1
|
Iluta S, Nistor M, Buruiana S, Dima D. Wnt Signaling Pathway in Tumor Biology. Genes (Basel) 2024; 15:1597. [PMID: 39766864 PMCID: PMC11675244 DOI: 10.3390/genes15121597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Relapse and metastasis are the major challenges that stand in the way of cancer healing and survival, mainly attributed to cancer stem cells (CSCs). Their capabilities of self-renewal and tumorigenic potential leads to treatment resistance development. CSCs function through signaling pathways such as the Wnt/β-catenin cascade. While commonly involved in embryogenesis and adult tissues homeostasis, the dysregulation of the Wnt pathway has direct correlations with tumorigenesis, metastasis, and drug resistance. The development of therapies that target CSCs and bulk tumors is both crucial and urgent. However, the extensive crosstalk present between Wnt and other signaling networks (Hedgehog and Notch) complicates the development of efficient long-term therapies with minimal side-effects on normal tissues. Despite the obstacles, the emergence of Wnt inhibitors and subsequent modulation of the signaling pathways would provide dynamic therapeutic approaches to impairing CSCs and reversing resistance mechanisms.
Collapse
Affiliation(s)
- Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj Napoca, Romania;
| | - Madalina Nistor
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj Napoca, Romania
| | - Sanda Buruiana
- Department of Hematology, Nicolae Testemitanu University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj Napoca, Romania
| |
Collapse
|
2
|
Hashemi M, Rezaei M, Rezaeiaghdam H, Jamali B, Koohpar ZK, Tanha M, Bizhanpour A, Asadi S, Jafari AM, Khosroshahi EM, Eslami M, Salimimoghadam S, Nabavi N, Rashidi M, Fattah E, Taheriazam A, Entezari M. Highlighting function of Wnt signalling in urological cancers: Molecular interactions, therapeutic strategies, and (nano)strategies. Transl Oncol 2024; 50:102145. [PMID: 39357465 PMCID: PMC11474201 DOI: 10.1016/j.tranon.2024.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/06/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer is a complex, multistep process characterized by abnormal cell growth and metastasis as well as the capacity of the tumor cells in therapy resistance development. The urological system is particularly susceptible to a group of malignancies known as urological cancers, where an accumulation of genetic alterations drives carcinogenesis. In various human cancers, Wnt singalling is dysregulated; following nuclear transfer of β-catenin, it promotes tumor progression and affects genes expression. Elevated levels of Wnt have been documented in urological cancers, where its overexpression enhances growth and metastasis. Additionally, increased Wnt singalling contributes to chemoresistance in urological cancers, leading to reduced sensitivity to chemotherapy agents like cisplatin, doxorubicin, and paclitaxel. Wnt upregulation can change radiotherapy response of urological cancers. The regulation of Wnt involves various molecular pathways, including Akt, miRNAs, lncRNAs, and circRNAs, all of which play roles in carcinogenesis. Targeting and silencing Wnt or its associated pathways can mitigate tumorigenesis in urological cancers. Anti-cancer compounds such as curcumin and thymoquinone have shown efficacy in suppressing tumorigenesis through the downregulation of Wnt singalling. Notably, nanoparticles have proven effective in treating urological cancers, with several studies in prostate cancer (PCa) using nanoparticles to downregulate Wnt and suppress tumor growth. Future research should focus on developing small molecules that inhibit Wnt singalling to further suppress tumorigenesis and advance the treatment of urological cancers. Moreover, Wnt can be used as reliable biomarker for the diagnosis and prognosis of urological cancers.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Rezaei
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Hadi Rezaeiaghdam
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Behdokht Jamali
- Department of Microbiology and Genetics, Kherad Institute of Higher Education, Bushehr, Iran
| | - Zeinab Khazaei Koohpar
- Department Of Cell and Molecular Biology, Faculty of Biological Sciences,Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Mahsa Tanha
- Department Of Biological Sciences, University Of Alabama, Tuscaloosa, Al, United States
| | - Anahita Bizhanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Maedeh Eslami
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8V 1P7, Canada
| | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Eisa Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
3
|
Vecchiotti D, Clementi L, Cornacchia E, Di Vito Nolfi M, Verzella D, Capece D, Zazzeroni F, Angelucci A. Evidence of the Link between Stroma Remodeling and Prostate Cancer Prognosis. Cancers (Basel) 2024; 16:3215. [PMID: 39335188 PMCID: PMC11430343 DOI: 10.3390/cancers16183215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer (PCa), the most commonly diagnosed cancer in men worldwide, is particularly challenging for oncologists when a precise prognosis needs to be established. Indeed, the entire clinical management in PCa has important drawbacks, generating an intense debate concerning the possibility to individuate molecular biomarkers able to avoid overtreatment in patients with pathological indolent cancers. To date, the paradigmatic change in the view of cancer pathogenesis prompts to look for prognostic biomarkers not only in cancer epithelial cells but also in the tumor microenvironment. PCa ecology has been defined with increasing details in the last few years, and a number of promising key markers associated with the reactive stroma are now available. Here, we provide an updated description of the most biologically significant and cited prognosis-oriented microenvironment biomarkers derived from the main reactive processes during PCa pathogenesis: tissue adaptations, inflammatory response and metabolic reprogramming. Proposed biomarkers include factors involved in stromal cell differentiation, cancer-normal cell crosstalk, angiogenesis, extracellular matrix remodeling and energy metabolism.
Collapse
Affiliation(s)
- Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Letizia Clementi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Emanuele Cornacchia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
4
|
Song P, Gao Z, Bao Y, Chen L, Huang Y, Liu Y, Dong Q, Wei X. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J Hematol Oncol 2024; 17:46. [PMID: 38886806 PMCID: PMC11184729 DOI: 10.1186/s13045-024-01563-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
The Wnt/β-catenin signaling pathway plays a crucial role in various physiological processes, encompassing development, tissue homeostasis, and cell proliferation. Under normal physiological conditions, the Wnt/β-catenin signaling pathway is meticulously regulated. However, aberrant activation of this pathway and downstream target genes can occur due to mutations in key components of the Wnt/β-catenin pathway, epigenetic modifications, and crosstalk with other signaling pathways. Consequently, these dysregulations contribute significantly to tumor initiation and progression. Therapies targeting the Wnt/β-catenin signaling transduction have exhibited promising prospects and potential for tumor treatment. An increasing number of medications targeting this pathway are continuously being developed and validated. This comprehensive review aims to summarize the latest advances in our understanding of the role played by the Wnt/β-catenin signaling pathway in carcinogenesis and targeted therapy, providing valuable insights into acknowledging current opportunities and challenges associated with targeting this signaling pathway in cancer research and treatment.
Collapse
Affiliation(s)
- Pan Song
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Zirui Gao
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yige Bao
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuhe Huang
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yanyan Liu
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Qiang Dong
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
5
|
Bhat GR, Sethi I, Sadida HQ, Rah B, Mir R, Algehainy N, Albalawi IA, Masoodi T, Subbaraj GK, Jamal F, Singh M, Kumar R, Macha MA, Uddin S, Akil ASAS, Haris M, Bhat AA. Cancer cell plasticity: from cellular, molecular, and genetic mechanisms to tumor heterogeneity and drug resistance. Cancer Metastasis Rev 2024; 43:197-228. [PMID: 38329598 PMCID: PMC11016008 DOI: 10.1007/s10555-024-10172-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
Cancer is a complex disease displaying a variety of cell states and phenotypes. This diversity, known as cancer cell plasticity, confers cancer cells the ability to change in response to their environment, leading to increased tumor diversity and drug resistance. This review explores the intricate landscape of cancer cell plasticity, offering a deep dive into the cellular, molecular, and genetic mechanisms that underlie this phenomenon. Cancer cell plasticity is intertwined with processes such as epithelial-mesenchymal transition and the acquisition of stem cell-like features. These processes are pivotal in the development and progression of tumors, contributing to the multifaceted nature of cancer and the challenges associated with its treatment. Despite significant advancements in targeted therapies, cancer cell adaptability and subsequent therapy-induced resistance remain persistent obstacles in achieving consistent, successful cancer treatment outcomes. Our review delves into the array of mechanisms cancer cells exploit to maintain plasticity, including epigenetic modifications, alterations in signaling pathways, and environmental interactions. We discuss strategies to counteract cancer cell plasticity, such as targeting specific cellular pathways and employing combination therapies. These strategies promise to enhance the efficacy of cancer treatments and mitigate therapy resistance. In conclusion, this review offers a holistic, detailed exploration of cancer cell plasticity, aiming to bolster the understanding and approach toward tackling the challenges posed by tumor heterogeneity and drug resistance. As articulated in this review, the delineation of cellular, molecular, and genetic mechanisms underlying tumor heterogeneity and drug resistance seeks to contribute substantially to the progress in cancer therapeutics and the advancement of precision medicine, ultimately enhancing the prospects for effective cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Gh Rasool Bhat
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Jammu and Kashmir, India
| | - Itty Sethi
- Institute of Human Genetics, University of Jammu, Jammu, Jammu and Kashmir, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Bilal Rah
- Iron Biology Group, Research Institute of Medical and Health Science, University of Sharjah, Sharjah, UAE
| | - Rashid Mir
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Prince Fahad Bin Sultan Chair for Biomedical Research, University of Tabuk, Tabuk, Saudi Arabia
| | | | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | | | - Farrukh Jamal
- Dr. Rammanohar, Lohia Avadh University, Ayodhya, India
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Institute of Medical Sciences (AIIMS), Dr. BRAIRCH, All India, New Delhi, India
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory Animal Research Centre, Qatar University, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Mohammad Haris
- Laboratory Animal Research Centre, Qatar University, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
6
|
Chen M, Liu X, Lu J, Teng H, Yu C, Liu Y, Zheng Y. Dysregulation of the circ_0087502/miR-1179/TGFBR2 pathway supports gemcitabine resistance in pancreatic cancer. Cancer Biol Ther 2023; 24:2258566. [PMID: 37844011 PMCID: PMC10580792 DOI: 10.1080/15384047.2023.2258566] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 05/12/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are a cohort of non-coding RNAs generated by back-splicing events. Accumulating evidence supports the crucial role of circRNAs in human tumorigenesis, metastasis, and chemoresistance. However, the role and mechanism of circRNA circ_0087502 in pancreatic cancer are yet unknown. METHODS The expression and function of circ_0087502 in pancreatic cancer were investigated using qRT-PCR and cell experiments. The predicted binding between circ_0087502 and microRNA-1179 (miR-1179), and between miR-1179 and TGFBR2, were examined using reporter assays. RESULTS Pancreatic cancer tissues and cell lines were discovered to express circ_0087502 at higher levels. Patients with pancreatic cancer who express circ_0087502 at high levels have a worse prognosis. In addition, circ_0087502 knockdown reduced the proliferation, migration, and invasion of pancreatic cancer cells and made them more sensitive to gemcitabine treatment. We found that circ_0087502 worked as a sponge for miR-1179, allowing miR-1179 to bind to the critical oncogene TGFBR2 in its 3'-untranslated region (3'-UTR). Pancreatic cancer cells were highly resistant to gemcitabine and had increased proliferation, migration, and invasion when miR-1179 was inhibited or overexpressed. CONCLUSION These results confirm that circ_0087502 activates the miR-1179/TGFBR2 axis to promote gemcitabine resistance in pancreatic cancer. Thus, our data might lay the groundwork for developing novel therapeutic strategies targeting circ_0087502 in pancreatic cancer patients.
Collapse
Affiliation(s)
- Mingliu Chen
- Department of hepatobiliary and pancreatic surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of hepatobiliary and pancreatic surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xinxiu Liu
- Department of ultrasound, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinpeng Lu
- Department of hepatobiliary and pancreatic surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Haiwen Teng
- Department of hepatobiliary and pancreatic surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chengui Yu
- Department of hepatobiliary and pancreatic surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yingchun Liu
- Department of Cell Biology and Genetics, Fujian Medical University, Fuzhou, China
| | - Yansong Zheng
- Department of hepatobiliary and pancreatic surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
7
|
Shree B, Das K, Sharma V. Emerging role of transforming growth factor-β-regulated long non-coding RNAs in prostate cancer pathogenesis. CANCER PATHOGENESIS AND THERAPY 2023; 1:195-204. [PMID: 38327834 PMCID: PMC10846338 DOI: 10.1016/j.cpt.2022.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/01/2022] [Accepted: 12/20/2022] [Indexed: 02/09/2024]
Abstract
Prostate cancer (PCa) is the most common malignancy in men. Despite aggressive therapy involving surgery and hormonal treatments, the recurrence and emergence of metastatic castration-resistant prostate cancer (CRPCa) remain a major challenge. Dysregulation of the transforming growth factor-β (TGF-β) signaling pathway is crucial to PCa development and progression. This also contributes to androgen receptor activation and the emergence of CRPC. In addition, TGF-β signaling regulates long non-coding RNA (lncRNA) expression in multiple cancers, including PCa. Here, we discuss the complex regulatory network of lncRNAs and TGF-β signaling in PCa and their potential applications in diagnosing, prognosis, and treating PCa. Further investigations on the role of lncRNAs in the TGF-β pathway will help to better understand PCa pathogenesis.
Collapse
Affiliation(s)
- Bakhya Shree
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Koyel Das
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Vivek Sharma
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad 500078, India
| |
Collapse
|
8
|
Choi WWY, Sánchez C, Li JJ, Dinarvand M, Adomat H, Ghaffari M, Khoja L, Vafaee F, Joshua AM, Chi KN, Guns EST, Hosseini-Beheshti E. Extracellular vesicles from biological fluids as potential markers in castration resistant prostate cancer. J Cancer Res Clin Oncol 2023; 149:4701-4717. [PMID: 36222898 PMCID: PMC10349738 DOI: 10.1007/s00432-022-04391-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/03/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Extracellular vesicles (EV) secreted from cancer cells are present in various biological fluids, carrying distinctly different cellular components compared to normal cells, and have great potential to be used as markers for disease initiation, progression, and response to treatment. This under-utilised tool provides insights into a better understanding of prostate cancer. METHODS EV from serum and urine of healthy men and castration-resistant prostate cancer (CRPC) patients were isolated and characterised by transmission electron microscopy, particle size analysis, and western blot. Proteomic and cholesterol liquid chromatography-mass spectrometry (LC-MS) analyses were conducted. RESULTS There was a successful enrichment of small EV/exosomes isolated from serum and urine. EV derived from biological fluids of CRPC patients had significant differences in composition when compared with those from healthy controls. Analysis of matched serum and urine samples from six prostate cancer patients revealed specific EV proteins common in both types of biological fluid for each patient. CONCLUSION Some of the EV proteins identified from our analyses have potential to be used as CRPC markers. These markers may depict a pattern in cancer progression through non-invasive sample collection.
Collapse
Affiliation(s)
- Wendy W Y Choi
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | | | - Jiao Jiao Li
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, 2065, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Mojdeh Dinarvand
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW, 2052, Australia
| | - Hans Adomat
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Mazyar Ghaffari
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Leila Khoja
- St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW, 2052, Australia
- UNSW Data Science Hub, University of New South Wales, Kensington, NSW, 2052, Australia
| | - Anthony M Joshua
- St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia
| | - Kim N Chi
- BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Emma S Tomlinson Guns
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
- BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Elham Hosseini-Beheshti
- Vancouver Prostate Centre, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- The Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
9
|
Smith BN, Mishra R, Billet S, Placencio-Hickok VR, Kim M, Zhang L, Duong F, Madhav A, Scher K, Moldawer N, Oppenheim A, Angara B, You S, Tighiouart M, Posadas EM, Bhowmick NA. Antagonizing CD105 and androgen receptor to target stromal-epithelial interactions for clinical benefit. Mol Ther 2023; 31:78-89. [PMID: 36045587 PMCID: PMC9840108 DOI: 10.1016/j.ymthe.2022.08.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/09/2022] [Accepted: 08/25/2022] [Indexed: 01/28/2023] Open
Abstract
Androgen receptor signaling inhibitors (ARSIs) are standard of care for advanced prostate cancer (PCa) patients. Eventual resistance to ARSIs can include the expression of androgen receptor (AR) splice variant, AR-V7, expression as a recognized means of ligand-independent androgen signaling. We demonstrated that interleukin (IL)-6-mediated AR-V7 expression requires bone morphogenic protein (BMP) and CD105 receptor activity in both PCa and associated fibroblasts. Chromatin immunoprecipitation supported CD105-dependent ID1- and E2F-mediated expression of RBM38. Further, RNA immune precipitation demonstrated RBM38 binds the AR-cryptic exon 3 to enable AR-V7 generation. The forced expression of AR-V7 by primary prostatic fibroblasts diminished PCa sensitivity to ARSI. Conversely, downregulation of AR-V7 expression in cancer epithelia and associated fibroblasts was achieved by a CD105-neutralizing antibody, carotuximab. These compelling pre-clinical findings initiated an interventional study in PCa patients developing ARSI resistance. The combination of carotuximab and ARSI (i.e., enzalutamide or abiraterone) provided disease stabilization in four of nine assessable ARSI-refractory patients. Circulating tumor cell evaluation showed AR-V7 downregulation in the responsive subjects on combination treatment and revealed a three-gene panel that was predictive of response. The systemic antagonism of BMP/CD105 signaling can support ARSI re-sensitization in pre-clinical models and subjects that have otherwise developed resistance due to AR-V7 expression.
Collapse
Affiliation(s)
- Bethany N Smith
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Rajeev Mishra
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA; School of Life Sciences & Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, Uttar Pradesh 208024, India
| | - Sandrine Billet
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | | | - Minhyung Kim
- Department of Surgery, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Le Zhang
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Frank Duong
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Anisha Madhav
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Kevin Scher
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Nancy Moldawer
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Amy Oppenheim
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Bryan Angara
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA; VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Mourad Tighiouart
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Edwin M Posadas
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Cancer, Los Angeles, CA 90048, USA; VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| |
Collapse
|
10
|
Owen JS, Clayton A, Pearson HB. Cancer-Associated Fibroblast Heterogeneity, Activation and Function: Implications for Prostate Cancer. Biomolecules 2022; 13:67. [PMID: 36671452 PMCID: PMC9856041 DOI: 10.3390/biom13010067] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The continuous remodeling of the tumor microenvironment (TME) during prostate tumorigenesis is emerging as a critical event that facilitates cancer growth, progression and drug-resistance. Recent advances have identified extensive communication networks that enable tumor-stroma cross-talk, and emphasized the functional importance of diverse, heterogeneous stromal fibroblast populations during malignant growth. Cancer-associated fibroblasts (CAFs) are a vital component of the TME, which mediate key oncogenic events including angiogenesis, immunosuppression, metastatic progression and therapeutic resistance, thus presenting an attractive therapeutic target. Nevertheless, how fibroblast heterogeneity, recruitment, cell-of-origin and differential functions contribute to prostate cancer remains to be fully delineated. Developing our molecular understanding of these processes is fundamental to developing new therapies and biomarkers that can ultimately improve clinical outcomes. In this review, we explore the current challenges surrounding fibroblast identification, discuss new mechanistic insights into fibroblast functions during normal prostate tissue homeostasis and tumorigenesis, and illustrate the diverse nature of fibroblast recruitment and CAF generation. We also highlight the promise of CAF-targeted therapies for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jasmine S. Owen
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Aled Clayton
- Tissue Microenvironment Group, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
11
|
Mašić S, Bacalja J, Vučić M, Čupić H, Tomas D, Ulamec M, Spajić B, Skenderi F, Krušlin B. CORRELATION OF EXPRESSION OF TGF- β AND MMP2 BETWEEN PROSTATIC ADENOCARCINOMA AND ADJACENT UNAFFECTED PARENCHYMA. Acta Clin Croat 2022; 61:9-14. [PMID: 36938549 PMCID: PMC10022412 DOI: 10.20471/acc.2022.61.s3.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
In prostate adenocarcinoma, both tumorous stroma and epithelium have important role in tumor progression. Transforming growth factor beta (TGF- β) is a promotor in advanced stages of prostate cancer. Matrix Metalloproteinase 2 (MMP2), the endopeptidase that degrades extracellular matrix is considered to be overexpressed in prostatic carcinoma related to its growth and aggressiveness. Therefore, the aim was to analyze the expression of proteins TGF- β and MMP2 between both epithelium and stroma of prostatic adenocarcinoma and adjacent unaffected parenchyma. The intensity of TGF- β and MMP2 expression in epithelium, tumorous stroma and adjacent unaffected parenchyma was analyzed in 62 specimens of prostatic adenocarcinoma by microarray-based immunohistochemistry. TGF- β was more expressed in tumorous than in prostate stroma (p =0.000), while no statistical significance in case of MMP2 (p = 0.097) was found. MMP2 was more expressed in tumorous than in prostate epithelium (p =0.000), while no statistical significance in case of TGF- β (p = 0.096) was observed. The study results indicate that both tumorous stroma and epithelium have a role in tumor progression and support potential role of TGF- β and MMP2 in prostatic adenocarcinoma progression.
Collapse
Affiliation(s)
- Silvija Mašić
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jasna Bacalja
- Department of Pathology, Centrallasarettet Växjö, Växjö, Sweden
| | - Majda Vučić
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Department of Pathology, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Hrvoje Čupić
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Department of Pathology, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Davor Tomas
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Monika Ulamec
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Department of Pathology, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Borislav Spajić
- Clinical Department of Urology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Faruk Skenderi
- Faculty of Health Sciences, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Božo Krušlin
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
12
|
Zhang L, Billet S, Gonzales G, Rohena-Rivera K, Muranaka H, Chu GCY, Yang Q, Kim H, Bhowmick NA, Smith B. Fatty Acid Signaling Impacts Prostate Cancer Lineage Plasticity in an Autocrine and Paracrine Manner. Cancers (Basel) 2022; 14:3449. [PMID: 35884514 PMCID: PMC9318639 DOI: 10.3390/cancers14143449] [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] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
Prostate cancer (PCa) affects an estimated 250,000 men every year and causes 34,000 deaths annually. A high-fat diet and obesity are associated with PCa progression and mortality. This study's premise was the novel observation of crosstalk between PCa epithelia and cancer-associated fibroblasts (CAF) in response to palmitate-mediated lineage plasticity. We found that cholesterol activated canonical Hedgehog (Hh) signaling by increasing cilium Gli activity in PCa cells, while palmitate activated Hh independent of Gli. Exogenous palmitate activated SOX2, a known mediator of lineage plasticity, in PCa cells cocultured with CAF. Stroma-derived Wnt5a was upregulated in CAF while cocultured with PCa cells and treated with palmitate. Wnt5a knockdown in CAF inhibited Hh and SOX2 expression in PCa cells from cocultures. These findings supported our proposed mechanism of a high-fat diet promoting Hh signaling-mediated transformation within the tumor microenvironment. SOX2 and Wnt5a expression were limited by the CD36 neutralizing antibody. Mice xenografted with PCa epithelia and CAF tumors were fed a high-fat diet, leading to elevated SOX2 expression and lineage plasticity reprogramming compared to mice fed an isocaloric rodent diet. CD36 inhibition with enzalutamide elevated apoptosis by TUNEL, but limited proliferation and SOX2 expression compared to enzalutamide alone. This study revealed a mechanism for a high-fat diet to affect prostate cancer progression. We found that saturated fat induced lineage plasticity reprogramming of PCa by interaction with CAF through Wnt5a and Hh signaling.
Collapse
Affiliation(s)
- Le Zhang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Sandrine Billet
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Gabrielle Gonzales
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Krizia Rohena-Rivera
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Hayato Muranaka
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Gina Chia-Yi Chu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Qian Yang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Hyung Kim
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| | - Neil A. Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
- Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Bethany Smith
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (L.Z.); (S.B.); (G.G.); (K.R.-R.); (H.M.); (G.C.-Y.C.); (Q.Y.); (H.K.)
| |
Collapse
|
13
|
Transforming growth factor-beta (TGF-β) in prostate cancer: A dual function mediator? Int J Biol Macromol 2022; 206:435-452. [PMID: 35202639 DOI: 10.1016/j.ijbiomac.2022.02.094] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta (TGF-β) is a member of a family of secreted cytokines with vital biological functions in cells. The abnormal expression of TGF-β signaling is a common finding in pathological conditions, particularly cancer. Prostate cancer (PCa) is one of the leading causes of death among men. Several genetic and epigenetic alterations can result in PCa development, and govern its progression. The present review attempts to shed some light on the role of TGF-β signaling in PCa. TGF-β signaling can either stimulate or inhibit proliferation and viability of PCa cells, depending on the context. The metastasis of PCa cells is increased by TGF-β signaling via induction of EMT and MMPs. Furthermore, TGF-β signaling can induce drug resistance of PCa cells, and can lead to immune evasion via reducing the anti-tumor activity of cytotoxic T cells and stimulating regulatory T cells. Upstream mediators such as microRNAs and lncRNAs, can regulate TGF-β signaling in PCa. Furthermore, some pharmacological compounds such as thymoquinone and valproic acid can suppress TGF-β signaling for PCa therapy. TGF-β over-expression is associated with poor prognosis in PCa patients. Furthermore, TGF-β up-regulation before prostatectomy is associated with recurrence of PCa. Overall, current review discusses role of TGF-β signaling in proliferation, metastasis and therapy response of PCa cells and in order to improve knowledge towards its regulation, upstream mediators of TGF-β such as non-coding RNAs are described. Finally, TGF-β regulation and its clinical application are discussed.
Collapse
|
14
|
Su S, Cao J, Meng X, Liu R, Vander Ark A, Woodford E, Zhang R, Stiver I, Zhang X, Madaj ZB, Bowman MJ, Wu Y, Xu HE, Chen B, Yu H, Li X. Enzalutamide-induced and PTH1R-mediated TGFBR2 degradation in osteoblasts confers resistance in prostate cancer bone metastases. Cancer Lett 2022; 525:170-178. [PMID: 34752846 PMCID: PMC9669895 DOI: 10.1016/j.canlet.2021.10.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 01/30/2023]
Abstract
Enzalutamide resistance has been observed in approximately 50% of patients with prostate cancer (PCa) bone metastases. Therefore, there is an urgent need to investigate the mechanisms and develop strategies to overcome resistance. We observed enzalutamide resistance in bone lesion development induced by PCa cells in mouse models. We found that the bone microenvironment was indispensable for enzalutamide resistance because enzalutamide significantly inhibited the growth of subcutaneous C4-2B tumors and the proliferation of C4-2B cells isolated from the bone lesions, and the resistance was recapitulated only when C4-2B cells were co-cultured with osteoblasts. In revealing how osteoblasts contribute to enzalutamide resistance, we found that enzalutamide decreased TGFBR2 protein expression in osteoblasts, which was supported by clinical data. This decrease was possibly through PTH1R-mediated endocytosis. We showed that PTH1R blockade rescued enzalutamide-mediated decrease in TGFBR2 levels and enzalutamide responses in C4-2B cells that were co-cultured with osteoblasts. This is the first study to reveal the contribution of the bone microenvironment to enzalutamide resistance and identify PTH1R as a feasible target to overcome the resistance in PCa bone metastases.
Collapse
Affiliation(s)
- Shang Su
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,Current address: Department of Cancer Biology, the University of Toledo, Toledo, OH, 43614
| | - Jingchen Cao
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503
| | - Xiangqi Meng
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,Current address: The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Ruihua Liu
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,Current address: Department of Cancer Biology, the University of Toledo, Toledo, OH, 43614;,Inner Mongolia University, Hohhot, 010021, China
| | - Alexandra Vander Ark
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503
| | - Erica Woodford
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503
| | - Reian Zhang
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,University of Michigan, Ann Arbor, MI, 48109
| | - Isabelle Stiver
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,University of Michigan, Ann Arbor, MI, 48109
| | - Xiaotun Zhang
- Anatomic/Clinical Pathology, Mayo Clinic, Rochester, MN, 55905
| | - Zachary B. Madaj
- Bioinformatics & Biostatistics Core, Van Andel Institute, Grand Rapids, MI, 49503
| | - Megan J. Bowman
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,Current address: Ball Horticultural Company, West Chicago, IL, 60185
| | - Yingying Wu
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503;,Current address: Center of Mathematical Sciences and Applications, Harvard University, Cambridge, MA 02138
| | - H. Eric Xu
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,Current address: Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bin Chen
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503
| | - Haiquan Yu
- Inner Mongolia University, Hohhot, 010021, China
| | - Xiaohong Li
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, 49503;,Current address: Department of Cancer Biology, the University of Toledo, Toledo, OH, 43614;,Corresponding author: Xiaohong Li, the University of Toledo, 3000 Transverse Drive, Toledo, OH 43614. Phone: +1-419-383-3982;
| |
Collapse
|
15
|
ChallaSivaKanaka S, Vickman RE, Kakarla M, Hayward SW, Franco OE. Fibroblast heterogeneity in prostate carcinogenesis. Cancer Lett 2022; 525:76-83. [PMID: 34715252 PMCID: PMC8788937 DOI: 10.1016/j.canlet.2021.10.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/17/2021] [Accepted: 10/19/2021] [Indexed: 01/30/2023]
Abstract
Our understanding of stromal components, specifically cancer-associated fibroblasts (CAF), in prostate cancer (PCa), has evolved from considering these cells as inert bystanders to acknowledging their significance as players in prostate tumorigenesis. CAF are multifaceted-they promote cancer cell growth, migration and remodel the tumor microenvironment. Although targeting CAF could be a promising strategy for PCa treatment, they incorporate a high but undefined degree of intrinsic cellular heterogeneity. The interaction between CAF subpopulations, with the normal and tumor epithelium and with other cell types is not yet characterized. Defining these interactions and the critical signaling nodes that support tumorigenesis will enable the development of novel strategies to control prostate cancer progression. Here we will discuss the origins, molecular and functional heterogeneity of CAF in PCa. We highlight the challenges associated with delineating CAF heterogeneity and discuss potential areas of research that would assist in expanding our knowledge of CAF and their role in PCa tumorigenesis.
Collapse
Affiliation(s)
- Sathyavathi ChallaSivaKanaka
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Renee E Vickman
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Mamatha Kakarla
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Simon W Hayward
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA
| | - Omar E Franco
- Department of Surgery, NorthShore University HealthSystem, Research Institute, 1001 University Place, Evanston, IL, 60201, USA. http://
| |
Collapse
|
16
|
Rada M, Kapelanski-Lamoureux A, Petrillo S, Tabariès S, Siegel P, Reynolds AR, Lazaris A, Metrakos P. Runt related transcription factor-1 plays a central role in vessel co-option of colorectal cancer liver metastases. Commun Biol 2021; 4:950. [PMID: 34376784 PMCID: PMC8355374 DOI: 10.1038/s42003-021-02481-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 06/17/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer liver metastasis (CRCLM) has two major histopathological growth patterns: angiogenic desmoplastic and non-angiogenic replacement. The replacement lesions obtain their blood supply through vessel co-option, wherein the cancer cells hijack pre-existing blood vessels of the surrounding liver tissue. Consequentially, anti-angiogenic therapies are less efficacious in CRCLM patients with replacement lesions. However, the mechanisms which drive vessel co-option in the replacement lesions are unknown. Here, we show that Runt Related Transcription Factor-1 (RUNX1) overexpression in the cancer cells of the replacement lesions drives cancer cell motility via ARP2/3 to achieve vessel co-option. Furthermore, overexpression of RUNX1 in the cancer cells is mediated by Transforming Growth Factor Beta-1 (TGFβ1) and thrombospondin 1 (TSP1). Importantly, RUNX1 knockdown impaired the metastatic capability of colorectal cancer cells in vivo and induced the development of angiogenic lesions in liver. Our results confirm that RUNX1 may be a potential target to overcome vessel co-option in CRCLM.
Collapse
Affiliation(s)
- Miran Rada
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | | | - Stephanie Petrillo
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Sébastien Tabariès
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Peter Siegel
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | | | - Anthoula Lazaris
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Peter Metrakos
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada.
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Despite significant progress, patients with metastatic prostate cancer continue to have poor prognosis. Immunotherapy has revolutionized cancer care for many tumor types but has a limited role in the treatment of prostate cancer. This review discusses the promise of immunotherapy in prostate cancer treatment with an emphasis on emerging therapeutic targets. RECENT FINDINGS Most prostate tumors have low tumor mutational burden and lack immunogenicity, representing significant hurdles to induction of anti-tumor immunity. However, recent research centered on deciphering key mechanisms of immune resistance in the prostate tumor microenvironment has led to the discovery of a range of new treatment targets. These discoveries are currently being translated into innovative immunotherapy clinical trials for patients with prostate cancer. Recent progress includes early evidence of activity for these novel approaches and the identification of potential predictive biomarkers of response. Novel treatment strategies using new antigen-directed therapies, drugs targeting the immunosuppressive tumor microenvironment, and combination immunotherapy therapies show great potential and are currently in clinical development. In addition, a deeper understanding of predictors of response and resistance to immunotherapy in prostate cancer is allowing for a more personalized approach to therapy.
Collapse
|
18
|
A cytokine in turmoil: Transforming growth factor beta in cancer. Biomed Pharmacother 2021; 139:111657. [PMID: 34243626 DOI: 10.1016/j.biopha.2021.111657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer remains one of the debilitating health threats to mankind in view of its incurable nature. Many factors are complicit in the initiation, progression and establishment of cancers. Early detection of cancer is the only window of hope that allows for appreciable management and possible limited survival. However, understanding of cancer biology and knowledge of the key factors that interplay at multi-level in the initiation and progression of cancer may hold possible avenues for cancer treatment and management. In particular, dysregulation of growth factor signaling such as that of transforming growth factor beta (TGF-β) and its downstream mediators play key roles in various cancer subtypes. Expanded understanding of the context/cell type-dependent roles of TGF-β and its downstream signaling mediators in cancer may provide leads for cancer pharmacotherapy. Reliable information contained in original articles, reviews, mini-reviews and expert opinions on TGF-β, cancer and the specific roles of TGF-β signaling in various cancer subtypes were retrieved from major scientific data bases including PubMed, Scopus, Medline, Web of Science core collections just to mention but a sample by using the following search terms: TGF-β in cancer, TGF-β and colorectal cancer, TGF-β and brain cancer, TGF-β in cancer initiation, TGF-β and cell proliferation, TGF-β and cell invasion, and TGF-β-based cancer therapy. Retrieved information and reports were carefully examined, contextualized and synchronized into a coherent scientific content to highlight the multiple roles of TGF-β signaling in normal and cancerous cells. From a conceptual standpoint, development of pharmacologically active agents that exert non-specific inhibitory effects on TGF-β signaling on various cell types will undoubtedly lead to a plethora of serious side effects in view of the multi-functionality and pleiotropic nature of TGF-β. Such non-specific targeting of TGF-β could derail any beneficial therapeutic intention associated with TGF-β-based therapy. However, development of pharmacologically active agents designed specifically to target TGF-β signaling in cancer cells may improve cancer pharmacotherapy. Similarly, specific targeting of downstream mediators of TGF-β such as TGF-β type 1 and II receptors (TβRI and TβRII), receptor-mediated Smads, mitogen activated protein kinase (MAPK) and importing proteins in cancer cells may be crucial for cancer pharmacotherapy.
Collapse
|
19
|
Gallazzi M, Baci D, Mortara L, Bosi A, Buono G, Naselli A, Guarneri A, Dehò F, Capogrosso P, Albini A, Noonan DM, Bruno A. Prostate Cancer Peripheral Blood NK Cells Show Enhanced CD9, CD49a, CXCR4, CXCL8, MMP-9 Production and Secrete Monocyte-Recruiting and Polarizing Factors. Front Immunol 2021; 11:586126. [PMID: 33569050 PMCID: PMC7868409 DOI: 10.3389/fimmu.2020.586126] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells, effector lymphocytes of the innate immunity, have been shown to be altered in several cancers, both at tissue and peripheral levels. We have shown that in Non-Small Cell Lung Cancer (NSCLC) and colon cancer, tumour associated circulating NK (TA-NK) and tumour infiltrating NK (TI-NK) exhibit pro-angiogenic phenotype/functions. However, there is still a lack of knowledge concerning the phenotype of peripheral blood (PB) NK (pNK) cells in prostate cancer (PCa). Here, we phenotypically and functionally characterized pNK from PCa patients (PCa TA-NKs) and investigated their interactions with endothelial cells and monocytes/macrophages. NK cell subset distribution in PB of PCa patients was investigated, by multicolor flow cytometry, for surface antigens expression. Protein arrays were performed to characterize the secretome on FACS-sorted pNK cells. Conditioned media (CM) from FACS-sorted PCa pTA-NKs were used to determine their ability to induce pro-inflammatory/pro-angiogenic phenotype/functions in endothelial cells, monocytes, and macrophages. CM from three different PCa (PC-3, DU-145, LNCaP) cell lines, were used to assess their effects on human NK cell polarization in vitro, by multicolor flow cytometry. We found that PCa pTA-NKs acquire the CD56brightCD9+CD49a+CXCR4+ phenotype, increased the expression of markers of exhaustion (PD-1, TIM-3) and are impaired in their degranulation capabilities. Similar effects were observed on healthy donor-derived pNK cells, exposed to conditioned media of three different PCa cell lines, together with increased production of pro-inflammatory chemokines/chemokine receptors CXCR4, CXCL8, CXCL12, reduced production of TNFα, IFNγ and Granzyme-B. PCa TA-NKs released factors able to support inflammatory angiogenesis in an in vitro model and increased the expression of CXCL8, ICAM-1, and VCAM-1 mRNA in endothelial cells. Secretome analysis revealed the ability of PCa TA-NKs to release pro-inflammatory cytokines/chemokines involved in monocyte recruitment and M2-like polarization. Finally, CMs from PCa pTA-NKs recruit THP-1 and peripheral blood CD14+ monocyte and polarize THP-1 and peripheral blood CD14+ monocyte-derived macrophage towards M2-like/TAM macrophages. Our results show that PCa pTA-NKs acquire properties related to the pro-inflammatory angiogenesis in endothelial cells, recruit monocytes and polarize macrophage to an M2-like type phenotype. Our data provides a rationale for a potential use of pNK profiling in PCa patients.
Collapse
Affiliation(s)
- Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Denisa Baci
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Annalisa Bosi
- Laboratory of Pharmacology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Angelo Naselli
- Unit of Urology, San Giuseppe Hospital, IRCCS MultiMedica, Milan, Italy
| | - Andrea Guarneri
- Unit of Urology, San Giuseppe Hospital, IRCCS MultiMedica, Milan, Italy
| | - Federico Dehò
- S.C. of Urology, ASST Settelaghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Paolo Capogrosso
- S.C. of Urology, ASST Settelaghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Adriana Albini
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, Milano, Italy
| | - Douglas M. Noonan
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, Milano, Italy
| | | |
Collapse
|
20
|
Lai KKY, Kahn M. Pharmacologically Targeting the WNT/β-Catenin Signaling Cascade: Avoiding the Sword of Damocles. Handb Exp Pharmacol 2021; 269:383-422. [PMID: 34463849 DOI: 10.1007/164_2021_523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
WNT/β-catenin signaling plays fundamental roles in numerous developmental processes and in adult tissue homeostasis and repair after injury, by controlling cellular self-renewal, activation, division, differentiation, movement, genetic stability, and apoptosis. As such, it comes as no surprise that dysregulation of WNT/β-catenin signaling is associated with various diseases, including cancer, fibrosis, neurodegeneration, etc. Although multiple agents that specifically target the WNT/β-catenin signaling pathway have been studied preclinically and a number have entered clinical trials, none has been approved by the FDA to date. In this chapter, we provide our insights as to the reason(s) it has been so difficult to safely pharmacologically target the WNT/β-catenin signaling pathway and discuss the significant efforts undertaken towards this goal.
Collapse
Affiliation(s)
- Keane K Y Lai
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Michael Kahn
- Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
21
|
Bordignon P, Bottoni G, Xu X, Popescu AS, Truan Z, Guenova E, Kofler L, Jafari P, Ostano P, Röcken M, Neel V, Dotto GP. Dualism of FGF and TGF-β Signaling in Heterogeneous Cancer-Associated Fibroblast Activation with ETV1 as a Critical Determinant. Cell Rep 2020; 28:2358-2372.e6. [PMID: 31461652 PMCID: PMC6718812 DOI: 10.1016/j.celrep.2019.07.092] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/17/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022] Open
Abstract
Heterogeneity of cancer-associated fibroblasts (CAFs) can result from activation of distinct signaling pathways. We show that in primary human dermal fibroblasts (HDFs), fibroblast growth factor (FGF) and transforming growth factor β (TGF-β) signaling oppositely modulate multiple CAF effector genes. Genetic abrogation or pharmacological inhibition of either pathway results in induction of genes responsive to the other, with the ETV1 transcription factor mediating the FGF effects. Duality of FGF/TGF-β signaling and differential ETV1 expression occur in multiple CAF strains and fibroblasts of desmoplastic versus non-desmoplastic skin squamous cell carcinomas (SCCs). Functionally, HDFs with opposite TGF-β versus FGF modulation converge on promoting cancer cell proliferation. However, HDFs with increased TGF-β signaling enhance invasive properties and epithelial-mesenchymal transition (EMT) of SCC cells, whereas HDFs with increased FGF signaling promote macrophage infiltration. The findings point to a duality of FGF versus TGF-β signaling in distinct CAF populations that promote cancer development through modulation of different processes. FGF and TGF-β signaling exert opposite control over multiple CAF effector genes ETV1 transcription factor mediates FGF effects and suppresses those of TGF-β Modulation of either pathway leads to different tumor-promoting CAF populations TGF-β-activated CAFs promote EMT, but FGF-activated CAFs increase inflammation
Collapse
Affiliation(s)
- Pino Bordignon
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Giulia Bottoni
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiaoying Xu
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Alma S Popescu
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Zinnia Truan
- Department of Otolaryngology-Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich 8091, Switzerland
| | - Lukas Kofler
- Department of Dermatology, Eberhard Karls University, Tübingen 72076, Germany
| | - Paris Jafari
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; International Cancer Prevention Institute, Epalinges 1066, Switzerland
| | - Paola Ostano
- Cancer Genomics Laboratory, Edo and Elvo Tempia Valenta Foundation, Biella 13900, Italy
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen 72076, Germany
| | - Victor Neel
- Department of Dermatology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - G Paolo Dotto
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; International Cancer Prevention Institute, Epalinges 1066, Switzerland.
| |
Collapse
|
22
|
Brasil da Costa FH, Lewis MS, Truong A, Carson DD, Farach-Carson MC. SULF1 suppresses Wnt3A-driven growth of bone metastatic prostate cancer in perlecan-modified 3D cancer-stroma-macrophage triculture models. PLoS One 2020; 15:e0230354. [PMID: 32413029 PMCID: PMC7228113 DOI: 10.1371/journal.pone.0230354] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/24/2020] [Indexed: 12/29/2022] Open
Abstract
Bone marrow stroma influences metastatic prostate cancer (PCa) progression, latency, and recurrence. At sites of PCa bone metastasis, cancer-associated fibroblasts and tumor-associated macrophages interact to establish a perlecan-rich desmoplastic stroma. As a heparan sulfate proteoglycan, perlecan (HSPG2) stores and stabilizes growth factors, including heparin-binding Wnt3A, a positive regulator of PCa cell growth. Because PCa cells alone do not induce CAF production of perlecan in the desmoplastic stroma, we sought to discover the sources of perlecan and its growth factor-releasing modifiers SULF1, SULF2, and heparanase in PCa cells and xenografts, bone marrow fibroblasts, and macrophages. SULF1, produced primarily by bone marrow fibroblasts, was the main glycosaminoglycanase present, a finding validated with primary tissue specimens of PCa metastases with desmoplastic bone stroma. Expression of both HSPG2 and SULF1 was concentrated in αSMA-rich stroma near PCa tumor nests, where infiltrating pro-tumor TAMs also were present. To decipher SULF1's role in the reactive bone stroma, we created a bone marrow biomimetic hydrogel incorporating perlecan, PCa cells, macrophages, and fibroblastic bone marrow stromal cells. Finding that M2-like macrophages increased levels of SULF1 and HSPG2 produced by fibroblasts, we examined SULF1 function in Wnt3A-mediated PCa tumoroid growth in tricultures. Comparing control or SULF1 knockout fibroblastic cells, we showed that SULF1 reduces Wnt3A-driven growth, cellularity, and cluster number of PCa cells in our 3D model. We conclude that SULF1 can suppress Wnt3A-driven growth signals in the desmoplastic stroma of PCa bone metastases, and SULF1 loss favors PCa progression, even in the presence of pro-tumorigenic TAMs.
Collapse
Affiliation(s)
- Fabio Henrique Brasil da Costa
- Biosciences Department, Rice University, Houston, TX, United States of America
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center School of Dentistry, Houston, TX, United States of America
| | - Michael S. Lewis
- Department of Pathology and Medicine, Cedars-Sinai Medical Center, West Hollywood, CA, United States of America
| | - Anna Truong
- Department of Chemistry, Rice University, Houston, TX, United States of America
| | - Daniel D. Carson
- Biosciences Department, Rice University, Houston, TX, United States of America
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Mary C. Farach-Carson
- Biosciences Department, Rice University, Houston, TX, United States of America
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center School of Dentistry, Houston, TX, United States of America
- Department of Bioengineering, Rice University, Houston, TX, United States of America
| |
Collapse
|
23
|
Hu M, Xie J, Liu Z, Wang X, Liu M, Wang J. Comprehensive Analysis Identifying Wnt Ligands Gene Family for Biochemical Recurrence in Prostate Adenocarcinoma and Construction of a Nomogram. J Comput Biol 2020; 27:1656-1667. [PMID: 32298604 DOI: 10.1089/cmb.2019.0397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
There is little research to explore the relationship between Wnt ligands gene family and biochemical recurrence of prostate adenocarcinoma. The purpose of this study was to systematically evaluate the role of Wnt ligands gene family in biochemical recurrence in prostate adenocarcinoma. RNA-seq transcriptome data and clinicopathological data of 489 prostate adenocarcinoma tissues and 51 nontumor tissues were obtained from The Cancer Genome Atlas. We developed a risk score model with the least absolute shrinkage and selection operator Cox regression algorithm. We used the X-tile program to derive the best threshold for risk scores, dividing patients into high-, intermediate-, and low-risk groups. Gene set enrichment analysis (GSEA) was performed. Nomogram was constructed based on the risk score and clinical features. The risk score = (0.192 × expression level of Wnt9A) + (0.732 × expression level of Wnt8B) + (0.051 × expression level of Wnt7B) + (-0.320 × expression level of Wnt3A). The risk score was an independent prognostic factor, with a hazard ratio of 1.298 (95% confidence interval: 1.046-1.612; p = 0.018). GSEA revealed that the Kyoto Encyclopedia of Genes and Genomes pathway of the four selected genes was closely related to malignancy-related biological processes. Nomogram was constructed based on the risk score and clinical features. The C index was 0.719, and the calibration curve showed that the nomogram performed well. In general, we comprehensively evaluated the association between Wnt ligands gene family and biochemical recurrence of prostate cancer. We developed a risk score model based on messenger RNA expression levels of several selected Wnt ligand family genes (Wnt3A, Wnt7B, Wnt8B, and Wnt9A), which was significantly associated with biochemical recurrence of prostate cancer. Our results might be helpful for future molecular studies focusing on the biochemical recurrence of prostate cancer.
Collapse
Affiliation(s)
- Maolin Hu
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, China.,Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jiangling Xie
- Department of Urology, Qianjiang Central Hospital of Chongqing Municipality, Chongqing, China
| | - Zhifeng Liu
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, China.,Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xuan Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Ming Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jianye Wang
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, China.,Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| |
Collapse
|
24
|
Suryawanshi A, Hussein MS, Prasad PD, Manicassamy S. Wnt Signaling Cascade in Dendritic Cells and Regulation of Anti-tumor Immunity. Front Immunol 2020; 11:122. [PMID: 32132993 PMCID: PMC7039855 DOI: 10.3389/fimmu.2020.00122] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 01/16/2020] [Indexed: 01/26/2023] Open
Abstract
Dendritic cells (DCs) control the strength and quality of antigen-specific adaptive immune responses. This is critical for launching a robust immunity against invading pathogens while maintaining a state of tolerance to self-antigens. However, this also represents a fundamental barrier to anti-tumor immune responses and cancer immunotherapy. DCs in the tumor microenvironment (TME) play a key role in this process. The factors in the TME and signaling networks that program DCs to a regulatory state are not fully understood. Recent advances point to novel mechanisms by which the canonical Wnt signaling cascade in DCs regulates immune suppression, and the same pathway in tumors is associated with the evasion of anti-tumor immunity. Here, we review these recent advances in the context of the pleiotropic effects of the Wnts in shaping anti-tumor immune responses by modulating DC functions. In addition, we will discuss how Wnt/β-catenin pathway in DCs can be targeted for successful cancer immunotherapy.
Collapse
Affiliation(s)
- Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Mohamed S Hussein
- Georgia Cancer Center, Augusta University, Augusta, GA, United States
| | - Puttur D Prasad
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Santhakumar Manicassamy
- Georgia Cancer Center, Augusta University, Augusta, GA, United States.,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
25
|
Vickman RE, Broman MM, Lanman NA, Franco OE, Sudyanti PAG, Ni Y, Ji Y, Helfand BT, Petkewicz J, Paterakos MC, Crawford SE, Ratliff TL, Hayward SW. Heterogeneity of human prostate carcinoma-associated fibroblasts implicates a role for subpopulations in myeloid cell recruitment. Prostate 2020; 80:173-185. [PMID: 31763714 PMCID: PMC12004274 DOI: 10.1002/pros.23929] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/10/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Carcinoma-associated fibroblasts (CAF) are a heterogeneous group of cells within the tumor microenvironment (TME) that can promote tumorigenesis in the prostate. By understanding the mechanism(s) by which CAF contributes to tumor growth, new therapeutic targets for the management of this disease may be identified. These studies determined whether unique sub-populations of human prostate CAF can be identified and functionally characterized. METHODS Single-cell RNA-seq of primary human prostate CAF followed by unsupervised clustering was utilized to generate cell clusters based on differentially expressed (DE) gene profiles. Potential communication between CAF and immune cells was analyzed using in vivo tissue recombination by combining CAF or normal prostate fibroblasts (NPF) with non-tumorigenic, initiated prostate epithelial BPH-1 cells. Resultant grafts were assessed for inflammatory cell recruitment. RESULTS Clustering of 3321 CAF allows for visualization of six subpopulations, demonstrating heterogeneity within CAF. Sub-renal capsule recombination assays show that the presence of CAF significantly increases myeloid cell recruitment to resultant tumors. This is supported by significantly increased expression of chemotactic chemokines CCL2 and CXCL12 in large clusters compared to other subpopulations. Bayesian analysis topologies also support differential communication signals between chemokine-related genes of individual clusters. Migration of THP-1 monocyte cells in vitro is stimulated in the presence of CAF conditioned medium (CM) compared with NPF CM. Further in vitro analyses suggest that CAF-derived chemokine CCL2 may be responsible for CAF-stimulated migration of THP-1 cells, since neutralization of this chemokine abrogates migration capacity. CONCLUSIONS CAF clustering based on DE gene expression supports the concept that clusters have unique functions within the TME, including a role in immune/inflammatory cell recruitment. These data suggest that CCL2 produced by CAF may be involved in the recruitment of inflammatory cells, but may also directly regulate the growth of the tumor. Further studies aimed at characterizing the subpopulation(s) of CAF which promote immune cell recruitment to the TME and/or stimulate prostate cancer growth and progression will be pursued.
Collapse
Affiliation(s)
- Renee E. Vickman
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Meaghan M. Broman
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Nadia A. Lanman
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Omar E. Franco
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | | | - Yang Ni
- Department of Statistics, Texas A&M University, College Station, Texas
| | - Yuan Ji
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Brian T. Helfand
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | | | | | - Susan E. Crawford
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Timothy L. Ratliff
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Simon W. Hayward
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| |
Collapse
|
26
|
Boumahdi S, de Sauvage FJ. The great escape: tumour cell plasticity in resistance to targeted therapy. Nat Rev Drug Discov 2020; 19:39-56. [PMID: 31601994 DOI: 10.1038/s41573-019-0044-1] [Citation(s) in RCA: 455] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2019] [Indexed: 01/05/2023]
Abstract
The success of targeted therapies in cancer treatment has been impeded by various mechanisms of resistance. Besides the acquisition of resistance-conferring genetic mutations, reversible mechanisms that lead to drug tolerance have emerged. Plasticity in tumour cells drives their transformation towards a phenotypic state that no longer depends on the drug-targeted pathway. These drug-refractory cells constitute a pool of slow-cycling cells that can either regain drug sensitivity upon treatment discontinuation or acquire permanent resistance to therapy and drive relapse. In the past few years, cell plasticity has emerged as a mode of targeted therapy evasion in various cancers, ranging from prostate and lung adenocarcinoma to melanoma and basal cell carcinoma. Our understanding of the mechanisms that control this phenotypic switch has also expanded, revealing the crucial role of reprogramming factors and chromatin remodelling. Further deciphering the molecular basis of tumour cell plasticity has the potential to contribute to new therapeutic strategies which, combined with existing anticancer treatments, could lead to deeper and longer-lasting clinical responses.
Collapse
Affiliation(s)
- Soufiane Boumahdi
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | | |
Collapse
|
27
|
Yu C, Hu K, Nguyen D, Wang ZA. From genomics to functions: preclinical mouse models for understanding oncogenic pathways in prostate cancer. Am J Cancer Res 2019; 9:2079-2102. [PMID: 31720076 PMCID: PMC6834478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/10/2019] [Indexed: 06/10/2023] Open
Abstract
Next-generation sequencing has revealed numerous genomic alterations that induce aberrant signaling activities in prostate cancer (PCa). Among them are pathways affecting multiple cancer types, including the PI3K/AKT/mTOR, p53, Rb, Ras/Raf/MAPK, Myc, FGF, and Wnt signaling pathways, as well as ones that are prominent in PCa, including alterations in genes of AR signaling, the ETS family, NKX3.1, and SPOP. Cross talk among the oncogenic pathways can confer PCa resistance to therapy, particularly in advanced tumors, which are castration-resistant or show neuroendocrine features. Various experimental models, such as cancer cell lines, animal models, and patient-derived xenografts and organoids have been utilized to dissect PCa progression mechanisms. Here, we review the current preclinical mouse models for studying the most commonly altered pathways in PCa, with an emphasis on their interplays. We highlight the power of genetically engineered mouse models (GEMMs) in translating genomic discoveries into understanding of the functions of these oncogenic events in vivo. Developing and analyzing PCa mouse models will undoubtedly continue to offer new insights into tumor biology and guide novel rationalized therapy.
Collapse
Affiliation(s)
- Chuan Yu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Kevin Hu
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Daniel Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| | - Zhu A Wang
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA 95064, USA
| |
Collapse
|
28
|
Kothari V, Goodwin JF, Zhao SG, Drake JM, Yin Y, Chang SL, Evans JR, Wilder-Romans K, Gabbara K, Dylgjeri E, Chou J, Sun G, Tomlins SA, Mehra R, Hege K, Filvaroff EH, Schaeffer EM, Karnes RJ, Quigley DA, Rathkopf DE, He HH, Speers C, Spratt DE, Gilbert LA, Ashworth A, Chinnaiyan AM, Raj GV, Knudsen KE, Feng FY. DNA-Dependent Protein Kinase Drives Prostate Cancer Progression through Transcriptional Regulation of the Wnt Signaling Pathway. Clin Cancer Res 2019; 25:5608-5622. [PMID: 31266829 PMCID: PMC6744969 DOI: 10.1158/1078-0432.ccr-18-2387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/07/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Protein kinases are known to play a prominent role in oncogenic progression across multiple cancer subtypes, yet their role in prostate cancer progression remains underexplored. The purpose of this study was to identify kinases that drive prostate cancer progression.Experimental Design: To discover kinases that drive prostate cancer progression, we investigated the association between gene expression of all known kinases and long-term clinical outcomes in tumor samples from 545 patients with high-risk disease. We evaluated the impact of genetic and pharmacologic inhibition of the most significant kinase associated with metastatic progression in vitro and in vivo. RESULTS DNA-dependent protein kinase (DNAPK) was identified as the most significant kinase associated with metastatic progression in high-risk prostate cancer. Inhibition of DNAPK suppressed the growth of both AR-dependent and AR-independent prostate cancer cells. Gene set enrichment analysis nominated Wnt as the top pathway associated with DNAPK. We found that DNAPK interacts with the Wnt transcription factor LEF1 and is critical for LEF1-mediated transcription. CONCLUSIONS Our data show that DNAPK drives prostate cancer progression through transcriptional regulation of Wnt signaling and is an attractive therapeutic target in aggressive prostate cancer.
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Radiation Oncology, University of California at San Francisco, CA
| | - Jonathan F Goodwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shuang G Zhao
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Justin M Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Yi Yin
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - S Laura Chang
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Joseph R Evans
- Department of Radiation Oncology, OSF Healthcare, Peoria, Illinois
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Kristina Gabbara
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jonathan Chou
- Department of Medicine, University of California at San Francisco, San Francisco, California
| | - Grace Sun
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Scott A Tomlins
- Department of Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Rohit Mehra
- Department of Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | - Edward M Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - David A Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | | | - Housheng H He
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Luke A Gilbert
- Department of Urology, University of California at San Francisco, San Francisco, California
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan-Ann Arbor, Ann Arbor, Michigan
| | - Ganesh V Raj
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Felix Y Feng
- Department of Radiation Oncology, University of California at San Francisco, CA.
- Department of Medicine, University of California at San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California
- Department of Urology, University of California at San Francisco, San Francisco, California
| |
Collapse
|
29
|
SILAC-Based Quantification of TGFBR2-Regulated Protein Expression in Extracellular Vesicles of Microsatellite Unstable Colorectal Cancers. Int J Mol Sci 2019; 20:ijms20174162. [PMID: 31454892 PMCID: PMC6747473 DOI: 10.3390/ijms20174162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022] Open
Abstract
Microsatellite unstable (MSI) colorectal cancers (CRCs) are characterized by mutational inactivation of Transforming Growth Factor Beta Receptor Type 2 (TGFBR2). TGFBR2-deficient CRCs present altered target gene and protein expression. Such cellular alterations modulate the content of CRC-derived extracellular vesicles (EVs). EVs function as couriers of proteins, nucleic acids, and lipids in intercellular communication. At a qualitative level, we have previously shown that TGFBR2 deficiency causes overall alterations in the EV protein content. To deepen the basic understanding of altered protein dynamics, this work aimed to determine TGFBR2-dependent EV protein signatures in a quantitative manner. Using a stable isotope labeling with amino acids in cell culture (SILAC) approach for mass spectrometry-based quantification, 48 TGFBR2-regulated proteins were identified in MSI CRC-derived EVs. Overall, TGFBR2 deficiency caused upregulation of several EV proteins related to the extracellular matrix and nucleosome as well as downregulation of proteasome-associated proteins. The present study emphasizes the general overlap of proteins between EVs and their parental CRC cells but also highlights the impact of TGFBR2 deficiency on EV protein composition. From a clinical perspective, TGFBR2-regulated quantitative differences of protein expression in EVs might nominate novel biomarkers for liquid biopsy-based MSI typing in the future.
Collapse
|
30
|
Zhang D, Li G, Chen X, Jing Q, Liu C, Lu S, Huang D, Wang Y, Tan P, Chen J, Zhang X, Qiu Y, Liu Y. Wnt3a protein overexpression predicts worse overall survival in laryngeal squamous cell carcinoma. J Cancer 2019; 10:4633-4638. [PMID: 31528227 PMCID: PMC6746142 DOI: 10.7150/jca.35009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/19/2019] [Indexed: 12/25/2022] Open
Abstract
As a classical ligand in the canonical Wnt/β-catenin signaling pathway, the role of Wnt3a in laryngeal squamous cell carcinoma (LSCC) remains unclear. Therefore, the expression pattern of the Wnt3a protein in 222 primary LSCC, and 19 corresponding adjacent non-carcinoma specimens, was detected by immunohistochemistry and further correlated with clinicopathological parameters. The results showed that LSCC tissue expressed higher levels of the Wnt3a protein when compared to the corresponding adjacent non-cancerous tissues. High expression of Wnt3a was closely related to histological grade (P = 0.031), clinical stage (I+II / III+IV; P = 0.004), and lymph node metastasis (P = 0.03). Kaplan-Meier analysis evidenced that a worse overall survival (OS) was correlated to the group with high Wnt3a expression (P = 0.003). When stratified survival analyses were performed, patients with lymph node metastasis/advanced clinical stages and high Wnt3a expression had worse OS rates than patients with other features (P < 0.001). Finally, multivariate analysis showed that Wnt3a expression was an independent prognosis factor for LSCC patients. The current findings suggest that Wnt3a is tightly related to the LSCC progression and could serve as a valuable clinic biomarker for LSCC patients.
Collapse
Affiliation(s)
- Diekuo Zhang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Guo Li
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Xiyu Chen
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Qiancheng Jing
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Department of Otolaryngology, Head and Neck Surgery, Changsha Central Hospital, 161 Shaoshan Road, Changsha, Hunan 410004, People's Republic of China
| | - Chao Liu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Shanhong Lu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Donghai Huang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Yunyun Wang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Pingqing Tan
- Department of Head and Neck Surgery, Hunan Cancer Hospital, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, 283 Tongzipo Road, Changsha, Hunan 410013, People's Republic of China
| | - Jie Chen
- Department of Head and Neck Surgery, Hunan Cancer Hospital, The Affiliated Tumor Hospital of Xiangya Medical School, Central South University, 283 Tongzipo Road, Changsha, Hunan 410013, People's Republic of China
| | - Xin Zhang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Yuanzheng Qiu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Yong Liu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Otolaryngology Major Disease Research, Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| |
Collapse
|
31
|
Park JH, Kim YH, Park EH, Lee SJ, Kim H, Kim A, Lee SB, Shim S, Jang H, Myung JK, Park S, Lee SJ, Kim MJ. Effects of metformin and phenformin on apoptosis and epithelial-mesenchymal transition in chemoresistant rectal cancer. Cancer Sci 2019; 110:2834-2845. [PMID: 31278880 PMCID: PMC6726705 DOI: 10.1111/cas.14124] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022] Open
Abstract
Recurrence and chemoresistance in colorectal cancer remain important issues for patients treated with conventional therapeutics. Metformin and phenformin, previously used in the treatment of diabetes, have been shown to have anticancer effects in various cancers, including breast, lung and prostate cancers. However, their molecular mechanisms are still unclear. In this study, we examined the effects of these drugs in chemoresistant rectal cancer cell lines. We found that SW837 and SW1463 rectal cancer cells were more resistant to ionizing radiation and 5‐fluorouracil than HCT116 and LS513 colon cancer cells. In addition, metformin and phenformin increased the sensitivity of these cell lines by inhibiting cell proliferation, suppressing clonogenic ability and increasing apoptotic cell death in rectal cancer cells. Signal transducer and activator of transcription 3 and transforming growth factor‐β/Smad signaling pathways were more activated in rectal cancer cells, and inhibition of signal transducer and activator of transcription 3 expression using an inhibitor or siRNA sensitized rectal cancer cells to chemoresistant by inhibition of the expression of antiapoptotic proteins, such as X‐linked inhibitor of apoptosis, survivin and cellular inhibitor of apoptosis protein 1. Moreover, metformin and phenformin inhibited cell migration and invasion by suppression of transforming growth factor β receptor 2‐mediated Snail and Twist expression in rectal cancer cells. Therefore, metformin and phenformin may represent a novel strategy for the treatment of chemoresistant rectal cancer by targeting signal transducer and activator of transcription 3 and transforming growth factor‐β/Smad signaling.
Collapse
Affiliation(s)
- Ji-Hye Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Young-Heon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Eun Hyeh Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sun-Joo Lee
- Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Hyewon Kim
- Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Areumnuri Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea.,Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea.,Laboratory of Experimental Pathology, Department of Pathology, Korea Institute of Radiological & Medical Science, Seoul, Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Min Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, Korea
| |
Collapse
|
32
|
Han Q, Wang X, Liao X, Han C, Yu T, Yang C, Li G, Han B, Huang K, Zhu G, Liu Z, Zhou X, Su H, Shang L, Gong Y, Song X, Peng T, Ye X. Diagnostic and prognostic value of WNT family gene expression in hepatitis B virus‑related hepatocellular carcinoma. Oncol Rep 2019; 42:895-910. [PMID: 31322232 PMCID: PMC6667889 DOI: 10.3892/or.2019.7224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to investigate the diagnostic and prognostic value of Wingless-type MMTV integration site (WNT) gene family expression in patients with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). The clinical data of the patients and gene expression levels were downloaded from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. Receiver operating characteristic curve analysis was used to investigate the diagnostic value of WNT genes. Cox proportional hazard regression analysis and Kaplan-Meier survival analysis were performed to evaluate the association of WNT gene expression level with overall survival (OS) and recurrence-free survival (RFS). A nomogram was constructed for the prediction of prognosis. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. Diagnostic receiver operating characteristic curve analysis suggested that WNT2 had a high diagnostic value, with an area under the curve (AUC) of >0.800 (P<0.0001, AUC=0.810, 95% CI: 0.767–0.852). Survival analysis indicated that the expression level of WNT1 was significantly associated with OS and RFS (adjusted P=0.033, adjusted HR=0.607, 95% CI: 0.384–0.960; and adjusted P=0.007, adjusted HR=0.592, 95% CI: 0.404–0.868, respectively). In the TCGA validation cohort, we also observed that WNT2 was significantly differentially expressed between HCC tissues and adjacent non-tumor tissues, and WNT1 was associated with both the OS and RFS of HCC. Therefore, through the GSE14520 HBV-related HCC cohort we concluded that WNT2 may serve as a diagnostic biomarker and WNT1 may serve as a prognostic biomarker. These results may also be extended to TCGA HCC verification cohort.
Collapse
Affiliation(s)
- Quanfa Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guanghui Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Bowen Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhengqian Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Liming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yizhen Gong
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaowei Song
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
33
|
Role of Wnt3a in the pathogenesis of cancer, current status and prospective. Mol Biol Rep 2019; 46:5609-5616. [PMID: 31236761 DOI: 10.1007/s11033-019-04895-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 05/24/2019] [Indexed: 12/18/2022]
Abstract
The Wnt signaling pathway plays a critical role in initiation, progression, invasion and metastasis of cancer. Wnt3a as a canonical Wnt ligand is strongly implicated in the etiology and pathology of a number of diseases including cancer. Depending on cancer type, Wnt3a enhances or suppresses metastasis, cell proliferation and apoptosis of cancer cells. This review summarizes the role of Wnt3a in the pathogenesis of different cancers including colorectal, prostate, hepatocellular, lung and leukemia, for promoting greater understanding and clinical management of these diseases.
Collapse
|
34
|
Bhattacharyya S, Feferman L, Tobacman JK. Dihydrotestosterone inhibits arylsulfatase B and Dickkopf Wnt signaling pathway inhibitor (DKK)-3 leading to enhanced Wnt signaling in prostate epithelium in response to stromal Wnt3A. Prostate 2019; 79:689-700. [PMID: 30801800 DOI: 10.1002/pros.23776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/23/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND In tissue microarrays, immunostaining of the enzyme arylsulfatase B (ARSB; N-acetylgalactosamine-4-sulfatase) was less in recurrent prostate cancers and in cancers with higher Gleason scores. In cultured prostate stem cells, decline in ARSB increased Wnt signaling through effects on Dickkopf Wnt Signaling Pathway Inhibitor (DKK)3. The effects of androgen exposure on ARSB and the impact of decline in ARSB on Wnt signaling in prostate tissue were unknown. METHODS Epithelial and stromal tissues from malignant and normal human prostate were obtained by laser capture microdissection. mRNA expression of ARSB, galactose-6-sulfate-sulfatase (GALNS) and Wnt-signaling targets was determined by QPCR. Non-malignant human epithelial and stromal prostate cells were grown in tissue culture, including two-cell layer cultures. ARSB was silenced by specific siRNA, and epithelial cells were treated with stromal spent media following treatment with IWP-2, an inhibitor of Wnt secretion, and by exogenous recombinant human Wnt3A. Promoter methylation was detected using specific DKK3 and ARSB promoter primers. The effects of DHT and of ARSB overexpression on DKK expression were determined. Cell proliferation was assessed by BrdU incorporation. RESULTS Normal stroma showed higher expression of vimentin, ARSB, and Wnt3A than epithelium. Normal epithelium had higher expression of E-cadherin, galactose 6-sulfate-sulfatase (GALNS), and DKK3 than stroma. In malignant epithelium, expression of ARSB and DKK3 declined, and expression of GALNS and Wnt signaling targets increased. In cultured prostate epithelial cells, Wnt-mediated signaling was greatest when ARSB was silenced and cells were exposed to exogenous Wnt3A. Exposure to 5α-dihydrotestosterone (DHT) increased ARSB and DKK3 promoter rmethylation, and effects of DHT on DKK3 expression were reversed when ARSB was overexpressed. CONCLUSIONS Androgen-induced declines in ARSB and DKK3 may contribute to prostate carcinogenesis by sustained activation of Wnt signaling in prostate epithelium in response to stromal Wnt3A.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- Department of Medicine, The University of Illinois at Chicago and Jesse Brown VAMC, Chicago, Illinois
| | - Leo Feferman
- Department of Medicine, The University of Illinois at Chicago and Jesse Brown VAMC, Chicago, Illinois
| | - Joanne K Tobacman
- Department of Medicine, The University of Illinois at Chicago and Jesse Brown VAMC, Chicago, Illinois
| |
Collapse
|
35
|
ZENGIN MEHMET. A different perspective to the tumor microenvironment in periampullary cancers: a neglected ring in tumorogenesis. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2019. [DOI: 10.32322/jhsm.453541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
36
|
Enabling precision medicine by unravelling disease pathophysiology: quantifying signal transduction pathway activity across cell and tissue types. Sci Rep 2019; 9:1603. [PMID: 30733525 PMCID: PMC6367506 DOI: 10.1038/s41598-018-38179-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
Signal transduction pathways are important in physiology and pathophysiology. Targeted drugs aim at modifying pathogenic pathway activity, e.g., in cancer. Optimal treatment choice requires assays to measure pathway activity in individual patient tissue or cell samples. We developed a method enabling quantitative measurement of functional pathway activity based on Bayesian computational model inference of pathway activity from measurements of mRNA levels of target genes of the pathway-associated transcription factor. Oestrogen receptor, Wnt, and PI3K-FOXO pathway assays have been described previously. Here, we report model development for androgen receptor, Hedgehog, TGFβ, and NFκB pathway assays, biological validation on multiple cell types, and analysis of data from published clinical studies (multiple sclerosis, amyotrophic lateral sclerosis, contact dermatitis, Ewing sarcoma, lymphoma, medulloblastoma, ependymoma, skin and prostate cancer). Multiple pathway analysis of clinical prostate cancer (PCa) studies showed increased AR activity in hyperplasia and primary PCa but variable AR activity in castrate resistant (CR) PCa, loss of TGFβ activity in PCa, increased Wnt activity in TMPRSS2:ERG fusion protein-positive PCa, active PI3K pathway in advanced PCa, and active PI3K and NFκB as potential hormonal resistance pathways. Potential value for future clinical practice includes disease subtyping and prediction and targeted therapy response prediction and monitoring.
Collapse
|
37
|
Sung JY, Yoon K, Ye SK, Goh SH, Park SY, Kim JH, Kang HG, Kim YN, Park BK. Upregulation of transforming growth factor-beta type I receptor by interferon consensus sequence-binding protein in osteosarcoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:761-772. [PMID: 30710564 DOI: 10.1016/j.bbamcr.2019.01.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
Abstract
Transforming growth factor-beta (TGF-β) is a known tumor suppressor, which also exerts a tumor promoting activity at an advanced stage of cancer. Previously, we reported that expression of interferon consensus sequence-binding protein (ICSBP), also known as interferon regulatory factor-8, is positively correlated with TGF-β type I receptor (TGF-β RI) expression in osteosarcoma patient tissues. In this study, we demonstrated that ICSBP upregulated TGF-β RI and induced epithelial-to-mesenchymal transition-like phenomena in human osteosarcoma cell lines. As determined by soft agar growth of osteosarcoma cells and xenografted mouse models, ICSBP increased tumorigenicity, which was reversed by ICSBP knock-down or a TGF-β RI inhibitor. To test whether ICSBP directly regulates the promoter activity of TGF-β RI, we performed a TGF-β RI promoter assay, an electro mobility shift assay, and a chromatin immunoprecipitation assay. We observed that TGF-β RI promoter was activated in ICSBP-overexpressing osteosarcoma cells. Exploiting serial deletions and mutations of the TGF-β RI promoter, we found a putative ICSBP-binding site at nucleotides -216/-211 (GGXXTC) in the TGF-β RI promoter. Our data suggest that ICSBP upregulates TGF-β RI expression by binding to this site, causing ICSBP-mediated tumor progression in osteosarcoma cells. In addition, we found a positive correlation between ICSBP and TGF-β RI expression in several types of tumors using the cBioportal database. SUMMARY: We demonstrated that interferon consensus sequence-binding protein upregulates transforming growth factor-beta type I receptor (TGF-β RI) expression by binding to nucleotides -216/-211 (GGXXTC) in the TGF-β RI promoter, which resulted in increased tumorigenicity and tumor progression in human osteosarcoma cells.
Collapse
Affiliation(s)
- Jee Young Sung
- Rare Cancer Branch, Division of Clinical Research, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Kyungsil Yoon
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Sang-Kyu Ye
- Department of Pharmacology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Sung-Ho Goh
- Precision Medicine Branch, Division of Precision Medicine, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - June Hyuk Kim
- Orthopaedic Oncology Clinic, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Hyun Guy Kang
- Orthopaedic Oncology Clinic, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Yong-Nyun Kim
- Comparative Biomedicine Research Branch, Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea.
| | - Byung-Kiu Park
- Rare Cancer Branch, Division of Clinical Research, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea.
| |
Collapse
|
38
|
Loss of Myeloid-Specific TGF-β Signaling Decreases CTHRC1 to Downregulate bFGF and the Development of H1993-Induced Osteolytic Bone Lesions. Cancers (Basel) 2018; 10:cancers10120463. [PMID: 30469488 PMCID: PMC6315699 DOI: 10.3390/cancers10120463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/05/2018] [Accepted: 11/21/2018] [Indexed: 12/19/2022] Open
Abstract
The role of myeloid cell-specific TGF-β signaling in non-small-cell lung cancer (NSCLC)-induced osteolytic bone lesion development is unknown. We used a genetically engineered mouse model, Tgfbr2LysMCre knockout (KO), which has a loss of TGF-β signaling specifically in myeloid lineage cells, and we found that the area of H1993 cell-induced osteolytic bone lesions was decreased in Tgfbr2LysMCre KO mice, relative to the area in control littermates. The bone lesion areas were correlated with tumor cell proliferation, angiogenesis, and osteoclastogenesis in the microenvironment. The smaller bone lesion area was partially rescued by bFGF, which was expressed by osteoblasts. Interestingly, bFGF was able to rescue the osteoclastogenesis, but not the tumor cell proliferation or angiogenesis. We then focused on identifying osteoclast factors that regulate bFGF expression in osteoblasts. We found that the expression and secretion of CTHRC1 was downregulated in osteoclasts from Tgfbr2LysMCre KO mice; CTHRC1 was able to promote bFGF expression in osteoblasts, possibly through the Wnt/β-catenin pathway. Functionally, bFGF stimulated osteoclastogenesis and inhibited osteoblastogenesis, but had no effect on H1993 cell proliferation. On the other hand, CTHRC1 promoted osteoblastogenesis and H1993 cell proliferation. Together, our data show that myeloid-specific TGF-β signaling promoted osteolytic bone lesion development and bFGF expression in osteoblasts; that osteoclast-secreted CTHRC1 stimulated bFGF expression in osteoblasts in a paracrine manner; and that CTHRC1 and bFGF had different cell-specific functions that contributed to bone lesion development.
Collapse
|
39
|
Heterogeneous cancer-associated fibroblast population potentiates neuroendocrine differentiation and castrate resistance in a CD105-dependent manner. Oncogene 2018; 38:716-730. [PMID: 30177832 PMCID: PMC7182071 DOI: 10.1038/s41388-018-0461-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/27/2018] [Accepted: 07/24/2018] [Indexed: 11/20/2022]
Abstract
Heterogeneous prostatic carcinoma associated fibroblasts (CAF) contribute to tumor progression and resistance to androgen signaling deprivation therapy (ADT). CAF subjected to extended passaging, compared to low passage CAF, were found to lose tumor expansion potential and heterogeneity. Cell surface endoglin (CD105), known to be expressed on proliferative endothelia and mesenchymal stem cells, was diminished in high passage CAF. RNA-sequencing revealed SFRP1 to be distinctly expressed by tumor-inductive CAF, which was further demonstrated to occur in a CD105-dependent manner. Moreover, ADT resulted in further expansion of the CD105+ fibroblastic population and downstream SFRP1 in 3-dimensional cultures and patient derived xenograft tissues. In patients, CD105+ fibroblasts were found to circumscribe epithelia with neuroendocrine differentiation. CAF-derived SFRP1, driven by CD105 signaling, was necessary and sufficient to induce prostate cancer neuroendocrine differentiation in a paracrine manner. A partially humanized CD105 neutralizing antibody, TRC105, inhibited fibroblastic SFRP1 expression and epithelial neuroendocrine differentiation. In a novel synthetic lethality paradigm, we found that simultaneously targeting the epithelia and its microenvironment with ADT and TRC105, respectively, reduced castrate resistant tumor progression, in a model where either ADT or TRC105 alone had little effect.
Collapse
|
40
|
Pacella I, Cammarata I, Focaccetti C, Miacci S, Gulino A, Tripodo C, Ravà M, Barnaba V, Piconese S. Wnt3a Neutralization Enhances T-cell Responses through Indirect Mechanisms and Restrains Tumor Growth. Cancer Immunol Res 2018; 6:953-964. [DOI: 10.1158/2326-6066.cir-17-0713] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 05/16/2018] [Accepted: 06/13/2018] [Indexed: 11/16/2022]
|
41
|
Vander Ark A, Cao J, Li X. Mechanisms and Approaches for Overcoming Enzalutamide Resistance in Prostate Cancer. Front Oncol 2018; 8:180. [PMID: 29911070 PMCID: PMC5992404 DOI: 10.3389/fonc.2018.00180] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
Enzalutamide, a second-generation small-molecule inhibitor of the androgen receptor (AR), has been approved for patients who failed with androgen deprivation therapy and have developed castration-resistant prostate cancer. More than 80% of these patients develop bone metastases. The binding of enzalutamide to the AR prevents the nuclear translocation of the receptor, thus inactivating androgen signaling. However, prostate cancer cells eventually develop resistance to enzalutamide treatment. Studies have found resistance both in patients and in laboratory models. The mechanisms of and approaches to overcoming such resistance are significant issues that need to be addressed. In this review, we focus on the major mechanisms of acquired enzalutamide resistance, including genetic mutations and splice variants of the AR, signaling pathways that bypass androgen signaling, intratumoral androgen biosynthesis by prostate tumor cells, lineage plasticity, and contributions from the tumor microenvironment. Approaches for overcoming these mechanisms to enzalutamide resistance along with the associated problems and solutions are discussed. Emerging questions, concerns, and new opportunities in studying enzalutamide resistance will be addressed as well.
Collapse
Affiliation(s)
- Alexandra Vander Ark
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Jingchen Cao
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Xiaohong Li
- Program for Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| |
Collapse
|
42
|
Sun Y, Xu K, He M, Fan G, Lu H. Overexpression of Glypican 5 (GPC5) Inhibits Prostate Cancer Cell Proliferation and Invasion via Suppressing Sp1-Mediated EMT and Activation of Wnt/β-Catenin Signaling. Oncol Res 2018; 26:565-572. [PMID: 28893348 PMCID: PMC7844840 DOI: 10.3727/096504017x15044461944385] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Glypican 5 (GPC5) belongs to the family of heparan sulfate proteoglycans (HSPGs). It was initially known as a regulator of growth factors and morphogens. Recently, there have been reports on its correlation with the tumorigenic process in the development of some cancers. However, little is known about its precise role in prostate cancer (PCa). In the present study, we explored the expression pattern and biological functions of GPC5 in PCa cells. Our results showed that GPC5 was lowly expressed in PCa cell lines. Upregulation of GPC5 significantly inhibited PCa cell proliferation and invasion in vitro as well as attenuated tumor growth in vivo. We also found that overexpression of GPC5 inhibited the epithelial-mesenchymal transition (EMT) and Wnt/β-catenin signaling activation, which was mediated by Sp1. Taken together, we suggest GPC5 as a tumor suppressor in PCa and provide promising therapeutic strategies for PCa.
Collapse
Affiliation(s)
- Yu Sun
- Department of Pathology, General Hospital of Daqing Oil Field, Daqing, P.R. China
| | - Kai Xu
- Department of Pathology, General Hospital of Daqing Oil Field, Daqing, P.R. China
| | - Miao He
- Department of Pathology, General Hospital of Daqing Oil Field, Daqing, P.R. China
| | - Guilian Fan
- Department of Pathology, General Hospital of Daqing Oil Field, Daqing, P.R. China
| | - Hongming Lu
- Department of Pathology, General Hospital of Daqing Oil Field, Daqing, P.R. China
| |
Collapse
|
43
|
Zhou H, Wu G, Ma X, Xiao J, Yu G, Yang C, Xu N, Zhang B, Zhou J, Ye Z, Wang Z. Attenuation of TGFBR2 expression and tumour progression in prostate cancer involve diverse hypoxia-regulated pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:89. [PMID: 29699590 PMCID: PMC5921809 DOI: 10.1186/s13046-018-0764-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Background Dysregulation of transforming growth factor β (TGF-β) signaling and hypoxic microenvironment have respectively been reported to be involved in disease progression in malignancies of prostate. Emerging evidence indicates that downregulation of TGFBR2, a pivotal regulator of TGF-β signaling, may contribute to carcinogenesis and progression of prostate cancer (PCa). However, the biological function and regulatory mechanism of TGFBR2 in PCa remain poorly understood. In this study, we propose to investigate the crosstalk of hypoxia and TGF-β signaling and provide insight into the molecular mechanism underlying the regulatory pathways in PCa. Methods Prostate cancer cell lines were cultured in hypoxia or normoxia to evaluate the effect of hypoxia on TGFBR2 expression. Methylation specific polymerase chain reaction (MSP) and demethylation agents was used to evaluate the methylation regulation of TGFBR2 promoter. Besides, silencing of EZH2 via specific siRNAs or chemical inhibitor was used to validate the regulatory effect of EZH2 on TGFBR2. Moreover, we conducted PCR, western blot, and luciferase assays which studied the relationship of miR-93 and TGFBR2 in PCa cell lines and specimens. We also detected the impacts of hypoxia on EZH2 and miR-93, and further examined the tumorigenic functions of miR-93 on proliferation and epithelial-mesenchymal transition via a series of experiments. Results TGFBR2 expression was attenuated under hypoxia. Hypoxia-induced EZH2 promoted H3K27me3 which caused TGFBR2 promoter hypermethylation and contributed to its epigenetic silencing in PCa. Besides, miR-93 was significantly upregulated in PCa tissues and cell lines, and negatively correlated with the expression of TGFBR2. Ectopic expression of miR-93 promoted cell proliferation, migration and invasion in PCa, and its expression could also be induced by hypoxia. In addition, TGFBR2 was identified as a bona fide target of miR-93. Conclusions Our findings elucidate diverse hypoxia-regulated pathways including EZH2-mediated hypermethylation and miR-93-induced silencing contribute to attenuation of TGFBR2 expression and promote cancer progression in prostate cancer. Electronic supplementary material The online version of this article (10.1186/s13046-018-0764-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guanqing Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Urology, Aerospace Center Hospital(ASCH), Beijing, 100076, China
| | - Xueyou Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gan Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunguang Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nan Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bao Zhang
- Department of Urology, Aerospace Center Hospital(ASCH), Beijing, 100076, China
| | - Jun Zhou
- Department of Urology, The third people Hospital of Hubei Province, Wuhan, 430030, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
44
|
Phi LTH, Sari IN, Yang YG, Lee SH, Jun N, Kim KS, Lee YK, Kwon HY. Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem Cells Int 2018; 2018:5416923. [PMID: 29681949 PMCID: PMC5850899 DOI: 10.1155/2018/5416923] [Citation(s) in RCA: 611] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/11/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer stem cells (CSCs), also known as tumor-initiating cells (TICs), are suggested to be responsible for drug resistance and cancer relapse due in part to their ability to self-renew themselves and differentiate into heterogeneous lineages of cancer cells. Thus, it is important to understand the characteristics and mechanisms by which CSCs display resistance to therapeutic agents. In this review, we highlight the key features and mechanisms that regulate CSC function in drug resistance as well as recent breakthroughs of therapeutic approaches for targeting CSCs. This promises new insights of CSCs in drug resistance and provides better therapeutic rationales to accompany novel anticancer therapeutics.
Collapse
Affiliation(s)
- Lan Thi Hanh Phi
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Ita Novita Sari
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Ying-Gui Yang
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Sang-Hyun Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Nayoung Jun
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Kwang Seock Kim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Yun Kyung Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| | - Hyog Young Kwon
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Asan, Republic of Korea
| |
Collapse
|
45
|
Qin T, Barron L, Xia L, Huang H, Villarreal MM, Zwaagstra J, Collins C, Yang J, Zwieb C, Kodali R, Hinck CS, Kim SK, Reddick RL, Shu C, O'Connor-McCourt MD, Hinck AP, Sun LZ. A novel highly potent trivalent TGF-β receptor trap inhibits early-stage tumorigenesis and tumor cell invasion in murine Pten-deficient prostate glands. Oncotarget 2018; 7:86087-86102. [PMID: 27863384 PMCID: PMC5349899 DOI: 10.18632/oncotarget.13343] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022] Open
Abstract
The effects of transforming growth factor beta (TGF-β) signaling on prostate tumorigenesis has been shown to be strongly dependent on the stage of development, with TGF-β functioning as a tumor suppressor in early stages of disease and as a promoter in later stages. To study in further detail the paradoxical tumor-suppressive and tumor-promoting roles of the TGF-β pathway, we investigated the effect of systemic treatment with a TGF-β inhibitor on early stages of prostate tumorigenesis. To ensure effective inhibition, we developed and employed a novel trivalent TGF-β receptor trap, RER, comprised of domains derived from the TGF-β type II and type III receptors. This trap was shown to completely block TβRII binding, to antagonize TGF-β1 and TGF-β3 signaling in cultured epithelial cells at low picomolar concentrations, and it showed equal or better anti-TGF-β activities than a pan TGF-β neutralizing antibody and a TGF-β receptor I kinase inhibitor in various prostate cancer cell lines. Systemic administration of RER inhibited prostate tumor cell proliferation as indicated by reduced Ki67 positive cells and invasion potential of tumor cells in high grade prostatic intraepithelial neoplasia (PIN) lesions in the prostate glands of Pten conditional null mice. These results provide evidence that TGF-β acts as a promoter rather than a suppressor in the relatively early stages of this spontaneous prostate tumorigenesis model. Thus, inhibition of TGF-β signaling in early stages of prostate cancer may be a novel therapeutic strategy to inhibit the progression as well as the metastatic potential in patients with prostate cancer.
Collapse
Affiliation(s)
- Tai Qin
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Vascular Surgery, Second Xiangya Hospital and Xiangya School of Medicine, Central South University, Hunan, China
| | - Lindsey Barron
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lu Xia
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Gynecology and Obstetrics, Xiangya Hospital and Xiangya School of Medicine, Central South University, Hunan, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Maria M Villarreal
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - John Zwaagstra
- National Research Council Human Health Therapeutics Portfolio, Montréal, Quebec, Canada, Maureen O'Connor-McCourt is currently affiliated with Formation Biologics, Montréal, Quebec, Canada
| | - Cathy Collins
- National Research Council Human Health Therapeutics Portfolio, Montréal, Quebec, Canada, Maureen O'Connor-McCourt is currently affiliated with Formation Biologics, Montréal, Quebec, Canada
| | - Junhua Yang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Christian Zwieb
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ravindra Kodali
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Sun Kyung Kim
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Robert L Reddick
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chang Shu
- Department of Vascular Surgery, Second Xiangya Hospital and Xiangya School of Medicine, Central South University, Hunan, China
| | - Maureen D O'Connor-McCourt
- National Research Council Human Health Therapeutics Portfolio, Montréal, Quebec, Canada, Maureen O'Connor-McCourt is currently affiliated with Formation Biologics, Montréal, Quebec, Canada
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, Texas, USA
| |
Collapse
|
46
|
Davies AH, Beltran H, Zoubeidi A. Cellular plasticity and the neuroendocrine phenotype in prostate cancer. Nat Rev Urol 2018; 15:271-286. [PMID: 29460922 DOI: 10.1038/nrurol.2018.22] [Citation(s) in RCA: 291] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The success of next-generation androgen receptor (AR) pathway inhibitors, such as abiraterone acetate and enzalutamide, in treating prostate cancer has been hampered by the emergence of drug resistance. This acquired drug resistance is driven, in part, by the ability of prostate cancer cells to change their phenotype to adopt AR-independent pathways for growth and survival. Around one-quarter of resistant prostate tumours comprise cells that have undergone cellular reprogramming to become AR-independent and to acquire a continuum of neuroendocrine characteristics. These highly aggressive and lethal tumours, termed neuroendocrine prostate cancer (NEPC), exhibit reactivation of developmental programmes that are associated with epithelial-mesenchymal plasticity and acquisition of stem-like cell properties. In the past few years, our understanding of the link between lineage plasticity and an emergent NEPC phenotype has considerably increased. This new knowledge can contribute to novel therapeutic modalities that are likely to improve the treatment and clinical management of aggressive prostate cancer.
Collapse
Affiliation(s)
- Alastair H Davies
- Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, BC, Canada
| | - Himisha Beltran
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, 413 East 69th Street, New York, NY, USA
| | - Amina Zoubeidi
- Vancouver Prostate Centre, 2660 Oak Street, Vancouver, BC, Canada.,Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2775 Laurel Street, Vancouver, BC, Canada
| |
Collapse
|
47
|
Loss of TGF-β signaling in osteoblasts increases basic-FGF and promotes prostate cancer bone metastasis. Cancer Lett 2018; 418:109-118. [PMID: 29337106 DOI: 10.1016/j.canlet.2018.01.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 01/02/2023]
Abstract
TGF-β plays a central role in prostate cancer (PCa) bone metastasis, and it is crucial to understand the bone cell-specific role of TGF-β signaling in this process. Thus, we used knockout (KO) mouse models having deletion of the Tgfbr2 gene specifically in osteoblasts (Tgfbr2Col1CreERT KO) or in osteoclasts (Tgfbr2LysMCre KO). We found that PCa-induced bone lesion development was promoted in the Tgfbr2Col1CreERT KO mice, but was inhibited in the Tgfbr2LysMCre KO mice, relative to their respective control Tgfbr2FloxE2 littermates. Since metastatic PCa cells attach to osteoblasts when colonized in the bone microenvironment, we focused on the mechanistic studies using the Tgfbr2Col1CreERT KO mouse model. We found that bFGF was upregulated in osteoblasts from PC3-injected tibiae of Tgfbr2Col1CreERT KO mice and correlated with increased tumor cell proliferation, angiogenesis, amounts of cancer-associated fibroblasts and osteoclasts. In vitro studies showed that osteoblastogenesis was inhibited, osteoclastogenesis was stimulated, but PC3 viability was not affected, by bFGF treatments. Lastly, the increased PC3-induced bone lesions in Tgfbr2Col1CreERT KO mice were significantly attenuated by blocking bFGF using neutralizing antibody, suggesting bFGF is a promising target inhibiting bone metastasis.
Collapse
|
48
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
49
|
Receptor for advanced glycation end product blockade enhances the chemotherapeutic effect of cisplatin in tongue squamous cell carcinoma by reducing autophagy and modulating the Wnt pathway. Anticancer Drugs 2017; 28:187-196. [PMID: 27831944 DOI: 10.1097/cad.0000000000000451] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tongue squamous cell carcinoma (TSCC) is one of the most severe types of cancer with poor outcomes. Cisplatin is used widely to treat cancer cells, but many patients develop acquired drug resistance. The receptor for advanced glycation end products (RAGE) is expressed widely in TSCC and associated with drug-induced chemotherapy resistance. However, the effect of RAGE and cisplatin on Tca-8113 cells remains unknown. We assayed the combined use of RAGE blockade and cisplatin effect on Tca-8113 cells' viability by MTT and apoptosis rate of Tca-8113 cells on RAGE blockade+cisplatin treatment; cisplatin alone; or RAGE blockade alone by flow cytometry. We observed the expressions of autophagy-related proteins beclin1, LC3II, p62; Wnt signaling-related proteins β-catenin, GSK3β, WNT5A, ROR-2; and apoptosis-related protein cleaved caspase-3, bcl-2-associated X proteins using western blot. We determined WNT5A and beclin1 expression on Tca-8113 cells by immunofluorescence. We further observed autophagy vacuoles by monodansylcadaverine staining. We found that RAGE blockade and cisplatin significantly decreased cell viability and increased the cell apoptosis rate compared with cisplatin alone. Furthermore, RAGE blockade suppressed the canonical Wnt pathway proteins β-catenin and GSK-3β, but upregulated noncanonical WNT5A and receptor ROR-2. We show that RAGE blockade suppressed the levels of autophagy-related protein LC3II/I, beclin1, accelerated degradation of autophagy for the increasing p62 expression, and increased cell apoptosis for the increasing expressions of cleaved caspase-3 and bcl-2-associated X proteins. We observed the location of WNT5A and beclin1 expressions on cells by immunofluorescence and their trends were consistent with western blotting. Taken together, our findings suggested that RAGE blockade+cisplatin improved chemotherapeutic effects by reducing autophagy and regulating Wnt/β-catenin to suppress the progression of TSCC.
Collapse
|
50
|
Valkenburg KC, De Marzo AM, Williams BO. Deletion of tumor suppressors adenomatous polyposis coli and Smad4 in murine luminal epithelial cells causes invasive prostate cancer and loss of androgen receptor expression. Oncotarget 2017; 8:80265-80277. [PMID: 29113300 PMCID: PMC5655195 DOI: 10.18632/oncotarget.17919] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 05/03/2017] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer is the most diagnosed non-skin cancer in the US and kills approximately 27,000 men per year in the US. Additional genetic mouse models are needed that recapitulate the heterogeneous nature of human prostate cancer. The Wnt/beta-catenin signaling pathway is important for human prostate tumorigenesis and metastasis, and also drives tumorigenesis in mouse models. Loss of Smad4 has also been found in human prostate cancer and drives tumorigenesis and metastasis when coupled with other genetic aberrations in mouse models. In this work, we concurrently deleted Smad4 and the tumor suppressor and endogenous Wnt/beta-catenin inhibitor adenomatous polyposis coli (Apc) in luminal prostate cells in mice. This double conditional knockout model produced invasive castration-resistant prostate carcinoma with no evidence of metastasis. We observed mixed differentiation phenotypes, including basaloid and squamous differentiation. Interestingly, tumor cells in this model commonly lose androgen receptor expression. In addition, tumors disappear in these mice during androgen cycling (castration followed by testosterone reintroduction). These mice model non-metastatic castration resistant prostate cancer and should provide novel information for tumors that have genetic aberrations in the Wnt pathway or Smad4.
Collapse
Affiliation(s)
- Kenneth C. Valkenburg
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Bart O. Williams
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|