchymal transition (EMT) through
iron-responsive pathways. Specific antibodies against β2-m have remarkable tumoricidal activity in cancer, through β2-m action on iron
flux, alterations of intracellular reactive oxygen species, DNA damage and repair enzyme activities, β-catenin activation and cadherin
switching, and tumor responsiveness to hypoxia. These novel functions of β2-m and β2-m signaling may be common to several solid
tumors including human lung, breast, renal, and prostate cancers. Our experimental results could lead to the development of a novel class
of antibody-based pharmaceutical agents for cancer growth control. In this review, we briefly summarize the recent data regarding β2-m
as a promising new cancer therapeutic target and discuss antagonizing this therapeutic target with antibody therapy for the treatment of
localized and disseminated cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Leland W K Chung
- Department of Urology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan.
| |
Collapse
|
33
|
Shao C, Liao CP, Hu P, Chu CY, Zhang L, Bui MHT, Ng CS, Josephson DY, Knudsen B, Tighiouart M, Kim HL, Zhau HE, Chung LWK, Wang R, Posadas EM. Detection of live circulating tumor cells by a class of near-infrared heptamethine carbocyanine dyes in patients with localized and metastatic prostate cancer. PLoS One 2014; 9:e88967. [PMID: 24551200 PMCID: PMC3925210 DOI: 10.1371/journal.pone.0088967] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 01/14/2014] [Indexed: 02/05/2023] Open
Abstract
Tumor cells are inherently heterogeneous and often exhibit diminished adhesion, resulting in the shedding of tumor cells into the circulation to form circulating tumor cells (CTCs). A fraction of these are live CTCs with potential of metastatic colonization whereas others are at various stages of apoptosis making them likely to be less relevant to understanding the disease. Isolation and characterization of live CTCs may augment information yielded by standard enumeration to help physicians to more accurately establish diagnosis, choose therapy, monitor response, and provide prognosis. We previously reported on a group of near-infrared (NIR) heptamethine carbocyanine dyes that are specifically and actively transported into live cancer cells. In this study, this viable tumor cell-specific behavior was utilized to detect live CTCs in prostate cancer patients. Peripheral blood mononuclear cells (PBMCs) from 40 patients with localized prostate cancer together with 5 patients with metastatic disease were stained with IR-783, the prototype heptamethine cyanine dye. Stained cells were subjected to flow cytometric analysis to identify live (NIR(+)) CTCs from the pool of total CTCs, which were identified by EpCAM staining. In patients with localized tumor, live CTC counts corresponded with total CTC numbers. Higher live CTC counts were seen in patients with larger tumors and those with more aggressive pathologic features including positive margins and/or lymph node invasion. Even higher CTC numbers (live and total) were detected in patients with metastatic disease. Live CTC counts declined when patients were receiving effective treatments, and conversely the counts tended to rise at the time of disease progression. Our study demonstrates the feasibility of applying of this staining technique to identify live CTCs, creating an opportunity for further molecular interrogation of a more biologically relevant CTC population.
Collapse
Affiliation(s)
- Chen Shao
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Department of Urology Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chun-Peng Liao
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America
| | - Peizhen Hu
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America
| | - Chia-Yi Chu
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America
| | - Lei Zhang
- Department of Epidemiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Matthew H T Bui
- Divsion of Urology- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Christopher S Ng
- Divsion of Urology- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - David Y Josephson
- Divsion of Urology- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Beatrice Knudsen
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Translational Pathology and Biobank, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Mourad Tighiouart
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Hyung L Kim
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Divsion of Urology- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Haiyen E Zhau
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America
| | - Leland W K Chung
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Division of Hematology Oncology-Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ruoxiang Wang
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Division of Hematology Oncology-Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Edwin M Posadas
- Uro-Oncolgy Research Laboratories, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Urologic Oncology Program, Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angles, California, United States of America ; Division of Hematology Oncology-Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
34
|
Nakamura H, Wang Y, Xue H, Romanish MT, Mager DL, Helgason CD, Wang Y. Genistein versus ICI 182, 780: an ally or enemy in metastatic progression of prostate cancer. Prostate 2013; 73:1747-60. [PMID: 24038102 DOI: 10.1002/pros.22712] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/20/2013] [Indexed: 11/09/2022]
Abstract
BACKGROUND Androgen signalling through the androgen receptor (AR) plays a critical role in prostate cancer (PCa) initiation and progression. Estrogen in synergy with androgen is essential for cell growth of the normal and malignant prostate. However, the exact role that estrogen and the estrogen receptor play in prostate carcinogenesis remains unclear. We have previously demonstrated the metastasis-promoting effect of an estrogen receptor beta (ERβ) agonist (genistein) in a patient-derived PCa xenograft model mimicking localized and metastatic disease. METHODS To test the hypothesis that the tumor-promoting activity of genistein was due to its estrogenic properties, we treated the xenograft-bearing mice with genistein and an anti-estrogen compound (ICI 182, 780) and compared the differential gene expression using microarrays. RESULTS Using a second xenograft model which was derived from another patient, we showed that genistein promoted disease progression in vivo and ICI 182, 780 inhibited metastatic spread. The microarray analysis revealed that the metallothionein (MT) gene family was differentially expressed in tumors treated by these compounds. Using qRT-PCR, the differences in expression levels were validated in the metastatic and non-metastatic LTL313 PCa xenograft tumor lines, both of which were originally derived from the same PCa patient. CONCLUSIONS Together our data provide evidence that genistein stimulates and ICI 182, 780 inhibits metastatic progression, suggesting that these effects may be mediated by ERβ signalling.
Collapse
Affiliation(s)
- Hisae Nakamura
- Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada; Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada; The Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The field of anatomic pathology has changed significantly over the last decades and, as a result of the technological developments in molecular pathology and genetics, has had increasing pressures put on it to become quantitative and to provide more information about protein expression on a cellular level in tissue sections. Multispectral imaging (MSI) has a long history as an advanced imaging modality and has been used for over a decade now in pathology to improve quantitative accuracy, enable the analysis of multicolor immunohistochemistry, and drastically reduce the impact of contrast-robbing tissue autofluorescence common in formalin-fixed, paraffin-embedded tissues. When combined with advanced software for the automated segmentation of different tissue morphologies (eg, tumor vs stroma) and cellular and subcellular segmentation, MSI can enable the per-cell quantitation of many markers simultaneously. This article covers the role that MSI has played in anatomic pathology in the analysis of formalin-fixed, paraffin-embedded tissue sections, discusses the technological aspects of why MSI has been adopted, and provides a review of the literature of the application of MSI in anatomic pathology.
Collapse
|
36
|
Hu P, Chung LWK, Berel D, Frierson HF, Yang H, Liu C, Wang R, Li Q, Rogatko A, Zhau HE. Convergent RANK- and c-Met-mediated signaling components predict survival of patients with prostate cancer: an interracial comparative study. PLoS One 2013; 8:e73081. [PMID: 24066029 PMCID: PMC3774681 DOI: 10.1371/journal.pone.0073081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 07/16/2013] [Indexed: 01/13/2023] Open
Abstract
We reported (PLoS One 6 (12):e28670, 2011) that the activation of c-Met signaling in RANKL-overexpressing bone metastatic LNCaP cell and xenograft models increased expression of RANK, RANKL, c-Met, and phosphorylated c-Met, and mediated downstream signaling. We confirmed the significance of the RANK-mediated signaling network in castration resistant clinical human prostate cancer (PC) tissues. In this report, we used a multispectral quantum dot labeling technique to label six RANK and c-Met convergent signaling pathway mediators simultaneously in formalin fixed paraffin embedded (FFPE) tissue specimens, quantify the intensity of each expression at the sub-cellular level, and investigated their potential utility as predictors of patient survival in Caucasian-American, African-American and Chinese men. We found that RANKL and neuropilin-1 (NRP-1) expression predicts survival of Caucasian-Americans with PC. A Gleason score ≥8 combined with nuclear p-c-Met expression predicts survival in African-American PC patients. Neuropilin-1, p-NF-κB p65 and VEGF are predictors for the overall survival of Chinese men with PC. These results collectively support interracial differences in cell signaling networks that can predict the survival of PC patients.
Collapse
Affiliation(s)
- Peizhen Hu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Leland W. K. Chung
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Dror Berel
- Biostatistics and Bioinformatics, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Henry F. Frierson
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Hua Yang
- Department of Pathology, Jilin University, Changchun, Jilin, China
| | - Chunyan Liu
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ruoxiang Wang
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Qinlong Li
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Andre Rogatko
- Biostatistics and Bioinformatics, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Haiyen E. Zhau
- Uro-Oncology Research Program, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
37
|
Abstract
Stromal features in carcinomas may provide a relatively consistent means to stratify patients afflicted with solid tumors. Stroma-derived transcriptome signatures can now be used to make predictions about patient survival, suggesting the potential for their clinical application in precision medicine to predict disease progression and emergence of therapeutic resistance.
Collapse
Affiliation(s)
- Michael R. Freeman
- Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA. 90048
- Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA. 90048
- Department of Biomedical Sciences, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA. 90048
| | - Quanlin Li
- Biostatistics and Bioinformatics Center, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA. 90048
| | - Leland W. K. Chung
- Department of Medicine, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA. 90048
- Department of Surgery, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA. 90048
| |
Collapse
|
38
|
Zhau HE, Li Q, Chung LWK. Interracial differences in prostate cancer progression among patients from the United States, China and Japan. Asian J Androl 2013; 15:705-7. [PMID: 23872663 DOI: 10.1038/aja.2013.86] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Although previous studies indicate interracial differences in prostate cancer epidemiology based on gene expression profiles among patients from the United States, China and Japan, evidence at the genetic and phenotypic levels that these differences exist and manifest along ethnic lines has been sparse. Recent studies, however, suggest that genetic differences, such as the lower incidence of Chinese prostate cancers harboring TMPRSS2-ERG translocations compared to patients from Western countries, should be carefully considered in the context of genotypic and phenotypic differences among interracial groups. New, more efficient technologies need to be developed to validate genetic, gene expression and/or phenotypic differences associated with prostate cancer tissue specimens obtained from interracial groups, to establish reliable clinical standards that take racial/ethnic data into account to improve the diagnosis, prognosis and treatment of patients with prostate cancer.
Collapse
|
39
|
Liu T, Mendes DE, Berkman CE. From AR to c-Met: androgen deprivation leads to a signaling pathway switch in prostate cancer cells. Int J Oncol 2013; 43:1125-30. [PMID: 23877345 PMCID: PMC3829778 DOI: 10.3892/ijo.2013.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/02/2013] [Indexed: 12/16/2022] Open
Abstract
Elucidating the role of androgen deprivation in the transition from androgen-dependence to independence may enable the development of more specific therapeutic strategies against prostate cancer. Our previous in vitro model was employed to further assess the effects of continuous androgen‑deprivation on prostate cancer cells (LNCaP) with respect to both androgen receptor (AR) and c-Met expression. The results indicated that long-term androgen deprivation resulted in a signaling pathway switch from AR to c-Met in androgen-sensitive cells, which was confirmed by immunofluorescence imaging and western blot analysis. This signaling pathway switch may be predictive of a more aggressive disease state following androgen deprivation therapy.
Collapse
Affiliation(s)
- Tiancheng Liu
- Department of Chemistry, Washington State University, Pullman, WA 99164, USA
| | | | | |
Collapse
|
40
|
Duan Z, Zou JX, Yang P, Wang Y, Borowsky AD, Gao AC, Chen HW. Developmental and androgenic regulation of chromatin regulators EZH2 and ANCCA/ATAD2 in the prostate Via MLL histone methylase complex. Prostate 2013; 73:455-66. [PMID: 23038103 DOI: 10.1002/pros.22587] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 08/16/2012] [Indexed: 02/03/2023]
Abstract
BACKGROUND Chromatin regulators ANCCA and EZH2 are overexpressed in prostate cancer and play crucial roles in androgen-stimulated and castration-refractory prostate tumor growth and survival. However, how their expression is regulated in the tumors and whether they play a role in prostate development remains unclear. METHODS Prostate tissue from different developmental stages of mouse and human were examined by IHC, qRT-PCR and Western for expression of ANCCA, EZH2, and Ki-67. Animals were castrated and T-implanted for the expression response in normal prostate and tumors. siRNA knockdown and ChIP were performed for the mechanism of ANCCA regulation of EZH2. RESULTS In contrast to their very low level expression in adult prostate, ANCCA and EZH2 are strongly expressed in the epithelium and mesenchyme of mouse and human UGS. Their expression becomes more restricted to epithelial cells during later development and displays a second peak during puberty, which correlates with the proliferative status of the epithelium. Importantly, their expression in normal prostate and tumors is strongly suppressed by castration and markedly induced by testosterone replacement. While androgen suppresses EZH2 in CRPC cells, in LNCaP cells, physiological concentrations of androgen stimulate expression of PRC2 genes (EZH2, SUZ12, and EED), which is mediated by androgen-induced ANCCA and involves E2F and histone H3K4me3 methylase MLL1 complex. CONCLUSION EZH2 and ANCCA are androgen regulated and strongly expressed in early prostate morphogenesis and during puberty, suggesting their important role in prostate development. Regulation of EZH2 by ANCCA emphasizes bromodomain protein ANCCA as a potential therapeutic target against prostate cancer.
Collapse
Affiliation(s)
- Zhijian Duan
- Cancer Center/Basic Sciences, University of California at Davis, Sacramento, California 95817, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Jin Z, Hildebrandt N. Semiconductor quantum dots for in vitro diagnostics and cellular imaging. Trends Biotechnol 2012; 30:394-403. [PMID: 22608980 DOI: 10.1016/j.tibtech.2012.04.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/18/2012] [Accepted: 04/18/2012] [Indexed: 12/23/2022]
Abstract
The need for companion diagnostics, point-of-care testing (POCT) and high-throughput screening in clinical diagnostics and personalized medicine has pushed the need for more biological information from a single sample at extremely low concentrations and volumes. Optical biosensors based on semiconductor quantum dots (QDs) can answer these requirements because their unique photophysical properties are ideally suited for highly sensitive multiplexed detection. Many different biological systems have been successfully scrutinized with a large variety of QDs over the past decade but their future as widely applied commercial biosensors is still open. In this review, we highlight recent in vitro diagnostic and cellular imaging applications of QDs and discuss milestones and obstacles on their way toward integration into real-life diagnostic and medical applications.
Collapse
Affiliation(s)
- Zongwen Jin
- Université Paris-Sud, Institut d'Electronique Fondamentale, Orsay, France
| | | |
Collapse
|