1
|
Eltit F, Wang Q, Jung N, Munshan S, Xie D, Xu S, Liang D, Mojtahedzadeh B, Liu D, Charest-Morin R, Corey E, True LD, Morrissey C, Wang R, Cox ME. Sclerotic prostate cancer bone metastasis: woven bone lesions with a twist. JBMR Plus 2024; 8:ziae091. [PMID: 39224570 PMCID: PMC11365963 DOI: 10.1093/jbmrpl/ziae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/07/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Bone metastases are the most severe and prevalent consequences of prostate cancer (PC), affecting more than 80% of patients with advanced PC. PCBMs generate pain, pathological fractures, and paralysis. As modern therapies increase survival, more patients are suffering from these catastrophic consequences. Radiographically, PCBMs are predominantly osteosclerotic, but the mechanisms of abnormal bone formation and how this pathological increase in bone density is related to fractures are unclear. In this study, we conducted a comprehensive analysis on a cohort of 76 cadaveric PCBM specimens and 12 cancer-free specimens as controls. We used micro-computed tomography to determine 3D organization and quantify bone characteristics, quantitative backscattering electron microscopy to characterize mineral content and details in bone structure, nanoindentation to determine mechanical properties, and histological and immunohistochemical analysis of bone structure and composition. We define 4 PCBM phenotypes: osteolytic, mixed lytic-sclerotic, and 2 subgroups of osteosclerotic lesions-those with residual trabeculae, and others without residual trabeculae. The osteosclerotic lesions are characterized by the presence of abnormal bone accumulated on trabeculae surfaces and within intertrabecular spaces. This abnormal bone is characterized by higher lacunae density, abnormal lacunae morphology, and irregular lacunae orientation. However, mineral content, hardness, and elastic modulus at micron-scale were indistinguishable between this irregular bone and residual trabeculae. The collagen matrix of this abnormal bone presents with irregular organization and a prominent collagen III composition. These characteristics suggest that osteosclerotic PCBMs initiate new bone deposition as woven bone; however, the lack of subsequent bone remodeling, absence of lamellar bone deposition on its surface, and presence of collagen III distinguish this pathologic matrix from conventional woven bone. Although the mineralized matrix retains normal bone hardness and stiffness properties, the lack of fibril anisotropy presents a compromised trabecular structure, which may have clinical implications.
Collapse
Affiliation(s)
- Felipe Eltit
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Qiong Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Naomi Jung
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Sheryl Munshan
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Dennis Xie
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Samuel Xu
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Doris Liang
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
- Centre for Aging SMART, Vancouver, BC V5Z 1M9, Canada
| | - Bita Mojtahedzadeh
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Danmei Liu
- Centre for Aging SMART, Vancouver, BC V5Z 1M9, Canada
| | - Raphaële Charest-Morin
- Department of Orthopaedics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- International Collaboration on Repair Discoveries, Vancouver, BC V5Z 1M9, Canada
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, United States
| | - Lawrence D True
- Department of Urology, University of Washington, Seattle, WA 98195, United States
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA 98195, United States
| | - Rizhi Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
- Centre for Aging SMART, Vancouver, BC V5Z 1M9, Canada
| | - Michael E Cox
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Centre for Aging SMART, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
2
|
Siripongsatian D, Jantarato A, Promteangtrong C, Kunawudhi A, Kiatkittikul P, Boonkawin N, Yaset S, Somboon S, Chotipanich C. Pretherapeutic PSMA PET-Derived Semiquantitative Parameters as Predictors of PSA Response in Patients with mCRPC Receiving [ 177 Lu]Lu-PSMA-617 Radioligand Therapy. Indian J Radiol Imaging 2024; 34:579-587. [PMID: 39318581 PMCID: PMC11419753 DOI: 10.1055/s-0044-1779634] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
Objective [ 177 Lu]Lu-prostate-specific membrane antigen (PSMA)-617 radioligand therapy (RLT) shows promise for metastatic castration-resistant prostate cancer (mCRPC) patients with positive PSMA positron emission tomography (PET) imaging. Identifying high-risk patients is crucial. We evaluated pretherapeutic PSMA PET-derived parameters to predict prostate-specific antigen (PSA) response in patients undergoing [ 177 Lu]Lu-PSMA-617 RLT. Materials and Methods We conducted a retrospective analysis among 27 patients (mean age: 71.0 ± 9.5 years; range: 52-85 years) who underwent PSMA PET/computed tomography (CT) and subsequent [ 177 Lu]Lu-PSMA-617 RLT between March 2019 and January 2023. After excluding patients with liver metastases, the number of patients left for analysis was 21 (14 responders and 7 nonresponders). Tumors were semiautomatically delineated with calculation of total tumor volume (PSMA-TV), lesion uptake (PSMA-TLU = PSMA-TV * standardized uptake value [SUV]mean), and lesion quotient (PSMA-TLQ = PSMA-TV/SUVmean) for each patient. Semiquantitative parameters were analyzed only in patients with mCRPC and no liver metastasis. Results In total, 17/27 patients (62.96%) had a decline in PSA levels; 15/27 patients (55.56%) experienced a decline of > 50%. Pretherapeutic PSMA PET/CT results revealed significant differences in PSMA-TV ( p = 0.003), PSMA-TLU ( p = 0.013), and PSMA-TLQ ( p = 0.011) between responders and nonresponders. SUVmax was significantly correlated to the best percentage change in PSA response after 177 Lu-PSMA-617 treatment ( r = -0.79, p = 0.006). No association was observed between PSMA-TV ( p = 0.367), PSMA-TLU ( p = 0.128), and PSMA-TLQ ( p = 0.556), with the best percentage change in PSA response after 177 Lu-PSMA-617 therapy. Conclusion Pretherapeutic PSMA PET-derived PSMA-TV, PSMA-TLU, and PSMA-TLQ were significant negative predictors of PSA response in patients with mCRPC and no liver metastasis receiving [ 177 Lu]Lu-PSMA-617 RLT.
Collapse
Affiliation(s)
| | - Attapon Jantarato
- National Cyclotron and PET Centre, Chulabhorn Hospital, Bangkok, Thailand
| | | | - Anchisa Kunawudhi
- National Cyclotron and PET Centre, Chulabhorn Hospital, Bangkok, Thailand
| | | | | | - Sukanya Yaset
- National Cyclotron and PET Centre, Chulabhorn Hospital, Bangkok, Thailand
| | - Sirinsuda Somboon
- National Cyclotron and PET Centre, Chulabhorn Hospital, Bangkok, Thailand
| | | |
Collapse
|
3
|
Yekedüz E, McKay RR, Gillessen S, Choueiri TK, Ürün Y. Visceral Metastasis Predicts Response to New Hormonal Agents in Metastatic Castration-Sensitive Prostate Cancer. Oncologist 2023:7135855. [PMID: 37084289 DOI: 10.1093/oncolo/oyad102] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/22/2023] [Indexed: 04/23/2023] Open
Abstract
Visceral metastasis (VM) and a higher number of bone metastasis generally define high volume/risk in patients with metastatic castration-sensitive prostate cancer (mCSPC). Subgroup analysis of pivotal trials did not show a clear benefit of second-generation non-steroidal anti-androgens (NSAAs) in patients with VM. However, subgroup analysis of the trial assessing abiraterone acetate, a CYP 17 inhibitor, plus prednisone (AAP) showed an improved overall survival (OS) in patients with mCSPC with VM. We searched MEDLINE, Web of Science, and congress abstracts for the phase III randomized controlled trials of second-generation NSAAs and AAP in patients with mCSPC. In this pooled analysis, we included 6485 patients from the 6 phase III trials. The rate of patients with VM was 15.2%. Interestingly, in contrast to NSAAs, AAP seems to be effective in improving OS among patients with VM (hazard ratio, HR: 0.89, 95% CI, 0.72-1.11, P = .30 for second-generation NSAAs; HR: 0.58, 95% CI, 0.40-0.84, P = .004 for AAP). In contrast, both second-generation NSAAs (HR: 0.63, 95% CI, 0.57-0.70, P < .001) and AAP (HR: 0.68, 95% CI, 0.57-0.81, P < .001) improved OS in patients without VM. In this pooled analysis, we demonstrate that while AAP provided an OS improvement in patients with VM, second-generation NSAAs did not demonstrate a similar OS benefit in this population.
Collapse
Affiliation(s)
- Emre Yekedüz
- Medical Oncology Clinic, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
- Department of Medical Oncology, Università della Svizzera Italiana, Lugano, Switzerland
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye
| |
Collapse
|
4
|
Yasir M, Park J, Han ET, Park WS, Han JH, Kwon YS, Lee HJ, Hassan M, Kloczkowski A, Chun W. Exploration of Flavonoids as Lead Compounds against Ewing Sarcoma through Molecular Docking, Pharmacogenomics Analysis, and Molecular Dynamics Simulations. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010414. [PMID: 36615603 PMCID: PMC9823950 DOI: 10.3390/molecules28010414] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023]
Abstract
Ewing sarcoma (ES) is a highly malignant carcinoma prevalent in children and most frequent in the second decade of life. It mostly occurs due to t(11;22) (q24;q12) translocation. This translocation encodes the oncogenic fusion protein EWS/FLI (Friend leukemia integration 1 transcription factor), which acts as an aberrant transcription factor to deregulate target genes essential for cancer. Traditionally, flavonoids from plants have been investigated against viral and cancerous diseases and have shown some promising results to combat these disorders. In the current study, representative flavonoid compounds from various subclasses are selected and used to disrupt the RNA-binding motif of EWS, which is required for EWS/FLI fusion. By blocking the RNA-binding motif of EWS, it might be possible to combat ES. Therefore, molecular docking experiments validated the binding interaction patterns and structural behaviors of screened flavonoid compounds within the active region of the Ewing sarcoma protein (EWS). Furthermore, pharmacogenomics analysis was used to investigate potential drug interactions with Ewing sarcoma-associated genes. Finally, molecular dynamics simulations were used to investigate the stability of the best selected docked complexes. Taken together, daidzein, kaempferol, and genistein exhibited a result comparable to ifosfamide in the proposed in silico study and can be further analyzed as possible candidate compounds in biological in vitro studies against ES.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Yong-Soo Kwon
- College of Pharmacy, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Hee-Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
- Correspondence: ; Tel.: +82-33-250-8853
| |
Collapse
|
5
|
Chen C, Huang R, Zhou J, Guo L, Xiang S. Formation of pre-metastatic bone niche in prostate cancer and regulation of traditional chinese medicine. Front Pharmacol 2022; 13:897942. [PMID: 36059977 PMCID: PMC9428453 DOI: 10.3389/fphar.2022.897942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Prostate cancer with bone metastasis has a high cancer-specific mortality. Thus, it is essential to delineate the mechanism of bone metastasis. Pre-metastatic niche (PMN) is a concept in tumor metastasis, which is characterized by tumor-secreted factors, reprogramming of stromal cells, and immunosuppression by myeloid-derived suppressor cells (MDSC), which is induced by bone marrow-derived cells (BMDC) in the target organ. However, PMN does not explain the predilection of prostate cancer towards bone metastasis. In this review, we discuss the initiation of bone metastasis of prostate cancer from the perspective of PMN and tumor microenvironment in a step-wise manner. Furthermore, we present a new concept called pre-metastatic bone niche, featuring inherent BMDC, to interpret bone metastasis. Moreover, we illustrate the regulation of traditional Chinese medicine on PMN.
Collapse
|
6
|
Zaragoza-Huesca D, Nieto-Olivares A, García-Molina F, Ricote G, Montenegro S, Sánchez-Cánovas M, Garrido-Rodríguez P, Peñas-Martínez J, Vicente V, Martínez F, Lozano ML, Carmona-Bayonas A, Martínez-Martínez I. Implication of Hepsin from Primary Tumor in the Prognosis of Colorectal Cancer Patients. Cancers (Basel) 2022; 14:cancers14133106. [PMID: 35804878 PMCID: PMC9264764 DOI: 10.3390/cancers14133106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Hepsin is a type II transmembrane serine protease whose deregulation promotes tumor invasion by proteolysis of the pericellular components. In colorectal cancer, the implication of hepsin is unknown. Consequently, we aimed to study the correlations between hepsin expression and different clinical-histopathological variables in 169 patients with localized colorectal cancer and 118 with metastases. Tissue microarrays were produced from samples at diagnosis of primary tumors and stained with an anti-hepsin antibody. Hepsin expression was correlated with clinical-histopathological variables by using the chi-square and Kruskal−Wallis tests, Kaplan−Meier and Aalen−Johansen estimators, and Cox and Fine and Gray multivariate models. In localized cancer patients, high-intensity hepsin staining was associated with reduced 5-year disease-free survival (p-value = 0.16). Medium and high intensity of hepsin expression versus low expression was associated with an increased risk of metastatic relapse (hazard ratio 2.83, p-value = 0.035 and hazard ratio 3.30, p-value = 0.012, respectively), being a better prognostic factor than classic histological variables. Additionally, in patients with localized tumor, 5-year thrombosis cumulative incidence increased with the increment of hepsin expression (p-value = 0.038). Medium and high intensities of hepsin with respect to low intensity were associated with an increase in thrombotic risk (hazard ratio 7.71, p-value = 0.043 and hazard ratio 9.02, p-value = 0.028, respectively). This relationship was independent of previous tumor relapse (p-value = 0.036). Among metastatic patients, low hepsin expression was associated with a low degree of tumor differentiation (p-value < 0.001) and with major metastatic dissemination (p-value = 0.023). Hepsin is a potential thrombotic and metastatic biomarker in patients with localized colorectal cancer. In metastatic patients, hepsin behaves in a paradoxical way with respect to differentiation and invasion processes.
Collapse
Affiliation(s)
- David Zaragoza-Huesca
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
| | - Andrés Nieto-Olivares
- Department of Pathology, Hospital General Universitario Morales Meseguer, 30008 Murcia, Spain;
| | - Francisco García-Molina
- Department of Pathology, Hospital General Universitario Reina Sofía, 30003 Murcia, Spain; (F.G.-M.); (F.M.)
| | - Guillermo Ricote
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
| | - Sofía Montenegro
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
| | - Manuel Sánchez-Cánovas
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
| | - Pedro Garrido-Rodríguez
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, U-765-CIBERER, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Julia Peñas-Martínez
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
| | - Vicente Vicente
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, U-765-CIBERER, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Francisco Martínez
- Department of Pathology, Hospital General Universitario Reina Sofía, 30003 Murcia, Spain; (F.G.-M.); (F.M.)
| | - María Luisa Lozano
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, U-765-CIBERER, Instituto de Salud Carlos III, 28220 Madrid, Spain
| | - Alberto Carmona-Bayonas
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
- Correspondence: (A.C.-B.); (I.M.-M.); Tel.: +34-968-341-990 (A.C.-B. & I.M.-M.)
| | - Irene Martínez-Martínez
- Centro Regional de Hemodonación, Department of Haematology and Medical Oncology, Hospital General Universitario Morales Meseguer, University of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain; (D.Z.-H.); (G.R.); (S.M.); (M.S.-C.); (P.G.-R.); (J.P.-M.); (V.V.); (M.L.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, U-765-CIBERER, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Correspondence: (A.C.-B.); (I.M.-M.); Tel.: +34-968-341-990 (A.C.-B. & I.M.-M.)
| |
Collapse
|
7
|
Yang F, Shi J, Zhang L, Wang H, Li Y. Role of fibulin-1 gene promoter methylation in the carcinogenesis and development of tongue squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2022; 133:432-440. [PMID: 35153187 DOI: 10.1016/j.oooo.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/26/2021] [Accepted: 10/06/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Recently, fibulin-1 (FBLN1) has been shown to be downregulated in various cancers via promoter hypermethylation. Our study aimed to determine the expression and methylation status of FBLN1 in tongue squamous cell carcinoma (TSCC) tissues and cells. METHODS Methylation-specific polymerase chain reaction was implemented to detect the methylation status of the FBLN1 gene in TSCC tissues and Western blot analysis was used to detect the expression of FBLN1 protein. The human TSCC cell lines CAL27 and SCC9 were cultured in vitro and treated with 5-aza-deoxycytidine (5-Aza-dC). CCK-8, colony formation, and Transwell assays were performed to test TSCC cell proliferation, migration, and invasion following 5-Aza-dC treatment or overexpression of FBLN1, which was further verified in in vivo experiments. RESULTS FBLN1 was hypermethylated and the protein expression was reduced in TSCC tissues. After human TSCC cell lines (CAL27 and SCC9) were treated with 5-Aza-dC or overexpressed FBLN1, FBLN1 expression was upregulated and the TSCC cell proliferation, migration, and invasion abilities were suppressed. In vivo experiments further showed that demethylation or overexpression of FBLN1 slowed tumor growth in nude mice. CONCLUSION This study demonstrated that 5-Aza-dC treatment or overexpression of FBLN1 inhibited the growth of human TSCC.
Collapse
Affiliation(s)
- Fang Yang
- Department of Oncology, Daqing Oilfield General Hospital, Daqing, China
| | - Jing Shi
- Department of Stomatology, Daqing Oilfield General Hospital, Daqing, China
| | - Liangyu Zhang
- Department of Oncology, Daqing Oilfield General Hospital, Daqing, China.
| | - Hongyan Wang
- Department of Stomatology, Daqing Oilfield General Hospital, Daqing, China
| | - Yang Li
- Department of Stomatology, Daqing Oilfield General Hospital, Daqing, China
| |
Collapse
|
8
|
Fang Y, Yang H, Hu G, Lu J, Zhou J, Gao N, Gu Y, Zhang C, Qiu J, Guo Y, Zhang Y, Wen Q, Qiao H. The POR rs10954732 polymorphism decreases susceptibility to hepatocellular carcinoma and hepsin as a prognostic biomarker correlated with immune infiltration based on proteomics. J Transl Med 2022; 20:88. [PMID: 35164791 PMCID: PMC8842912 DOI: 10.1186/s12967-022-03282-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/25/2022] [Indexed: 11/10/2022] Open
Abstract
The effect of the cytochrome P450 oxidoreductase (POR) rs10954732 (G > A) polymorphism on hepatocellular carcinoma (HCC) susceptibility is unknown. Here we found that A allele carriers showed a 69% decrease in susceptibility to HCC with overall survival (OS) prolonged to 199%, accompanied by lower activity for cytochrome P450 2E1. A total of 222 differentially expressed proteins were mainly enriched in neutrophil and T cell activation and involved in the immune and inflammatory responses, constituting the altered immune tumor microenvironment related with A allele by proteomics analysis. Hepsin (HPN) showed significant down-regulation in HCC and up-regulation in A allele carriers. A lower HPN level was associated with increased susceptibility to HCC and a worse prognosis. Moreover, HPN is a potential independent prognostic biomarker for HCC and is strongly associated with clinicopathological features, tumor-infiltrating status of immune cells both in our discovery cohort and database surveys. Our findings provide a new potential mechanism by which HPN may play an important role in the susceptibility of rs10954732 A allele carriers to HCC and their prognosis through tumor immune infiltration, thus offering potential insights for future studies on tumor immunotherapy.
Collapse
|
9
|
LI J, HU S, LI H, JIANG J, WANG J. Clinical prognosis and gene expression profiles of prostate cancer patients with bone and lymphatic metastases. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.57221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
| | | | - Huafu LI
- Sun Yat-Sen University, China; Sun Yat-Sen University, China
| | | | | |
Collapse
|
10
|
Dzobo K, Dandara C. Architecture of Cancer-Associated Fibroblasts in Tumor Microenvironment: Mapping Their Origins, Heterogeneity, and Role in Cancer Therapy Resistance. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 24:314-339. [PMID: 32496970 DOI: 10.1089/omi.2020.0023] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumor stroma, a key component of the tumor microenvironment (TME), is a key determinant of response and resistance to cancer treatment. The stromal cells, extracellular matrix (ECM), and blood vessels influence cancer cell response to therapy and play key roles in tumor relapse and therapeutic outcomes. Of the stromal cells present in the TME, much attention has been given to cancer-associated fibroblasts (CAFs) as they are the most abundant and important in cancer initiation, progression, and therapy resistance. Besides releasing several factors, CAFs also synthesize the ECM, a key component of the tumor stroma. In this expert review, we examine the role of CAFs in the regulation of tumor cell behavior and reveal how CAF-derived factors and signaling influence tumor cell heterogeneity and development of novel strategies to combat cancer. Importantly, CAFs display both phenotypic and functional heterogeneity, with significant ramifications on CAF-directed therapies. Principal anti-cancer therapies targeting CAFs take the form of: (1) CAFs' ablation through use of immunotherapies, (2) re-education of CAFs to normalize the cells, (3) cellular therapies involving CAFs delivering drugs such as oncolytic adenoviruses, and (4) stromal depletion via targeting the ECM and its related signaling. The CAFs' heterogeneity could be a result of different cellular origins and the cancer-specific tumor microenvironmental effects, underscoring the need for further multiomics and biochemical studies on CAFs and the subsets. Lastly, we present recent advances in therapeutic targeting of CAFs and the success of such endeavors or their lack thereof. We recommend that to advance global public health and personalized medicine, treatments in the oncology clinic should be combinatorial in nature, strategically targeting both cancer cells and stromal cells, and their interactions.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Vaught DB, Merkel AR, Lynch CC, Edwards J, Tantawy MN, Hilliard T, Wang S, Peterson T, Johnson RW, Sterling JA, Brantley‐Sieders D. EphA2 Is a Clinically Relevant Target for Breast Cancer Bone Metastatic Disease. JBMR Plus 2021; 5:e10465. [PMID: 33869989 PMCID: PMC8046157 DOI: 10.1002/jbm4.10465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/22/2020] [Accepted: 01/12/2021] [Indexed: 11/23/2022] Open
Abstract
EphA2 receptor tyrosine kinase (RTK) is highly expressed in breast tumor cells across multiple molecular subtypes and correlates with poor patient prognosis. In this study, the potential role of EphA2 in this clinically relevant phenomenon is investigated as metastasis of breast cancer to bone is a major cause of morbidity and mortality in patients. It was found that the EphA2 function in breast cancer cells promotes osteoclast activation and the development of osteolytic bone disease. Blocking EphA2 function molecularly and pharmacologically in breast tumors reduced the number and size of bone lesions and the degree of osteolytic disease in intratibial and intracardiac mouse models, which correlated with a significant decrease in the number of osteoclasts at the tumor-bone interface. EphA2 loss of function in tumor cells impaired osteoclast progenitor differentiation in coculture, which is mediated, at least in part, by reduced expression of IL-6. EPHA2 transcript levels are enriched in human breast cancer bone metastatic lesions relative to visceral metastatic sites; EphA2 protein expression was detected in breast tumor cells in bone metastases in patient samples, supporting the clinical relevance of the study's findings. These data provide a strong rationale for the development and application of molecularly targeted therapies against EphA2 for the treatment of breast cancer bone metastatic disease. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- David B Vaught
- Department of Cancer BiologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Alyssa R Merkel
- Vanderbilt Center for Bone BiologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Conor C Lynch
- Department of Tumor BiologyH. Lee Moffitt Cancer CenterTampaFLUSA
| | | | - Mohammed Noor Tantawy
- Radiology and Vanderbilt Institute of Imaging SciencesVanderbilt University School of MedicineNashvilleTNUSA
| | - Timothy Hilliard
- Radiology and Vanderbilt Institute of Imaging SciencesVanderbilt University School of MedicineNashvilleTNUSA
| | - Shan Wang
- Department of Medicine, Division of Rheumatology and ImmunologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Todd Peterson
- Radiology and Vanderbilt Institute of Imaging SciencesVanderbilt University School of MedicineNashvilleTNUSA
| | - Rachelle W Johnson
- Vanderbilt Center for Bone BiologyVanderbilt University School of MedicineNashvilleTNUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University School of MedicineNashvilleTNUSA
- Department of Tumor BiologyH. Lee Moffitt Cancer CenterTampaFLUSA
- Division of Clinical PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Julie A Sterling
- Vanderbilt Center for Bone BiologyVanderbilt University School of MedicineNashvilleTNUSA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (VISN 9)Vanderbilt UniversityNashvilleTNUSA
| | - Dana Brantley‐Sieders
- Vanderbilt‐Ingram Cancer CenterVanderbilt University School of MedicineNashvilleTNUSA
- Department of Tumor BiologyH. Lee Moffitt Cancer CenterTampaFLUSA
- Radiology and Vanderbilt Institute of Imaging SciencesVanderbilt University School of MedicineNashvilleTNUSA
- Department of Medicine, Division of Rheumatology and ImmunologyVanderbilt University School of MedicineNashvilleTNUSA
| |
Collapse
|
12
|
Labrecque MP, Brown LG, Coleman IM, Nguyen HM, Lin DW, Corey E, Nelson PS, Morrissey C. Cabozantinib can block growth of neuroendocrine prostate cancer patient-derived xenografts by disrupting tumor vasculature. PLoS One 2021; 16:e0245602. [PMID: 33471819 PMCID: PMC7817027 DOI: 10.1371/journal.pone.0245602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
With the advent of potent second-line anti-androgen therapy, we and others have observed an increased incidence of androgen receptor (AR)-null small cell or neuroendocrine prostate cancer (SCNPC) in metastatic castration-resistant prostate cancer (mCRPC). Our study was designed to determine the effect of cabozantinib, a multi-targeted tyrosine kinase inhibitor that inhibits VEGFR2, MET and RET on SCNPC. Transcriptome analysis of the University of Washington rapid autopsy and SU2C mCRPC datasets revealed upregulated MET and RET expression in SCNPCs relative to adenocarcinomas. Additionally, increased MET expression correlated with attenuated AR expression and activity. In vitro treatment of SCNPC patient-derived xenograft (PDX) cells with the MET inhibitor AMG-337 had no impact on cell viability in LuCaP 93 (MET+/RET+) and LuCaP 173.1 (MET-/RET-), whereas cabozantinib decreased cell viability of LuCaP 93, but not LuCaP 173.1. Notably, MET+/RET+ LuCaP 93 and MET-/RET- LuCaP 173.1 tumor volumes were significantly decreased with cabozantinib treatment in vivo, and this activity was independent of MET or RET expression in LuCaP 173.1. Tissue analysis indicated that cabozantinib did not inhibit tumor cell proliferation (Ki67), but significantly decreased microvessel density (CD31) and increased hypoxic stress and glycolysis (HK2) in LuCaP 93 and LuCaP 173.1 tumors. RNA-Seq and gene set enrichment analysis revealed that hypoxia and glycolysis pathways were increased in cabozantinib-treated tumors relative to control tumors. Our data suggest that the most likely mechanism of cabozantinib-mediated tumor growth suppression in SCNPC PDX models is through disruption of the tumor vasculature. Thus, cabozantinib may represent a potential therapy for patients with metastatic disease in tumor phenotypes that have a significant dependence on the tumor vasculature for survival and proliferation.
Collapse
Affiliation(s)
- Mark P. Labrecque
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Lisha G. Brown
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ilsa M. Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Holly M. Nguyen
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Daniel W. Lin
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
13
|
Liu Y, Xiong S, Liu S, Chen J, Yang H, Liu G, Li G. Analysis of Gene Expression in Bladder Cancer: Possible Involvement of Mitosis and Complement and Coagulation Cascades Signaling Pathway. J Comput Biol 2020; 27:987-998. [PMID: 31545079 DOI: 10.1089/cmb.2019.0237] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Ying Liu
- Key Laboratory of Southwest Rice Biology and Genetic Breeding, Ministry of Agriculture, Rice and Sorghum Research Institute, Sichuan Academy of Agricultural Sciences, Deyang City, P.R. China
| | - Shenghua Xiong
- Department of Anesthesia, Sichuan Province Transportation Hall Hospital, Chengdu City, P.R. China
| | - Shiying Liu
- Department of Medical Laboratory, The General Hospital of Western Theater Command, Chengdu City, P.R. China
| | - Jie Chen
- Department of Medical Laboratory, The General Hospital of Western Theater Command, Chengdu City, P.R. China
| | - Hao Yang
- Department of Medical Laboratory, People's Hospital of Deyang City, Deyang City, P.R. China
| | - Gang Liu
- Department of Medical Laboratory, The General Hospital of Western Theater Command, Chengdu City, P.R. China
| | - Gengmi Li
- Key Laboratory of Southwest Rice Biology and Genetic Breeding, Ministry of Agriculture, Rice and Sorghum Research Institute, Sichuan Academy of Agricultural Sciences, Deyang City, P.R. China
- Department of Medical Laboratory, The General Hospital of Western Theater Command, Chengdu City, P.R. China
| |
Collapse
|
14
|
Connor MJ, Shah TT, Horan G, Bevan CL, Winkler M, Ahmed HU. Cytoreductive treatment strategies for de novo metastatic prostate cancer. Nat Rev Clin Oncol 2019; 17:168-182. [PMID: 31712648 DOI: 10.1038/s41571-019-0284-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
In the past decade, a revolution in the treatment of metastatic prostate cancer has occurred with the advent of novel hormonal agents and life-prolonging chemotherapy regimens in combination with standard androgen-deprivation therapy. Notwithstanding, the use of systemic therapy alone can result in a castrate-resistant state; therefore, increasing focus is being placed on the additional survival benefits that could potentially be achieved with local cytoreductive and/or metastasis-directed therapies. Local treatment of the primary tumour with the established modalities of radiotherapy and radical prostatectomy has been explored in this context, and the use of novel minimally invasive ablative therapies has been proposed. In addition, evidence of the potential clinical benefits of metastasis-directed therapy with ionizing radiation (primarily stereotactic ablative radiotherapy) is accumulating. Herein, we summarize the pathobiological rationale for local cytoreduction and the potentially systemic immunological responses to radiotherapy and ablative therapies in patients with metastatic prostate cancer. We also discuss the current evidence base for a cytoreductive strategy, including metastasis-directed therapy, in the current era of sequential multimodal therapy incorporating novel treatments. Finally, we outline further research questions relating to this complex and evolving treatment landscape.
Collapse
Affiliation(s)
- Martin J Connor
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK. .,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK.
| | - Taimur T Shah
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Gail Horan
- Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Charlotte L Bevan
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Mathias Winkler
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Hashim U Ahmed
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
15
|
Manna FL, Karkampouna S, Zoni E, De Menna M, Hensel J, Thalmann GN, Kruithof-de Julio M. Metastases in Prostate Cancer. Cold Spring Harb Perspect Med 2019; 9:a033688. [PMID: 29661810 PMCID: PMC6396340 DOI: 10.1101/cshperspect.a033688] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Prostate cancer (PCa) prognosis and clinical outcome is directly dependent on metastatic occurrence. The bone microenvironment is a favorable metastatic niche. Different biological processes have been suggested to contribute to the osteotropism of PCa such as hemodynamics, bone-specific signaling interactions, and the "seed and soil" hypothesis. However, prevalence of disseminating tumor cells in the bone is not proportional to the actual occurrence of metastases, as not all patients will develop bone metastases. The fate and tumor-reforming ability of a metastatic cell is greatly influenced by the microenvironment. In this review, the molecular mechanisms of bone and soft-tissue metastasis in PCa are discussed. Specific attention is dedicated to the residual disease, novel approaches, and animal models used in oncological translational research are illustrated.
Collapse
Affiliation(s)
- Federico La Manna
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Sofia Karkampouna
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Eugenio Zoni
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Marta De Menna
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Janine Hensel
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - George N Thalmann
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department of Urology, Inselspital, Bern University Hospital, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
16
|
Lam HM, Nguyen HM, Corey E. Generation of Prostate Cancer Patient-Derived Xenografts to Investigate Mechanisms of Novel Treatments and Treatment Resistance. Methods Mol Biol 2018; 1786:1-27. [PMID: 29786784 DOI: 10.1007/978-1-4939-7845-8_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Treatment advances lead to survival benefits of patients with advanced prostate cancer. These treatments are highly efficacious in a subset of patients; however, similarly to other cancers, after initial responses the tumors develop resistance (acquired resistance) and the patients succumb to the disease. Furthermore, there is a subset of patients who do not respond to the treatment at all (de novo resistance). Preclinical testing using patient-derived xenografts (PDXs) has led to successful drug development, and PDXs will continue to provide valuable resources to generate clinically relevant data with translational potential. PDXs demonstrate tumor heterogeneity observed in patients, preserve tumor-microenvironment architecture, and provide clinically relevant treatment responses. In view of the evolving biology of the advanced prostate cancer associated with new treatments, PDXs representing these new tumor phenotypes are urgently needed for the study of treatment responses and resistance. In this chapter, we describe methodologies used to establish prostate cancer PDXs and use of these PDXs to study de novo and acquired resistance.
Collapse
Affiliation(s)
- Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Holly M Nguyen
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Abstract
Tumours display considerable variation in the patterning and properties of angiogenic blood vessels, as well as in their responses to anti-angiogenic therapy. Angiogenic programming of neoplastic tissue is a multidimensional process regulated by cancer cells in concert with a variety of tumour-associated stromal cells and their bioactive products, which encompass cytokines and growth factors, the extracellular matrix and secreted microvesicles. In this Review, we discuss the extrinsic regulation of angiogenesis by the tumour microenvironment, highlighting potential vulnerabilities that could be targeted to improve the applicability and reach of anti-angiogenic cancer therapies.
Collapse
Affiliation(s)
- Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Daniela Biziato
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Division of Experimental Pathology, University of Lausanne and University of Lausanne Hospital, 1066 Lausanne, Switzerland
| |
Collapse
|
18
|
Liu Y, Wilson D, Leach RJ, Chen Y. MBDDiff: an R package designed specifically for processing MBDcap-seq datasets. BMC Genomics 2016; 17 Suppl 4:432. [PMID: 27556923 PMCID: PMC5001203 DOI: 10.1186/s12864-016-2794-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background Since its initial discovery in 1975, DNA methylation has been intensively studied and shown to be involved in various biological processes, such as development, aging and tumor progression. Many experimental techniques have been developed to measure the level of DNA methylation. Methyl-CpG binding domain-based capture followed by high-throughput sequencing (MBDCap-seq) is a widely used method for characterizing DNA methylation patterns in a genome-wide manner. However, current methods for processing MBDCap-seq datasets does not take into account of the region-specific genomic characteristics that might have an impact on the measurements of the amount of methylated DNA (signal) and background fluctuation (noise). Thus, specific software needs to be developed for MBDCap-seq experiments. Results A new differential methylation quantification algorithm for MBDCap-seq, MBDDiff, was implemented. To evaluate the performance of the MBDDiff algorithm, a set of simulated signal based on negative binomial and Poisson distribution with parameters estimated from real MBDCap-seq datasets accompanied with different background noises were generated, and then performed against a set of commonly used algorithms for MBDCap-seq data analysis in terms of area under the ROC curve (AUC), number of false discoveries and statistical power. In addition, we also demonstrated the effective of MBDDiff algorithm to a set of in-house prostate cancer samples, endometrial cancer samples published earlier, and a set of public-domain triple negative breast cancer samples to identify potential factors that contribute to cancer development and recurrence. Conclusions In this paper we developed an algorithm, MBDDiff, designed specifically for datasets derived from MBDCap-seq. MBDDiff contains three modules: quality assessment of datasets and quantification of DNA methylation; determination of differential methylation of promoter regions; and visualization functionalities. Simulation results suggest that MBDDiff performs better compared to MEDIPS and DESeq in terms of AUC and the number of false discoveries at different levels of background noise. MBDDiff outperforms MEDIPS with increased backgrounds noise, but comparable performance when noise level is lower. By applying MBDDiff to several MBDCap-seq datasets, we were able to identify potential targets that contribute to the corresponding biological processes. Taken together, MBDDiff provides user an accurate differential methylation analysis for data generated by MBDCap-seq, especially under noisy conditions. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2794-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuanhang Liu
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Cellular and Structure Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Desiree Wilson
- Department of Cellular and Structure Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Robin J Leach
- Department of Cellular and Structure Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA. .,Department of Epidemiology & Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
19
|
Zhou H, Manthey J, Lioutikova E, Yang W, Yoshigoe K, Yang MQ, Wang H. The up-regulation of Myb may help mediate EGCG inhibition effect on mouse lung adenocarcinoma. Hum Genomics 2016; 10 Suppl 2:19. [PMID: 27461468 PMCID: PMC4965723 DOI: 10.1186/s40246-016-0072-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Green tea polyphenol epigallocatechin-3-gallate (EGCG) has been demonstrated to inhibit cancer in experimental studies through its antioxidant activity and modulations on cellular functions by binding specific proteins. By means of computational analysis and functional genomic approaches, we previously identified a set of protein coding genes and microRNAs whose expressions were significantly modulated in response to the EGCG treatment in tobacco carcinogen-induced lung adenocarcinoma in A/J mice. However, to what degree these genes are involved in the cancer inhibition of EGCG remains unclear. Results In this study, we further employed statistical methods and literature research to analyze these data in combination with The Cancer Genome Atlas (TCGA) lung adenocarcinoma datasets for additional data mining. Under the assumption that, if a gene mediates EGCG’s cancer inhibition, its expression level change caused by EGCG should be opposite to what occurred in the carcinogenesis, we identified Myb and Peg3 as the primary putative genes involved in the cancer inhibitory activity. Further analysis suggested that the regulation of Myb could be mediated through an EGCG-upregulated microRNA, miR-449c-5p. Conclusions Although the actions of EGCG involve multiple targets/pathways, further analysis by mining the existing genomic datasets revealed that the upregulations of Myb and Peg3 are likely the key anti-cancer events of EGCG in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s40246-016-0072-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Mathematical Science, School of Health and Natural Sciences, University of Saint Joseph, 1678 Asylum Avenue, West Hartford, CT, 06117, USA.
| | - Joseph Manthey
- Department of Mathematical Science, School of Health and Natural Sciences, University of Saint Joseph, 1678 Asylum Avenue, West Hartford, CT, 06117, USA
| | - Ekaterina Lioutikova
- Department of Mathematical Science, School of Health and Natural Sciences, University of Saint Joseph, 1678 Asylum Avenue, West Hartford, CT, 06117, USA
| | - William Yang
- Joint Bioinformatics Ph.D. Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - Kenji Yoshigoe
- Joint Bioinformatics Ph.D. Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, 2801 S. University Avenue, Little Rock, AR, 72204, USA
| | - Mary Qu Yang
- Joint Bioinformatics Ph.D. Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, 2801 S. University Avenue, Little Rock, AR, 72204, USA.
| | - Hong Wang
- Susan L. Cullman Laboratory for Cancer Research, Department of Chemical Biology and Centre for Cancer Prevention Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 164 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
20
|
Chen Z, Zhu S, Hong J, Soutto M, Peng D, Belkhiri A, Xu Z, El-Rifai W. Gastric tumour-derived ANGPT2 regulation by DARPP-32 promotes angiogenesis. Gut 2016; 65:925-34. [PMID: 25779598 PMCID: PMC4573388 DOI: 10.1136/gutjnl-2014-308416] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/27/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Overexpression of dopamine and cAMP-regulated phosphoprotein, Mr 32000 (DARPP-32), and its truncated isoform (t-DARPP) are associated with gastric tumorigenesis. Herein, we investigated the role of DARPP-32 proteins in regulating angiopoietin 2 (ANGPT2) and promoting tumour angiogenesis. DESIGN Quantitative real-time RT-PCR, immunoblotting, luciferase reporter, immunofluorescence, immunohistochemistry and angiogenesis assays were applied to investigate the regulation of angiogenesis by DARPP-32 proteins. RESULTS Overexpression of DARPP-32 significantly increased the mRNA and protein levels of ANGPT2 in gastric cancer cells. The overexpression of DARPP-32 T34A mutant or the N-terminal truncated isoform, t-DARPP, led to similar effects ruling out the T34-dependent regulation of protein phosphatase 1 activity in regulating ANGPT2. DARPP-32 proteins induced a secreted form of ANGPT2, which was detectable in the media, functionally active, and able to induce angiogenesis, measured by the human umbilical vein endothelial cells tube formation assay. Antibody blocking of the secreted ANGPT2 abrogated its function. To identify the mechanism by which DARPP-32 regulates ANGPT2, we examined the activities of NF-κB and signal transducer and activator of transcription 3 (STAT3), known regulators of angiogenesis. The results ruled out NF-κB and showed induction of STAT3 phosphorylation, activation and nuclear localisation. Inhibition or knockdown of STAT3 significantly attenuated the induction of ANGPT2 by DARPP-32 proteins. In vivo xenograft models demonstrated that overexpression of DARPP-32 promotes angiogenesis and tumour growth. Analyses of human gastric cancer tissues showed a strong correlation between DARPP-32 and ANGPT2. CONCLUSIONS Our novel findings establish the role of DARPP-32-STAT3 axis in regulating ANGPT2 in cancer cells to promote angiogenesis and tumorigenesis.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shoumin Zhu
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jun Hong
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mohammed Soutto
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - DunFa Peng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wael El-Rifai
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
21
|
Kumar A, Coleman I, Morrissey C, Zhang X, True LD, Gulati R, Etzioni R, Bolouri H, Montgomery B, White T, Lucas JM, Brown LG, Dumpit RF, DeSarkar N, Higano C, Yu EY, Coleman R, Schultz N, Fang M, Lange PH, Shendure J, Vessella RL, Nelson PS. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat Med 2016; 22:369-78. [PMID: 26928463 PMCID: PMC5045679 DOI: 10.1038/nm.4053] [Citation(s) in RCA: 546] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/01/2016] [Indexed: 12/17/2022]
Abstract
Tumor heterogeneity may reduce the efficacy of molecularly guided systemic therapy for cancers that have metastasized. To determine whether the genomic alterations in a single metastasis provide a reasonable assessment of the major oncogenic drivers of other dispersed metastases in an individual, we analyzed multiple tumors from men with disseminated prostate cancer through whole-exome sequencing, array comparative genomic hybridization (CGH) and RNA transcript profiling, and we compared the genomic diversity within and between individuals. In contrast to the substantial heterogeneity between men, there was limited diversity among metastases within an individual. The number of somatic mutations, the burden of genomic copy number alterations and aberrations in known oncogenic drivers were all highly concordant, as were metrics of androgen receptor (AR) activity and cell cycle activity. AR activity was inversely associated with cell proliferation, whereas the expression of Fanconi anemia (FA)-complex genes was correlated with elevated cell cycle progression, expression of the E2F transcription factor 1 (E2F1) and loss of retinoblastoma 1 (RB1). Men with somatic aberrations in FA-complex genes or in ATM serine/threonine kinase (ATM) exhibited significantly longer treatment-response durations to carboplatin than did men without defects in genes encoding DNA-repair proteins. Collectively, these data indicate that although exceptions exist, evaluating a single metastasis provides a reasonable assessment of the major oncogenic driver alterations that are present in disseminated tumors within an individual, and thus may be useful for selecting treatments on the basis of predicted molecular vulnerabilities.
Collapse
Affiliation(s)
- Akash Kumar
- Department of Genome Sciences, University of Washington, 3720 15 Ave. NE, Seattle, WA
| | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Colm Morrissey
- Department of Urology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Xiaotun Zhang
- Department of Urology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Lawrence D. True
- Department of Pathology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Hamid Bolouri
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Bruce Montgomery
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Thomas White
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Jared M. Lucas
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Lisha G. Brown
- Department of Urology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Ruth F. Dumpit
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Navonil DeSarkar
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Celestia Higano
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Evan Y. Yu
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Roger Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Min Fang
- Department of Pathology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| | - Paul H. Lange
- Department of Urology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, 3720 15 Ave. NE, Seattle, WA
| | - Robert L. Vessella
- Department of Urology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
| | - Peter S. Nelson
- Department of Genome Sciences, University of Washington, 3720 15 Ave. NE, Seattle, WA
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
- Department of Urology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
- Department of Pathology, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA
| |
Collapse
|
22
|
Cho WJ, Oliveira DSM, Najy AJ, Mainetti LE, Aoun HD, Cher ML, Heath E, Kim HRC, Bonfil RD. Gene expression analysis of bone metastasis and circulating tumor cells from metastatic castrate-resistant prostate cancer patients. J Transl Med 2016; 14:72. [PMID: 26975354 PMCID: PMC4791970 DOI: 10.1186/s12967-016-0829-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/05/2016] [Indexed: 01/15/2023] Open
Abstract
Background Characterization of genes linked to bone metastasis is critical for identification of novel prognostic or predictive biomarkers and potential therapeutic targets in metastatic castrate-resistant prostate cancer (mCRPC). Although bone marrow core biopsies (BMBx) can be obtained for gene profiling, the procedure itself is invasive and uncommon practice in mCRPC patients. Conversely, circulating tumor cells (CTCs), which are likely to stem from bone metastases, can be isolated from blood. The goals of this exploratory study were to establish a sensitive methodology to analyze gene expression in BMBx and CTCs, and to determine whether the presence or absence of detectable gene expression is concordant in matching samples from mCRPC patients. Methods The CellSearch® platform was used to enrich and enumerate CTCs. Low numbers of PC3 prostate cancer (PCa) cells were spiked into normal blood to assess cell recovery rate. RNA extracted from recovered PC3 cells was amplified using an Eberwine-based procedure to obtain antisense mRNA (aRNA), and assess the linearity of the RNA amplification method. In this pilot study, RNAs extracted from CTCs and PCa cells microdissected from formalin-fixed paraffin-embedded BMBx, were amplified to obtain aRNA and assess the expression of eight genes functionally relevant to PCa bone metastasis using RT-PCR. Results RNAs were successfully extracted from as few as 1–5 PCa cells in blood samples. The relative expression levels of reference genes were maintained after RNA amplification. The integrity of the amplified RNA was also demonstrated by RT-PCR analysis using primer sets that target the 5′-end, middle, and 3′-end of reference mRNA. We found that in 21 out of 28 comparisons, the presence or absence of detectable gene expression in CTCs and PCa cells microdissected from single bone lesions of the same patients was concordant. Conclusions This exploratory analysis suggests that aRNA amplification through in vitro transcription may be useful as a method to detect gene expression in small numbers of CTCs and tumor cells microdissected from bone metastatic lesions. In some cases, gene expression in CTCs and BMBxs was not concordant, raising questions about using CTC gene expression to make clinical decisions. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0829-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Won Jin Cho
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA
| | - Daniel S M Oliveira
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA
| | - Abdo J Najy
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Leandro E Mainetti
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA
| | - Hussein D Aoun
- Department of Radiology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Michael L Cher
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA.,Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Elisabeth Heath
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - Hyeong-Reh C Kim
- Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA
| | - R Daniel Bonfil
- Department of Urology, Wayne State University School of Medicine and Karmanos Cancer Institute, 540 E. Canfield, Scott Hall # 9105, Detroit, MI, 4820, USA. .,Department of Pathology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
23
|
Chen L, Yang J, Huang T, Kong X, Lu L, Cai YD. Mining for novel tumor suppressor genes using a shortest path approach. J Biomol Struct Dyn 2015. [PMID: 26209080 DOI: 10.1080/07391102.2015.1042915] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cancer, being among the most serious diseases, causes many deaths every year. Many investigators have devoted themselves to designing effective treatments for this disease. Cancer always involves abnormal cell growth with the potential to invade or spread to other parts of the body. In contrast, tumor suppressor genes (TSGs) act as guardians to prevent a disordered cell cycle and genomic instability in normal cells. Studies on TSGs can assist in the design of effective treatments against cancer. In this study, we propose a computational method to discover potential TSGs. Based on the known TSGs, a number of candidate genes were selected by applying the shortest path approach in a weighted graph that was constructed using protein-protein interaction network. The analysis of selected genes shows that some of them are new TSGs recently reported in the literature, while others may be novel TSGs.
Collapse
Affiliation(s)
- Lei Chen
- a College of Life Science , Shanghai University , Shanghai 200444 , P.R. China.,b College of Information Engineering , Shanghai Maritime University , Shanghai 201306 , P.R. China
| | - Jing Yang
- c The Key Laboratory of Stem Cell Biology , Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) , Shanghai 200025 , P.R. China
| | - Tao Huang
- c The Key Laboratory of Stem Cell Biology , Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) , Shanghai 200025 , P.R. China
| | - Xiangyin Kong
- c The Key Laboratory of Stem Cell Biology , Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) , Shanghai 200025 , P.R. China
| | - Lin Lu
- d Department of Radiology , Columbia University Medical Center , New York , NY 10032 , USA
| | - Yu-Dong Cai
- a College of Life Science , Shanghai University , Shanghai 200444 , P.R. China
| |
Collapse
|
24
|
Wu Y, Schoenborn JR, Morrissey C, Xia J, Larson S, Brown LG, Qu X, Lange PH, Nelson PS, Vessella RL, Fang M. High-Resolution Genomic Profiling of Disseminated Tumor Cells in Prostate Cancer. J Mol Diagn 2015; 18:131-43. [PMID: 26607774 DOI: 10.1016/j.jmoldx.2015.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 08/18/2015] [Accepted: 08/25/2015] [Indexed: 12/27/2022] Open
Abstract
Circulating tumor cells and disseminated tumor cells (DTCs) are of great interest because they provide a minimally invasive window for assessing aspects of cancer biology, including tumor heterogeneity, a means to discover biomarkers of disease behavior, and a way to identify and prioritize therapeutic targets in the emerging era of precision oncology. However, the rarity of circulating tumor cells and DTCs poses a substantial challenge to the consistent success in analyzing their molecular features, including genomic aberrations. Herein, we describe optimized and robust methods to reproducibly detect genomic copy number alterations in samples of 2 to 40 cells after whole-genome amplification with the use of a high-resolution single-nuclear polymorphism-array platform and refined computational algorithms. We have determined the limit of detection for heterogeneity within a sample as 50% and also demonstrated success in analyzing single cells. We validated the genes in genomic regions that are frequently amplified or deleted by real-time quantitative PCR and nCounter copy number quantification. We further applied these methods to DTCs isolated from individuals with advanced prostate cancer to confirm their highly aberrant nature. We compared copy number alterations of DTCs with matched metastatic tumors isolated from the same individual to gain biological insight. These developments provide high-resolution genomic profiling of single and rare cell populations and should be applicable to a wide-range of sample sources.
Collapse
Affiliation(s)
- Yu Wu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jamie R Schoenborn
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Jing Xia
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sandy Larson
- Department of Urology, University of Washington, Seattle, Washington
| | - Lisha G Brown
- Department of Urology, Puget Sound VA Health Care System, Seattle, Washington
| | - Xiaoyu Qu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Paul H Lange
- Department of Urology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Urology, University of Washington, Seattle, Washington
| | - Robert L Vessella
- Department of Urology, University of Washington, Seattle, Washington; Department of Urology, Puget Sound VA Health Care System, Seattle, Washington
| | - Min Fang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Urology, University of Washington, Seattle, Washington.
| |
Collapse
|
25
|
Swartz MF, Simon B, Atallah-Yunes N, Cholette JM, Orie J, Gensini F, Alfieris GM. Distal Transverse Arch to Left Carotid Artery Ratio Helps to Identify Infants With Aortic Arch Hypoplasia. Ann Thorac Surg 2015. [PMID: 26212512 DOI: 10.1016/j.athoracsur.2015.04.107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Aortic coarctation (CoA) with concomitant aortic arch hypoplasia (AAH) is associated with an increased risk of hypertension after surgical repair. The differentiation of CoA with or without AAH may be critical to delineate the ideal surgical approach that best ameliorates postoperative hypertension. Since 2000, we have defined CoA with AAH when the diameter of the distal transverse aortic arch is equal to or less than the diameter of the left carotid artery. We hypothesized that, based on our definition, aortic tissue from infants having CoA with AAH would demonstrate distinct genetic expression patterns as compared with infants having CoA alone. METHODS From 6 infants (AAH, 3; CoA, 3), an Affymetrix 1.0 genome array identified genes in the coarctation/arch region that were differentially expressed between infants having CoA with AAH versus CoA alone. Reverse transcription polymerase chain reaction validated genetic differences from a cohort of 21 infants (CoA with AAH, 10; CoA, 11). To evaluate the clinical outcomes based on our definition of CoA with AAH, we reviewed infants repaired using this algorithm from 2000 to 2010. RESULTS Microarray data demonstrated genes differentially expressed between groups. Reverse transcription polymerase chain reaction confirmed that CoA with AAH was associated with an increased expression of genes involved in cardiac and vascular development and growth, including hepsin, fibroblast growth factor-18, and T-box 2. The clinical outcomes of 79 infants (AAH, 26; CoA, 53) demonstrated that 90.1% were free of hypertension at 13 years when managed with this surgical strategy. CONCLUSIONS These findings provide evidence that the ratio of the diameter of the distal transverse arch to the left carotid artery may be helpful to identify CoA with AAH and, when used to delineate the surgical approach, may minimize hypertension.
Collapse
Affiliation(s)
- Michael F Swartz
- Pediatric Cardiac Consortium of Upstate New York, Rochester, New York; University of Rochester Medical Center, Strong Memorial Hospital, Rochester, New York.
| | - Bartholomew Simon
- University of Rochester Medical Center, Strong Memorial Hospital, Rochester, New York
| | - Nader Atallah-Yunes
- Pediatric Cardiac Consortium of Upstate New York, Rochester, New York; University of Rochester Medical Center, Strong Memorial Hospital, Rochester, New York
| | - Jill M Cholette
- Pediatric Cardiac Consortium of Upstate New York, Rochester, New York; University of Rochester Medical Center, Strong Memorial Hospital, Rochester, New York
| | - Joseph Orie
- Pediatric Cardiac Consortium of Upstate New York, Rochester, New York
| | - Francisco Gensini
- Pediatric Cardiac Consortium of Upstate New York, Rochester, New York; University of Rochester Medical Center, Strong Memorial Hospital, Rochester, New York
| | - George M Alfieris
- Pediatric Cardiac Consortium of Upstate New York, Rochester, New York; University of Rochester Medical Center, Strong Memorial Hospital, Rochester, New York
| |
Collapse
|
26
|
Xu Z, Chen H, Liu D, Huo J. Fibulin-1 is downregulated through promoter hypermethylation in colorectal cancer: a CONSORT study. Medicine (Baltimore) 2015; 94:e663. [PMID: 25837757 PMCID: PMC4554035 DOI: 10.1097/md.0000000000000663] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Fibulin-1 (FBLN1) is involved in the progression of some types of cancer. However, the role of FBLN1 in colorectal cancer (CRC) has not been examined. The purpose of this study was to understand the molecular mechanisms and clinical significance of FBLN1 inactivation in CRC. The expression of FBLN1 in CRC tissues and adjacent normal tissues was analyzed by immunohistochemical analysis and quantitative real-time polymerase chain reaction (qRT-PCR). Methylation-specific polymerase chain reaction (MSP) and bisulfite sequencing PCR (BSP) were performed to examine the methylation status of the FBLN1 gene promoter. Furthermore, the methylated level of FBLN1 was analyzed with the clinicopathological characteristics. Immunohistochemical analysis and qRT-PCR analysis showed that FBLN1 protein and messenger RNA (mRNA) levels in tumor tissues were both significantly decreased compared with that in adjacent nontumor tissues. The methylation rate of FBLN1 promoter was significantly higher in CRC tissues than that in adjacent nontumor tissues (P < 0.001). In addition, the correlation between FBLN1 hypermethylation, protein expression, and overall survival (OS) was statistically significant. Our results indicated that the FBLN1 gene may be a novel candidate of tumor suppressor gene in CRC, and that promoter hypermethylation of FBLN1 is an important reason for its downregulation and is also a good predictor of OS for CRC.
Collapse
Affiliation(s)
- Zhiying Xu
- From the Department of Gastroenterology (ZX, DL, JH), 2nd Xiangya Hospital, Central South University, Changsha, Hunan; and Department of Gastroenterology (ZX, HC), People's Hospital of Taizhou, Taizhou, Jiangsu, China
| | | | | | | |
Collapse
|
27
|
Kamińska K, Szczylik C, Bielecka ZF, Bartnik E, Porta C, Lian F, Czarnecka AM. The role of the cell-cell interactions in cancer progression. J Cell Mol Med 2015; 19:283-96. [PMID: 25598217 PMCID: PMC4407603 DOI: 10.1111/jcmm.12408] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 07/18/2014] [Indexed: 12/11/2022] Open
Abstract
In the field of cancer research, scientific investigations are based on analysing differences in the secretome, the proteome, the transcriptome, the expression of cell surface molecules, and the deregulation of signal transduction pathways between neoplastic and normal cells. Accumulating evidence indicates a crucial role in carcinogenesis concerning not only stromal cells but also normal cells from target organs and tissue where tumours emerge. The tumour microenvironment (TME) definitively plays an important role in regulating neighbouring cell behaviour. To date, limited attention has been focused upon interactions between cancer cells and normal cells. This review concentrates on the interactions between stromal and healthy cells from the TME in cancer development. In the article, the authors also describe mutations, genes and proteins expression pattern that are involved in tumour development in target organ.
Collapse
Affiliation(s)
- Katarzyna Kamińska
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| | | | | | | | | | | | | |
Collapse
|
28
|
Xiao W, Wang J, Li H, Xia D, Yu G, Yao W, Yang Y, Xiao H, Lang B, Ma X, Guo X, Guan W, Xu H, Liu J, Zhang X, Ye Z. Fibulin-1 is epigenetically down-regulated and related with bladder cancer recurrence. BMC Cancer 2014; 14:677. [PMID: 25234557 PMCID: PMC4180143 DOI: 10.1186/1471-2407-14-677] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/10/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Bladder cancer is one of the most common cancers worldwide. Fibulin-1, a multi-functional extracellular matrix protein, has been demonstrated to be involved in many kinds of cancers, while its function in bladder cancer remains unclear. So here we investigated the expression and function of fibulin-1 in Bladder cancer. METHODS We used real-time PCR, Western blot analysis and immunohistochemistry to determine the expression of fibulin-1 in Bladder cancer cells and patient tissues respectively. Methylation-specific PCR and quantitative sequencing were used to examine the methylation status of FBLN1 gene promoter. Eukaryotic expression plasmid and lentiviral vector were used to overexpress fibulin-1 in Bladder cancer cells 5637, HT-1376 to investigate its function in vitro and in vivo. RESULTS We identified that fibulin-1 was significantly down-regulated in bladder cancer, and its dysregulation was associated with non-muscle-invasive bladder cancer (NMIBC) grade and recurrence. The promoter region of FBLN1 was generally methylated in bladder cancer cell lines and tissues, further investigation in patient tissues showed that the methylation status was associated with the fibulin-1 expression. Overexpression of fibulin-1 significantly suppressed tumor growth, induced tumor cell apoptosis, decreased cell motility, and inhibited angiogenesis in cultured bladder cancer cells and xenograft tumor in nude mice. CONCLUSIONS Altogether, our results indicated that fibulin-1 expression is associated with NMIBC grade and recurrence, it is epigenetically down-regulated and functions as a tumor suppressor gene and angiogenesis inhibitor in bladder cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | | | | | | |
Collapse
|
29
|
Costa FC, Saito A, Gonçalves KA, Vidigal PM, Meirelles GV, Bressan GC, Kobarg J. Ki-1/57 and CGI-55 ectopic expression impact cellular pathways involved in proliferation and stress response regulation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2944-56. [PMID: 25205453 DOI: 10.1016/j.bbamcr.2014.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 08/27/2014] [Accepted: 08/28/2014] [Indexed: 10/24/2022]
Abstract
Ki-1/57 (HABP4) and CGI-55 (SERBP1) are regulatory proteins and paralogs with 40.7% amino acid sequence identity and 67.4% similarity. Functionally, they have been implicated in the regulation of gene expression on both the transcriptional and mRNA metabolism levels. A link with tumorigenesis is suggested, since both paralogs show altered expression levels in tumor cells and the Ki-1/57 gene is found in a region of chromosome 9q that represents a haplotype for familiar colon cancer. However, the target genes regulated by Ki-1/57 and CGI-55 are unknown. Here, we analyzed the alterations of the global transcriptome profile after Ki-1/57 or CGI-55 overexpression in HEK293T cells by DNA microchip technology. We were able to identify 363 or 190 down-regulated and 50 or 27 up-regulated genes for Ki-1/57 and CGI-55, respectively, of which 20 were shared between both proteins. Expression levels of selected genes were confirmed by qRT-PCR both after protein overexpression and siRNA knockdown. The majority of the genes with altered expression were associated to proliferation, apoptosis and cell cycle control processes, prompting us to further explore these contexts experimentally. We observed that overexpression of Ki-1/57 or CGI-55 results in reduced cell proliferation, mainly due to a G1 phase arrest, whereas siRNA knockdown of CGI-55 caused an increase in proliferation. In the case of Ki-1/57 overexpression, we found protection from apoptosis after treatment with the ER-stress inducer thapsigargin. Together, our data give important new insights that may help to explain these proteins putative involvement in tumorigenic events.
Collapse
Affiliation(s)
- Fernanda C Costa
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil.
| | - Angela Saito
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil; Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Kaliandra A Gonçalves
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil; Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Pedro M Vidigal
- Laboratório de Bioinformática, Instituto de Biotecnologia Aplicada à Agropecuária-BIOAGRO, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brasil.
| | - Gabriela V Meirelles
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil.
| | - Gustavo C Bressan
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil; Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Jörg Kobarg
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, Brasil; Departamento de Bioquímica-Programa de Pós-graduação em Biologia Funcional e Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil; Departamento de Genética, Evolução e Bioagentes - Programa de Pós-graduação em Genética e Biologia Molecular, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| |
Collapse
|
30
|
Nabhan C, Patel A, Villines D, Tolzien K, Kelby SK, Lestingi TM. Lenalidomide Monotherapy in Chemotherapy-Naive, Castration-Resistant Prostate Cancer Patients: Final Results of a Phase II Study. Clin Genitourin Cancer 2014; 12:27-32. [DOI: 10.1016/j.clgc.2013.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 08/30/2013] [Accepted: 09/04/2013] [Indexed: 11/29/2022]
|
31
|
Mengele K, Napieralski R, Magdolen V, Reuning U, Gkazepis A, Sweep F, Brünner N, Foekens J, Harbeck N, Schmitt M. Characteristics of the level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn 2014; 10:947-62. [DOI: 10.1586/erm.10.73] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
32
|
Wu BJ, Zhou ZP, Li WP, Ding W, Wu YZ, Zhou ZW, Zhang RQ, Liu QF, Jiang H. Abnormal hypermethylation and clinicopathological significance of FBLN1 gene in cutaneous melanoma. Tumour Biol 2014; 35:123-7. [PMID: 23907575 DOI: 10.1007/s13277-013-1015-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/11/2013] [Indexed: 12/18/2022] Open
Abstract
Fibulin-1 (FBLN1) is involved in the progression of some types of cancer. However, the role of FBLN1 in cutaneous melanoma (CM) has not been examined. The purpose of this study was to understand the molecular mechanisms and clinical significance of FBLN1 inactivation in CM. The expression of FBLN1 mRNA in CM tissues and adjacent normal skin tissues was analyzed by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR). Methylation-specific polymerase chain reaction was performed to examine the methylation status of the FBLN1 gene promoter. Furthermore, the methylation status of FBLN1 was analyzed with the clinicopathological characteristics and overall survival. qRT-PCR showed FBLN1 mRNA levels in cancerous tissues to be significantly decreased compared with that in adjacent normal skin tissues. The rate of FBLN1 promoter methylation was significantly higher in CM tissues than in adjacent normal skin tissues (P < 0.001). Downregulation of FBLN1 was strongly correlated with promoter methylation (P = 0.021). Promoter hypermethylation of FBLN1 was significantly associated with tumor stage (P = 0.019). In addition, FBLN1 methylation status was associated with significantly shorter survival time and was an independent predictor of overall survival. In conclusion, our results indicated that FBLN1 is a novel candidate of tumor suppressor gene and that promoter hypermethylation of FBLN1 is associated with tumor progression in CM.
Collapse
Affiliation(s)
- Bao-Jin Wu
- Department of Plastic Surgery, Huashan Hospital, Fudan University, No. 12, Wulumuqi Zhong Rd., Shanghai, 200040, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Qu X, Randhawa G, Friedman C, Kurland BF, Glaskova L, Coleman I, Mostaghel E, Higano CS, Porter C, Vessella R, Nelson PS, Fang M. A three-marker FISH panel detects more genetic aberrations of AR, PTEN and TMPRSS2/ERG in castration-resistant or metastatic prostate cancers than in primary prostate tumors. PLoS One 2013; 8:e74671. [PMID: 24098661 PMCID: PMC3787014 DOI: 10.1371/journal.pone.0074671] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 08/04/2013] [Indexed: 12/31/2022] Open
Abstract
TMPRSS2/ERG rearrangement, PTEN gene deletion, and androgen receptor (AR) gene amplification have been observed in various stages of human prostate cancer. We hypothesized that using these markers as a combined panel would allow better differentiation between low-risk and high-risk prostate cancer. We analyzed 110 primary prostate cancer samples, 70 metastatic tumor samples from 11 patients, and 27 xenograft tissues derived from 22 advanced prostate cancer patients using fluorescence in situ hybridization (FISH) analysis with probes targeting the TMPRSS2/ERG, PTEN, and AR gene loci. Heterogeneity of the aberrations detected was evaluated. Genetic patterns were also correlated with transcript levels. Among samples with complete data available, the three-marker FISH panel detected chromosomal abnormalities in 53% of primary prostate cancers and 87% of metastatic (Met) or castration-resistant (CRPC) tumors. The number of markers with abnormal FISH result had a different distribution between the two groups (P<0.001). At the patient level, Met/CRPC tumors are 4.5 times more likely to show abnormalities than primary cancer patients (P<0.05). Heterogeneity among Met/CRPC tumors is mostly inter-patient. Intra-patient heterogeneity is primarily due to differences between the primary prostate tumor and the metastases while multiple metastatic sites show consistent abnormalities. Intra-tumor variability is most prominent with the AR copy number in primary tumors. AR copy number correlated well with the AR mRNA expression (rho = 0.52, P<0.001). Especially among TMPRSS2:ERG fusion-positive CRPC tumors, AR mRNA and ERG mRNA levels are strongly correlated (rho = 0.64, P<0.001). Overall, the three-marker FISH panel may represent a useful tool for risk stratification of prostate cancer patients.
Collapse
Affiliation(s)
- Xiaoyu Qu
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Grace Randhawa
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Cynthia Friedman
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Brenda F. Kurland
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lena Glaskova
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Elahe Mostaghel
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Celestia S. Higano
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Christopher Porter
- Virginia Mason Medical Center, Seattle, Washington, United States of America
| | - Robert Vessella
- University of Washington, Seattle, Washington, United States of America
- Puget Sound VA Health Care System, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
| | - Min Fang
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Seattle Cancer Care Alliance, Seattle, Washington, United States of America
- University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
34
|
Langenkamp E, Kamps JAAM, Mrug M, Verpoorte E, Niyaz Y, Horvatovich P, Bischoff R, Struijker-Boudier H, Molema G. Innovations in studying in vivo cell behavior and pharmacology in complex tissues--microvascular endothelial cells in the spotlight. Cell Tissue Res 2013; 354:647-69. [PMID: 24072341 DOI: 10.1007/s00441-013-1714-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/18/2013] [Indexed: 02/06/2023]
Abstract
Many studies on the molecular control underlying normal cell behavior and cellular responses to disease stimuli and pharmacological intervention are conducted in single-cell culture systems, while the read-out of cellular engagement in disease and responsiveness to drugs in vivo is often based on overall tissue responses. As the majority of drugs under development aim to specifically interact with molecular targets in subsets of cells in complex tissues, this approach poses a major experimental discrepancy that prevents successful development of new therapeutics. In this review, we address the shortcomings of the use of artificial (single) cell systems and of whole tissue analyses in creating a better understanding of cell engagement in disease and of the true effects of drugs. We focus on microvascular endothelial cells that actively engage in a wide range of physiological and pathological processes. We propose a new strategy in which in vivo molecular control of cells is studied directly in the diseased endothelium instead of at a (far) distance from the site where drugs have to act, thereby accounting for tissue-controlled cell responses. The strategy uses laser microdissection-based enrichment of microvascular endothelium which, when combined with transcriptome and (phospho)proteome analyses, provides a factual view on their status in their complex microenvironment. Combining this with miniaturized sample handling using microfluidic devices enables handling the minute sample input that results from this strategy. The multidisciplinary approach proposed will enable compartmentalized analysis of cell behavior and drug effects in complex tissue to become widely implemented in daily biomedical research and drug development practice.
Collapse
Affiliation(s)
- Elise Langenkamp
- University Medical Center Groningen, Department of Pathology and Medical Biology, Medical Biology section, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hudson BD, Kulp KS, Loots GG. Prostate cancer invasion and metastasis: insights from mining genomic data. Brief Funct Genomics 2013; 12:397-410. [PMID: 23878130 DOI: 10.1093/bfgp/elt021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed malignancy in men in the Western world and the second leading cause of cancer-related deaths among men worldwide. Although most cancers have the potential to metastasize under appropriate conditions, PCa favors the skeleton as a primary site of metastasis, suggesting that the bone microenvironment is conducive to its growth. PCa metastasis proceeds through a complex series of molecular events that include angiogenesis at the site of the original tumor, local migration within the primary site, intravasation into the blood stream, survival within the circulation, extravasation of the tumor cells to the target organ and colonization of those cells within the new site. In turn, each one of these steps involves a complicated chain of events that utilize multiple protein-protein interactions, protein signaling cascades and transcriptional changes. Despite the urgent need to improve current biomarkers for diagnosis, prognosis and drug resistance, advances have been slow. Global gene expression methods such as gene microarrays and RNA sequencing enable the study of thousands of genes simultaneously and allow scientists to examine molecular pathways of cancer pathogenesis. In this review, we summarize the current literature that explored high-throughput transcriptome analysis toward the advancement of biomarker discovery for PCa. Novel biomarkers are strongly needed to enable more accurate detection of PCa, improve prediction of tumor aggressiveness and facilitate the discovery of new therapeutic targets for tailored medicine. Promising molecular markers identified from gene expression profiling studies include HPN, CLU1, WT1, WNT5A, AURKA and SPARC.
Collapse
Affiliation(s)
- Bryan D Hudson
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA.
| | | | | |
Collapse
|
36
|
Xiao W, Wang J, Li H, Guan W, Xia D, Yu G, Xiao H, Lang B, Ma X, Liu J, Zhang X, Ye Z, Xu H. Fibulin-1 is Down-Regulated Through Promoter Hypermethylation and Suppresses Renal Cell Carcinoma Progression. J Urol 2013; 190:291-301. [PMID: 23391467 DOI: 10.1016/j.juro.2013.01.098] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 01/30/2013] [Indexed: 11/20/2022]
Affiliation(s)
- Wei Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Androgen receptor promotes ligand-independent prostate cancer progression through c-Myc upregulation. PLoS One 2013; 8:e63563. [PMID: 23704919 PMCID: PMC3660401 DOI: 10.1371/journal.pone.0063563] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/02/2013] [Indexed: 01/27/2023] Open
Abstract
The androgen receptor (AR) is the principal therapeutic target in prostate cancer. For the past 70 years, androgen deprivation therapy (ADT) has been the major therapeutic focus. However, some patients do not benefit, and those tumors that do initially respond to ADT eventually progress. One recently described mechanism of such an effect is growth and survival-promoting effects of the AR that are exerted independently of the AR ligands, testosterone and dihydrotestosterone. However, specific ligand-independent AR target genes that account for this effect were not well characterized. We show here that c-Myc, which is a key mediator of ligand-independent prostate cancer growth, is a key ligand-independent AR target gene. Using microarray analysis, we found that c-Myc and AR expression levels strongly correlated with each other in tumors from patients with castration-resistant prostate cancer (CRPC) progressing despite ADT. We confirmed that AR directly regulates c-Myc transcription in a ligand-independent manner, that AR and c-Myc suppression reduces ligand-independent prostate cancer cell growth, and that ectopic expression of c-Myc attenuates the anti-growth effects of AR suppression. Importantly, treatment with the bromodomain inhibitor JQ1 suppressed c-Myc function and suppressed ligand-independent prostate cancer cell survival. Our results define a new link between two critical proteins in prostate cancer – AR and c-Myc – and demonstrate the potential of AR and c-Myc-directed therapies to improve prostate cancer control.
Collapse
|
38
|
Boyce BF. Advances in osteoclast biology reveal potential new drug targets and new roles for osteoclasts. J Bone Miner Res 2013; 28:711-22. [PMID: 23436579 PMCID: PMC3613781 DOI: 10.1002/jbmr.1885] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/22/2013] [Accepted: 01/28/2013] [Indexed: 01/06/2023]
Abstract
Osteoclasts are multinucleated myeloid lineage cells formed in response to macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) by fusion of bone marrow-derived precursors that circulate in the blood and are attracted to sites of bone resorption in response to factors, such as sphingosine-1 phosphate signaling. Major advances in understanding of the molecular mechanisms regulating osteoclast functions have been made in the past 20 years, mainly from mouse and human genetic studies. These have revealed that osteoclasts express and respond to proinflammatory and anti-inflammatory cytokines. Some of these cytokines activate NF-κB and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) signaling to induce osteoclast formation and activity and also regulate communication with neighboring cells through signaling proteins, including ephrins and semaphorins. Osteoclasts also positively and negatively regulate immune responses and osteoblastic bone formation. These advances have led to development of new inhibitors of bone resorption that are in clinical use or in clinical trials; and more should follow, based on these advances. This article reviews current understanding of how bone resorption is regulated both positively and negatively in normal and pathologic states.
Collapse
Affiliation(s)
- Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
39
|
Tan IDA, Ricciardelli C, Russell DL. The metalloproteinase ADAMTS1: a comprehensive review of its role in tumorigenic and metastatic pathways. Int J Cancer 2013; 133:2263-76. [PMID: 23444028 DOI: 10.1002/ijc.28127] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/14/2013] [Indexed: 01/07/2023]
Abstract
As it was first characterized in 1997, the ADAMTS (A Disintegrin and Metalloprotease with ThromboSpondin motifs) metalloprotease family has been associated with many physiological and pathological conditions. Of the 19 proteases belonging to this family, considerable attention has been devoted to the role of its first member ADAMTS1 in cancer. Elevated ADAMTS1 promotes pro-tumorigenic changes such as increased tumor cell proliferation, inhibited apoptosis and altered vascularization. Importantly, it facilitates significant peritumoral remodeling of the extracellular matrix environment to promote tumor progression and metastasis. However, discrepancy exists, as several studies also depict ADAMTS1 as a tumor suppressor. This article reviews the current understanding of ADAMTS1 regulation and the consequence of its dysregulation in primary cancer and ADAMTS1-mediated pathways of cancer progression and metastasis.
Collapse
Affiliation(s)
- Izza de Arao Tan
- Robinson Institute, School of Paediatrics and Reproductive Health, Department of Obstetrics and Gynaecology, Univeristy of Adelaide, South Australia, Australia
| | | | | |
Collapse
|
40
|
Haiman CA, Han Y, Feng Y, Xia L, Hsu C, Sheng X, Pooler LC, Patel Y, Kolonel LN, Carter E, Park K, Le Marchand L, Van Den Berg D, Henderson BE, Stram DO. Genome-wide testing of putative functional exonic variants in relationship with breast and prostate cancer risk in a multiethnic population. PLoS Genet 2013; 9:e1003419. [PMID: 23555315 PMCID: PMC3610631 DOI: 10.1371/journal.pgen.1003419] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 02/12/2013] [Indexed: 12/19/2022] Open
Abstract
Rare variation in protein coding sequence is poorly captured by GWAS arrays and has been hypothesized to contribute to disease heritability. Using the Illumina HumanExome SNP array, we successfully genotyped 191,032 common and rare non-synonymous, splice site, or nonsense variants in a multiethnic sample of 2,984 breast cancer cases, 4,376 prostate cancer cases, and 7,545 controls. In breast cancer, the strongest associations included either SNPs in or gene burden scores for genes LDLRAD1, SLC19A1, FGFBP3, CASP5, MMAB, SLC16A6, and INS-IGF2. In prostate cancer, one of the most associated SNPs was in the gene GPRC6A (rs2274911, Pro91Ser, OR = 0.88, P = 1.3 × 10(-5)) near to a known risk locus for prostate cancer; other suggestive associations were noted in genes such as F13A1, ANXA4, MANSC1, and GP6. For both breast and prostate cancer, several of the most significant associations involving SNPs or gene burden scores (sum of minor alleles) were noted in genes previously reported to be associated with a cancer-related phenotype. However, only one of the associations (rs145889899 in LDLRAD1, p = 2.5 × 10(-7) only seen in African Americans) for overall breast or prostate cancer risk was statistically significant after correcting for multiple comparisons. In addition to breast and prostate cancer, other cancer-related traits were examined (body mass index, PSA level, and alcohol drinking) with a number of known and potentially novel associations described. In general, these findings do not support there being many protein coding variants of moderate to high risk for breast and prostate cancer with odds ratios over a range that is probably required for protein coding variation to play a truly outstanding role in risk heritability. Very large sample sizes will be required to better define the role of rare and less penetrant coding variation in prostate and breast cancer disease genetics.
Collapse
Affiliation(s)
- Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Ying Han
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Ye Feng
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Lucy Xia
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Chris Hsu
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Xin Sheng
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Loreall C. Pooler
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Yesha Patel
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Laurence N. Kolonel
- Epidemiology Program, Cancer Research Center, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Erin Carter
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Karen Park
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Loic Le Marchand
- Epidemiology Program, Cancer Research Center, University of Hawaii, Honolulu, Hawaii, United States of America
| | - David Van Den Berg
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Daniel O. Stram
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
41
|
Eph receptors and their ligands: promising molecular biomarkers and therapeutic targets in prostate cancer. Biochim Biophys Acta Rev Cancer 2013; 1835:243-57. [PMID: 23396052 DOI: 10.1016/j.bbcan.2013.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/21/2013] [Accepted: 01/25/2013] [Indexed: 01/01/2023]
Abstract
Although at present, there is a high incidence of prostate cancer, particularly in the Western world, mortality from this disease is declining and occurs primarily only from clinically significant late stage tumors with a poor prognosis. A major current focus of this field is the identification of new biomarkers which can detect earlier, and more effectively, clinically significant tumors from those deemed "low risk", as well as predict the prognostic course of a particular cancer. This strategy can in turn offer novel avenues for targeted therapies. The large family of Receptor Tyrosine Kinases, the Ephs, and their binding partners, the ephrins, has been implicated in many cancers of epithelial origin through stimulation of oncogenic transformation, tumor angiogenesis, and promotion of increased cell survival, invasion and migration. They also show promise as both biomarkers of diagnostic and prognostic value and as targeted therapies in cancer. This review will briefly discuss the complex roles and biological mechanisms of action of these receptors and ligands and, with regard to prostate cancer, highlight their potential as biomarkers for both diagnosis and prognosis, their application as imaging agents, and current approaches to assessing them as therapeutic targets. This review demonstrates the need for future studies into those particular family members that will prove helpful in understanding the biology and potential as targets for treatment of prostate cancer.
Collapse
|
42
|
Qu X, Randhawa G, Friedman C, O'Hara-Larrivee S, Kroeger K, Dumpit R, True L, Vakar-Lopez F, Porter C, Vessella R, Nelson P, Fang M. A novel four-color fluorescence in situ hybridization assay for the detection of TMPRSS2 and ERG rearrangements in prostate cancer. Cancer Genet 2013; 206:1-11. [PMID: 23352841 DOI: 10.1016/j.cancergen.2012.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/11/2012] [Accepted: 12/12/2012] [Indexed: 12/23/2022]
Abstract
Since the identification of the TMPRSS2-ERG rearrangement as the most common fusion event in prostate cancer, various methods have been developed to detect this rearrangement and to study its prognostic significance. We report a novel four-color fluorescence in situ hybridization (FISH) assay that detects not only the typical TMPRSS2-ERG fusion but also alternative rearrangements of the TMPRSS2 or ERG gene. We validated this assay on fresh, frozen, or formalin-fixed paraffin-embedded prostate cancer specimens, including cell lines, primary prostate cancer tissues, xenograft tissues derived from metastatic prostate cancer, and metastatic tissues from castration-resistant prostate cancer (CRPC) patients. When compared with either reverse transcription-polymerase chain reaction or the Gen-Probe method as the technical reference, analysis using the four-color FISH assay demonstrated an analytical sensitivity of 94.5% (95% confidence interval [CI] 0.80-0.99) and specificity of 100% (95% CI 0.89-1.00) for detecting the TMPRSS2-ERG fusion. The TMPRSS2-ERG fusion was detected in 41% and 43% of primary prostate cancer (n = 59) and CRPC tumors (n = 82), respectively. Rearrangements other than the typical TMPRSS2-ERG fusion were confirmed by karyotype analysis and found in 7% of primary cancer and 13% of CRPC tumors. Successful karyotype analyses are reported for the first time on four of the xenograft samples, complementing the FISH results. Analysis using the four-color FISH assay provides sensitive detection of TMPRSS2 and ERG gene rearrangements in prostate cancer.
Collapse
Affiliation(s)
- Xiaoyu Qu
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Webb SL, Sanders AJ, Mason MD, Jiang WG. Matriptase-2 inhibits HECV motility and tubule formation in vitro and tumour angiogenesis in vivo. Mol Cell Biochem 2012; 375:207-17. [PMID: 23238872 DOI: 10.1007/s11010-012-1544-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 12/06/2012] [Indexed: 12/12/2022]
Abstract
The type II transmembrane serine proteases (TTSP) are cell surface proteolytic enzymes that mediate a diverse range of cellular functions, including tumour invasion and metastasis. Matriptase-2 is a member of the TTSP family and has been shown to have a key role in cancer progression. The role of matriptase-2 in angiogenesis and angiogenesis-related cancer progression is currently poorly understood. This study aims to elucidate the role of matriptase-2 in tumour angiogenesis. Matriptase-2 was over-expressed in human vascular endothelial cells, HECV, using a mammalian expression plasmid. The altered cells were used in a number of in vitro and in vivo assays designed to investigate the involvement of matriptase-2 in angiogenesis. Over-expression had no significant effect on the growth and adhesion of HECV cells. However, there was a significant reduction in the motility of the cells and their ability to form tubules in an artificial basement membrane (p < 0.01 for both). HECV(mat2 exp) cells inoculated into CD-1 athymic mice along with either PC-3 prostate cancer cells or MDA-MB-231 breast cancer cells showed a dramatic decrease in tumour development and growth in the prostate tumours (p < 0.01) and a lesser, non-significant, decrease in the breast tumours (p = 0.08). Over-expression of matriptase-2 also decreased urokinase type plasminogen activator total protein levels in HECV and prostate cells. The study concludes that matriptase-2 has the ability to suppress the angiogenic nature of HECV cells in vitro and in vivo. It also suggests that matriptase-2 could have a potential role in prostate and breast tumour suppression through its anti-angiogenic properties.
Collapse
Affiliation(s)
- Siobhan L Webb
- Metastasis & Angiogenesis Research Group, Institute of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | | | | | | |
Collapse
|
44
|
Lee YJ, Hsieh WY, Chen LY, Li C. Protein arginine methylation of SERBP1 by protein arginine methyltransferase 1 affects cytoplasmic/nuclear distribution. J Cell Biochem 2012; 113:2721-8. [PMID: 22442049 DOI: 10.1002/jcb.24151] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Protein arginine methylation regulates a broad array of cellular processes. SERBP1 implicated in tumor progression through its putative involvement in the plaminogen activator protease cascade, is an RNA-binding protein containing an RG-rich domain and an RGG box domain that might be methylated by protein arginine N-methyltransferases (PRMTs). Asymmetric dimethylarginine (aDMA) was detected in SERBP1 and an indirect methyltransferase inhibitor adenosine dialdehyde (AdOx) significantly reduced the methylation signals. Arginines in the middle RG and C-terminal RGG region of SERBP1 are methylated based on the analyses of different deletion constructs. The predominant type I protein arginine methyltransferase PRMT1 co-immunoprecipitated with SERBP1 and the level of bound PRMT1 decreased upon the addition of AdOx. Recombinant PRMT1 methylated SERBP1 and knockdown of PRMT1 significantly reduced the aDMA level of SERBP1, indicating that SERBP1 is specifically methylated by PRMT1. Immunofluorescent analyses of endogenous SERBP1 showed predominant cytoplasmic localization of SERBP1. Treatment of AdOx or PRMT1 siRNA increased the nuclear localization of SERBP1. Analyses of different deletions indicated that the middle RG region is important for the nuclear localization while both N- and C- terminus are required for nuclear export. Low methylation of the C-terminal RGG region also favors nuclear localization. In conclusion, the RG-rich and RGG box of SERBP1 is asymmetrically dimethylated by PRMT1 and the modification affects protein interaction and intracellular localization of the protein. These findings provide the basis for dissecting the roles of SERBP1.
Collapse
Affiliation(s)
- Yu-Jen Lee
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | |
Collapse
|
45
|
Abstract
Cancer cells rely on intercellular communication throughout the different stages of their transformation and progression into metastasis. They do so by co-opting different processes such as cell-cell junctions, growth factors, receptors, and vesicular release. Initially characterized in neuronal and vascular tissues, Ephs and Ephrins, the largest family of receptor tyrosine kinases, comprised of two classes (i.e., A and B types), is increasingly scrutinized by cancer researchers. These proteins possess the particular features of both the receptors and ligands being membrane-bound which, via mandatory direct cell-cell interactions, undergo a bidirectional signal transduction initiated from both the receptor and the ligand. Following cell-cell interactions, Ephs/Ephrins behave as guidance molecules which trigger both repulsive and attractive signals, so as to direct the movement of cells through their immediate microenvironment. They also direct processes which include sorting and positioning and cytoskeleton rearrangements, thus making them perfect candidates for the control of the metastatic process. In fact, the role of Ephs and Ephrins in cancer progression has been demonstrated for many of the family members and they, surprisingly, have both tumor promoter and suppressor functions in different cellular contexts. They are also able to coordinate between multiple processes including cell survival, proliferation, differentiation, adhesion, motility, and invasion. This review is an attempt to summarize the data available on these Ephs/Ephrins' biological functions which contribute to the onset of aggressive cancers. I will also provide an overview of the factors which could explain the functional differences demonstrated by Ephs and Ephrins at different stages of tumor progression and whose elucidation is warranted for any future therapeutic targeting of this signaling pathway in cancer metastasis.
Collapse
|
46
|
Langenkamp E, Vom Hagen FM, Zwiers PJ, Moorlag HE, Schouten JP, Hammes HP, Gouw ASH, Molema G. Tumor Vascular Morphology Undergoes Dramatic Changes during Outgrowth of B16 Melanoma While Proangiogenic Gene Expression Remains Unchanged. ISRN ONCOLOGY 2011; 2011:409308. [PMID: 22235379 PMCID: PMC3249352 DOI: 10.5402/2011/409308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 09/07/2011] [Indexed: 12/28/2022]
Abstract
In established tumors, angiogenic endothelial cells (ECs) coexist next to “quiescent” EC in matured vessels. We hypothesized that angio-gene expression of B16.F10 melanoma would differ depending on the growth stage. Unraveling the spatiotemporal nature thereof is essential for drug regimen design aimed to affect multiple neovascularization stages. We determined the angiogenic phenotype—represented by 52 angio-genes—and vascular morphology of small, intermediate, and large s.c. growing mouse B16.F10 tumors and demonstrated that expression of these genes did not differ between the different growth stages. Yet vascular morphology changed dramatically from small vessels without lumen in small to larger vessels with increased lumen size in intermediate/large tumors. Separate analysis of these vascular morphologies revealed a significant difference in αSMA expression in relation to vessel morphology, while no relation with VEGF, HIF-1α, nor Dll4 expression levels was observed. We conclude that the tumor vasculature remains actively engaged in angiogenesis during B16.F10 melanoma outgrowth and that the major change in tumor vascular morphology does not follow molecular concepts generated in other angiogenesis models.
Collapse
Affiliation(s)
- Elise Langenkamp
- Medical Biology Section, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhang X, Morrissey C, Sun S, Ketchandji M, Nelson PS, True LD, Vakar-Lopez F, Vessella RL, Plymate SR. Androgen receptor variants occur frequently in castration resistant prostate cancer metastases. PLoS One 2011; 6:e27970. [PMID: 22114732 PMCID: PMC3219707 DOI: 10.1371/journal.pone.0027970] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 10/29/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Although androgens are depleted in castration resistant prostate cancer (CRPC), metastases still express nuclear androgen receptor (AR) and androgen regulated genes. We recently reported that C-terminal truncated constitutively active AR splice variants contribute to CRPC development. Since specific antibodies detecting all C-terminal truncated AR variants are not available, our aim was to develop an approach to assess the prevalence and function of AR variants in prostate cancer (PCa). METHODOLOGY/PRINCIPAL FINDINGS Using 2 antibodies against different regions of AR protein (N- or C-terminus), we successfully showed the existence of AR variant in the LuCaP 86.2 xenograft. To evaluate the prevalence of AR variants in human PCa tissue, we used this method on tissue microarrays including 50 primary PCa and 162 metastatic CRPC tissues. RT-PCR was used to confirm AR variants. We observed a significant decrease in nuclear C-terminal AR staining in CRPC but no difference between N- and C-terminal AR nuclear staining in primary PCa. The expression of the AR regulated proteins PSA and PSMA were marginally affected by the decrease in C-terminal staining in CRPC samples. These data suggest that there is an increase in the prevalence of AR variants in CRPC based on our ability to differentiate nuclear AR expression using N- and C-terminal AR antibodies. These findings were validated using RT-PCR. Importantly, the loss of C-terminal immunoreactivity and the identification of AR variants were different depending on the site of metastasis in the same patient. CONCLUSIONS We successfully developed a novel immunohistochemical approach which was used to ascertain the prevalence of AR variants in a large number of primary PCa and metastatic CRPC. Our results showed a snapshot of overall high frequency of C-terminal truncated AR splice variants and site specific AR loss in CRPC, which could have utility in stratifying patients for AR targeted therapeutics.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Alternative Splicing
- Blotting, Western
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/secondary
- Castration
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Male
- Middle Aged
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Prognosis
- Prostate/metabolism
- Prostate/pathology
- Prostatic Hyperplasia/genetics
- Prostatic Hyperplasia/metabolism
- Prostatic Hyperplasia/pathology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Soft Tissue Neoplasms/genetics
- Soft Tissue Neoplasms/metabolism
- Soft Tissue Neoplasms/secondary
Collapse
Affiliation(s)
- Xiaotun Zhang
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Shihua Sun
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Melanie Ketchandji
- Department of Urology, University of Washington, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lawrence D. True
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Funda Vakar-Lopez
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Robert L. Vessella
- Department of Urology, University of Washington, Seattle, Washington, United States of America
- GRECC and Research Service, Department of Veterans Affairs Medical Center, Seattle, Washington, United States of America
| | - Stephen R. Plymate
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- GRECC and Research Service, Department of Veterans Affairs Medical Center, Seattle, Washington, United States of America
| |
Collapse
|
48
|
Miao H, Wang B. EphA receptor signaling--complexity and emerging themes. Semin Cell Dev Biol 2011; 23:16-25. [PMID: 22040915 DOI: 10.1016/j.semcdb.2011.10.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 10/17/2011] [Indexed: 01/09/2023]
Abstract
The impact of Eph and ephrin signaling on cell behavior is complex and highly context dependent. Forward signaling initiated by Eph receptor activation and reverse signaling initiated by ephrin activation often mediate opposite effects. The apparent ligand-independent functions of Eph receptors recognized recently add another layer of complexity. This review will attempt to sort out the information generated recently on signaling by the A subfamily of Eph receptors and ephrin ligands. We will focus on EphA/ephrin-A signaling in the context of several physiological and disease processes, where new progresses have been made lately and unifying themes are emerging amid previous confusions. For more comprehensive survey of literature on Eph/ephrin signaling pathways and networks, readers are referred to outstanding reviews both in this volume and in other recent publications.
Collapse
Affiliation(s)
- Hui Miao
- Rammelkamp Center for Research, MetroHealth Campus, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA.
| | | |
Collapse
|
49
|
Ganesan R, Kolumam GA, Lin SJ, Xie MH, Santell L, Wu TD, Lazarus RA, Chaudhuri A, Kirchhofer D. Proteolytic activation of pro-macrophage-stimulating protein by hepsin. Mol Cancer Res 2011; 9:1175-86. [PMID: 21875933 DOI: 10.1158/1541-7786.mcr-11-0004] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Macrophage-stimulating protein (MSP) is a plasminogen-related growth factor and ligand for the receptor tyrosine kinase RON. The MSP/RON system promotes wound healing and invasive tumor growth and suppresses proinflammatory immune response. MSP binding to RON requires proteolytic conversion of the inactive single-chain form (pro-MSP) into the disulfide-linked α/β heterodimer. The pro-MSP cleavage sequence (Ser-Lys-Leu-Arg(483)↓Val(484)) closely matches the substrate recognition sequences of hepsin, a type II transmembrane serine protease, that is overexpressed in several cancers. Here, we show that recombinant hepsin cleaves pro-MSP at the consensus site Arg(483)-Val(484) with superior efficiency compared with the known activators MT-SP1 and hepatocyte growth factor activator (HGFA). At least 50% of pro-MSP was processed within 1 hour at a hepsin concentration of 2.4 nmol/L and at a molar enzyme to substrate ratio of 1:500. An uncleavable single-chain variant of MSP weakly bound to a RON-Fc fusion protein, whereas hepsin-cleaved MSP bound with a K(D) of 10.3 nmol/L, suggesting that the high-affinity binding site in MSP β-chain was properly formed. LNCaP prostate cancer cells overexpressing hepsin on the cell surface efficiently activated pro-MSP, which was blocked by a specific anti-hepsin antibody. Incubation of pro-MSP with hepsin led to robust RON-mediated phosphorylation of mitogen-activated protein kinase, ribosomal S6 protein, and Akt in human A2780 ovarian carcinoma cells stably expressing RON protein. In macrophages, pro-MSP with hepsin induced chemotaxis and attenuated lipopolysaccharide-dependent production of nitric oxide. These findings suggest that the MSP/RON signaling pathway may be regulated by hepsin in tissue homeostasis and in disease pathologies, such as in cancer and immune disorders.
Collapse
Affiliation(s)
- Rajkumar Ganesan
- Department of Early Discovery Biochemistry, Genentech, Inc., 1 DNA Way, MS #27, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sottnik JL, Zhang J, Macoska JA, Keller ET. The PCa Tumor Microenvironment. CANCER MICROENVIRONMENT 2011; 4:283-97. [PMID: 21728070 DOI: 10.1007/s12307-011-0073-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 06/23/2011] [Indexed: 12/31/2022]
Abstract
The tumor microenvironment (TME) is a very complex niche that consists of multiple cell types, supportive matrix and soluble factors. Cells in the TME consist of both host cells that are present at tumor site at the onset of tumor growth and cells that are recruited in either response to tumor- or host-derived factors. PCa (PCa) thrives on crosstalk between tumor cells and the TME. Crosstalk results in an orchestrated evolution of both the tumor and microenvironment as the tumor progresses. The TME reacts to PCa-produced soluble factors as well as direct interaction with PCa cells. In return, the TME produces soluble factors, structural support and direct contact interactions that influence the establishment and progression of PCa. In this review, we focus on the host side of the equation to provide a foundation for understanding how different aspects of the TME contribute to PCa progression. We discuss immune effector cells, specialized niches, such as the vascular and bone marrow, and several key protein factors that mediate host effects on PCa. This discussion highlights the concept that the TME offers a potentially very fertile target for PCa therapy.
Collapse
Affiliation(s)
- Joseph L Sottnik
- Department of Urology, University of Michigan, RM 5308 CC, Ann Arbor, MI, 48109-8940, USA
| | | | | | | |
Collapse
|