1
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2025; 33:295-315. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
2
|
Bialas P, Kobayashi T, Hellsten R, Krzyzanowska A, Persson M, Marginean F, Trudel D, Garraway IP, Trock BJ, Taimen P, Saad F, Mirtti T, Knudsen B, De Marzo AM, Bjartell A. pSTAT3 Expression is Increased in Advanced Prostate Cancer in Post-Initiation of Androgen Deprivation Therapy. Prostate 2025; 85:252-264. [PMID: 39523927 PMCID: PMC11720397 DOI: 10.1002/pros.24820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The transcription factor Signal Transducer and Activator of Transcription 3 (STAT3) plays a role in carcinogenesis and is involved in processes, such as proliferation, differentiation, drug resistance and immunosuppression. STAT3 can be activated by phosphorylation of tyrosine at position 705 (pSTAT3Tyr705) or serine at 727 (pSTAT3Ser727). High expression levels of pSTAT3 are implicated in advanced stages of prostate cancer (PCa) and are known to interact with the androgen receptor signaling pathway. However, not much is known about how androgen deprivation therapy (ADT) in advanced disease affects pSTAT3 expression. The aim of this study was to determine the influence of ADT on pSTAT3 expression in PCa tissue. METHODS The study cohort came from a PCa tissue microarray resource containing prostate specimens from patients before and post-initiation of ADT. Tissue samples from 111 patients were immunostained for pSTAT3Tyr705 and pSTAT3Ser727. H-score was used to evaluate the intensity and the percentage of pSTAT3 expression in malignant epithelial and stromal compartments. Univariate and multivariable Cox regression analyses were used to assess pSTAT3Tyr705 and pSTAT3Ser727 as biomarkers of oncological outcome in patients undergoing ADT. RESULTS Post-ADT PCa samples demonstrated increased nuclear and cytoplasmic levels of pSTAT3Ser727 in the stroma compared to pre-ADT samples, whereas pSTAT3Tyr705 expression was increased significantly in both stromal and malignant epithelial compartments except for stromal cytoplasm. High cytoplasmic pSTAT3Ser727 in stromal compartments correlated with reduced overall survival, shorter time to castration-resistant PCa development, and decreased metastasis-free survival. An increase in nuclear and cytoplasmic pSTAT3Ser727 expression within the stromal compartment of post-ADT samples corresponded to a shorter time to CRPC development, which was not observed for pSTAT3Tyr705. Multivariable survival analysis using Cox's regression identified that high cytoplasmic pSTAT3Ser727 expression in the stroma of post-ADT samples and pT3 or pT4-stage were associated with worse overall survival and 5-year metastasis-free survival (MFS). CONCLUSIONS This study presents novel insights into the impact of ADT on the expression levels of pSTAT3Tyr705 and pSTAT3Ser727 in PCa. Cytoplasmic pSTAT3Ser727 status of cancer-associated stromal cells in post-ADT samples may serve as an independent prognostic marker for OS and 5-year MFS, identifying prostate cancer patients prone to developing resistance to ADT.
Collapse
Affiliation(s)
- Piotr Bialas
- Department of Translational Medicine, Division of Urological CancersLund UniversityMalmöSweden
- Chair and Department of Cell BiologyPoznan University of Medical SciencesPoznanPoland
| | - Tamae Kobayashi
- Department of Translational Medicine, Division of Urological CancersLund UniversityMalmöSweden
| | - Rebecka Hellsten
- Department of Translational Medicine, Division of Urological CancersLund UniversityMalmöSweden
| | - Agnieszka Krzyzanowska
- Department of Translational Medicine, Division of Urological CancersLund UniversityMalmöSweden
| | - Margareta Persson
- Department of Laboratory Medicine, Translational Cancer ResearchLund UniversityLundSweden
| | - Felicia Marginean
- Department of Translational Medicine, Division of Urological CancersLund UniversityMalmöSweden
| | - Dominique Trudel
- Centre de recherche du Centre hospitalier de l'Université de Montréal et Institut du cancer de MontréalMontrealQuebecCanada
- Department of Pathology and Cellular BiologyUniversité de MontréalMontrealQuebecCanada
| | - Isla P. Garraway
- Department of Urology, Jonsson Comprehensive Cancer CenterDavid Geffen School of Medicine at University of CaliforniaLos AngelesCaliforniaUSA
- Division of UrologyGreater Los Angeles VA Healthcare SystemLos AngelesCaliforniaUSA
| | - Bruce J. Trock
- Department of Urology and Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Pekka Taimen
- Institute of Biomedicine and FICAN West Cancer CentreUniversity of TurkuTurkuFinland
- Department of PathologyTurku University HospitalTurkuFinland
| | - Fred Saad
- Department of SurgeryUniversité de MontréalMontrealQuebecCanada
| | - Tuomas Mirtti
- HUS Diagnostic Center, Department of PathologyHUS Helsinki University HospitalHelsinkiFinland
- Medicum and Research Program In Systems OncologyFaculty of Medicine, University of HelsinkiHelsinkiFinland
| | - Beatrice Knudsen
- Digital and Computational PathologyUniversity of UtahSalt Lake CityUtahUSA
| | - Angelo M. De Marzo
- Department of Urology and Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center at Johns HopkinsBaltimoreMarylandUSA
| | - Anders Bjartell
- Department of Translational Medicine, Division of Urological CancersLund UniversityMalmöSweden
- Department of UrologySkåne University HospitalMalmöSweden
| |
Collapse
|
3
|
Rahman R, Rahaman MH, Hanson AR, Choo N, Xie J, Townley SL, Shrestha R, Hassankhani R, Islam S, Ramm S, Simpson KJ, Risbridger GP, Best G, Centenera MM, Balk SP, Kichenadasse G, Taylor RA, Butler LM, Tilley WD, Conn SJ, Lawrence MG, Wang S, Selth LA. CDK9 inhibition inhibits multiple oncogenic transcriptional and epigenetic pathways in prostate cancer. Br J Cancer 2024; 131:1092-1105. [PMID: 39117800 PMCID: PMC11405875 DOI: 10.1038/s41416-024-02810-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Cyclin-dependent kinase 9 (CDK9) stimulates oncogenic transcriptional pathways in cancer and CDK9 inhibitors have emerged as promising therapeutic candidates. METHODS The activity of an orally bioavailable CDK9 inhibitor, CDKI-73, was evaluated in prostate cancer cell lines, a xenograft mouse model, and patient-derived tumor explants and organoids. Expression of CDK9 was evaluated in clinical specimens by mining public datasets and immunohistochemistry. Effects of CDKI-73 on prostate cancer cells were determined by cell-based assays, molecular profiling and transcriptomic/epigenomic approaches. RESULTS CDKI-73 inhibited proliferation and enhanced cell death in diverse in vitro and in vivo models of androgen receptor (AR)-driven and AR-independent models. Mechanistically, CDKI-73-mediated inhibition of RNA polymerase II serine 2 phosphorylation resulted in reduced expression of BCL-2 anti-apoptotic factors and transcriptional defects. Transcriptomic and epigenomic approaches revealed that CDKI-73 suppressed signaling pathways regulated by AR, MYC, and BRD4, key drivers of dysregulated transcription in prostate cancer, and reprogrammed cancer-associated super-enhancers. These latter findings prompted the evaluation of CDKI-73 with the BRD4 inhibitor AZD5153, a combination that was synergistic in patient-derived organoids and in vivo. CONCLUSION Our work demonstrates that CDK9 inhibition disrupts multiple oncogenic pathways and positions CDKI-73 as a promising therapeutic agent for prostate cancer, particularly aggressive, therapy-resistant subtypes.
Collapse
Affiliation(s)
- Razia Rahman
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
| | - Muhammed H Rahaman
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Adrienne R Hanson
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
| | - Nicholas Choo
- Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Jianling Xie
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
| | - Scott L Townley
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
| | - Raj Shrestha
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
- Flinders University, Freemasons Centre for Male Health and Wellbeing, Bedford Park, SA, Australia
| | - Ramin Hassankhani
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Saiful Islam
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Susanne Ramm
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, Australia
| | - Gail P Risbridger
- Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, Melbourne, VIC, Australia
- Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Giles Best
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
| | - Margaret M Centenera
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Steven P Balk
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ganessan Kichenadasse
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
- Department of Medical Oncology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, SA, South Australia
| | - Renea A Taylor
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, Melbourne, VIC, Australia
- Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
- Biomedicine Discovery Institute Cancer Program, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Lisa M Butler
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Wayne D Tilley
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Simon J Conn
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia
| | - Mitchell G Lawrence
- Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Cabrini Institute, Cabrini Health, Malvern, Melbourne, VIC, Australia
- Melbourne Urological Research Alliance (MURAL), Monash Biomedicine Discovery Institute Cancer Program, Monash University, Clayton, VIC, Australia
| | - Shudong Wang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Luke A Selth
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, Australia.
- Flinders University, Freemasons Centre for Male Health and Wellbeing, Bedford Park, SA, Australia.
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
4
|
Mavingire N, Moore JC, Johnson JR, Dwead AM, Cropp CD, Mechref Y, Kobeissy F, Rais-Bahrami S, Woods-Burnham L. Revisiting HER2 in Prostate Cancer from an Inclusive Perspective: From Biomarkers to Omics. Cancers (Basel) 2024; 16:3262. [PMID: 39409883 PMCID: PMC11476348 DOI: 10.3390/cancers16193262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a major driver of disease progression, treatment resistance, and worse survival for patients with various types of cancers, including prostate cancer. However, key bench studies and clinical trials have failed to evaluate the role of HER2 in prostate cancer using racially diverse experimental designs and protocols. This lack of diversity represents what has been the status quo of cancer research in the United States for decades. In the case of prostate cancer, homogenic study designs are problematic as Black men are much more likely to be diagnosed and die from aggressive and incurable forms of the disease. Therefore, the strategic inclusion of biospecimens collected from Black patients as well as the recruitment and enrollment of Black men into prostate cancer clinical trials is necessary to comprehensively evaluate genetic and molecular factors that contribute to variable outcomes in this high-risk population. Additionally, a higher prevalence of HER2 expression in Black men was recently reported in a small cohort of prostate cancer patients and may contribute to worsened prognosis. In this review, we carefully consider the role of HER2 in prostate cancer while, for the first time, taking into account the influences of race and genetic ancestry.
Collapse
Affiliation(s)
- Nicole Mavingire
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.); (J.C.M.)
| | - Janelle C. Moore
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.); (J.C.M.)
| | - Jabril R. Johnson
- Department of Microbiology, Biochemistry, & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Abdulrahman M. Dwead
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.); (J.C.M.)
| | - Cheryl D. Cropp
- Department of Pharmacology & Toxicology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35294, USA;
- Department of Radiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL 35294, USA
| | - Leanne Woods-Burnham
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA 30310, USA; (N.M.); (J.C.M.)
| |
Collapse
|
5
|
Thakur N, Singh P, Bagri A, Srivastava S, Dwivedi V, Singh A, Jaiswal SK, Dholpuria S. Therapy resistance in prostate cancer: mechanism, signaling and reversal strategies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1110-1134. [PMID: 39351434 PMCID: PMC11438573 DOI: 10.37349/etat.2024.00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate cancer (PC) depicts a major health challenge all over the globe due to its complexities in the treatment and diverse clinical trajectories. Even in the advances in the modern treatment strategies, the spectrum of resistance to the therapies continues to be a significant challenge. This review comprehensively examines the underlying mechanisms of the therapy resistance occurred in PC, focusing on both the tumor microenvironment and the signaling pathways implicated in the resistance. Tumor microenvironment comprises of stromal and epithelial cells, which influences tumor growth, response to therapy and progression. Mechanisms such as microenvironmental epithelial-mesenchymal transition (EMT), anoikis suppression and stimulation of angiogenesis results in therapy resistance. Moreover, dysregulation of signaling pathways including androgen receptor (AR), mammalian target of rapamycin/phosphoinositide 3 kinase/AKT (mTOR/PI3K/AKT), DNA damage repair and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways drive therapy resistance by promoting tumor survival and proliferation. Understanding these molecular pathways is important for developing targeted therapeutic interventions which overcomes resistance. In conclusion, a complete grasp of mechanisms and pathways underlying medication resistance in PC is important for the development of individualized treatment plans and enhancements of clinical outcomes. By studying and understanding the complex mechanisms of signaling pathways and microenvironmental factors contributing to therapy resistance, this study focuses and aims to guide the development of innovative therapeutic approaches to effectively overcome the PC progression and improve the survival rate of patients.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Aditi Bagri
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Saumya Srivastava
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Vinay Dwivedi
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Asha Singh
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Sunil Kumar Jaiswal
- School of Biological and Life Sciences, Galgotias University, Greater Noida, Uttar Pradesh 203201, India
| | - Sunny Dholpuria
- Department of Life Sciences, J. C. Bose University of Science and Technology, YMCA Faridabad, Faridabad, Haryana 121006, India
| |
Collapse
|
6
|
Mishra J, Chakraborty S, Nandi P, Manna S, Baral T, Niharika, Roy A, Mishra P, Patra SK. Epigenetic regulation of androgen dependent and independent prostate cancer. Adv Cancer Res 2024; 161:223-320. [PMID: 39032951 DOI: 10.1016/bs.acr.2024.05.007] [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] [Indexed: 07/23/2024]
Abstract
Prostate cancer is one of the most common malignancies among men worldwide. Besides genetic alterations, epigenetic modulations including DNA methylation, histone modifications and miRNA mediated alteration of gene expression are the key driving forces for the prostate tumor development and cancer progression. Aberrant expression and/or the activity of the epigenetic modifiers/enzymes, results in aberrant expression of genes involved in DNA repair, cell cycle regulation, cell adhesion, apoptosis, autophagy, tumor suppression and hormone response and thereby disease progression. Altered epigenome is associated with prostate cancer recurrence, progression, aggressiveness and transition from androgen-dependent to androgen-independent phenotype. These epigenetic modifications are reversible and various compounds/drugs targeting the epigenetic enzymes have been developed that are effective in cancer treatment. This chapter focuses on the epigenetic alterations in prostate cancer initiation and progression, listing different epigenetic biomarkers for diagnosis and prognosis of the disease and their potential as therapeutic targets. This chapter also summarizes different epigenetic drugs approved for prostate cancer therapy and the drugs available for clinical trials.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Prahallad Mishra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India.
| |
Collapse
|
7
|
Echeverría CE, Oyarzún VI, López-Cortés A, Cancino J, Sotomayor PC, Goncalves MD, Godoy AS. Biological role of fructose in the male reproductive system: Potential implications for prostate cancer. Prostate 2024; 84:8-24. [PMID: 37888416 PMCID: PMC10872645 DOI: 10.1002/pros.24631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Over the last 20 years, fructose has gradually emerged as a potential metabolic substrate capable of promoting the growth and progression of various cancers, including prostate cancer (PCa). The biological and molecular mechanisms that underlie the effects of fructose on cancer are beginning to be elucidated. METHODS This review summarizes the biological function of fructose as a potential carbon source for PCa cells and its role in the functionality of the male reproductive tract under normal conditions. RESULTS The most recent biological advances related to fructose transport and metabolism as well as their implications in PCa growth and progression suggest that fructose represent a potential carbon source for PCa cells. Consequently, fructose derivatives may represent efficient radiotracers for obtaining PCa images via positron emission tomography and fructose transporters/fructose-metabolizing enzymes could be utilized as potential diagnostic and/or predictive biomarkers for PCa. CONCLUSION The existing data suggest that restriction of fructose from the diet could be a useful therapeutic strategy for patients with PCa.
Collapse
Affiliation(s)
- Carolina E. Echeverría
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical, New York, NY, USA
| | - Vanessa I. Oyarzún
- Laboratory of Ocular and Systemic Autoimmune Diseases, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Jorge Cancino
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Paula C. Sotomayor
- Departamento de Urología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcus D. Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medical, New York, NY, USA
| | - Alejandro S. Godoy
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo New York, USA
| |
Collapse
|
8
|
Zhang C, Sun Q, Zhao J, Jiang N, Hao Y, Luo J, Karim S, Wu L, de Perrot M, Peng C, Zhao X. JSI-124 inhibits cell proliferation and tumor growth by inducing autophagy and apoptosis in murine malignant mesothelioma. Mol Carcinog 2023; 62:1888-1901. [PMID: 37642305 DOI: 10.1002/mc.23623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023]
Abstract
Malignant pleural mesothelioma (MPM), mainly caused by asbestos exposure, has a poor prognosis and lacks effective treatment compared with other cancer types. The intracellular transcription factor signal transducer and activator of transcription 3 (STAT3) is overexpressed and hyperactivated in most human cancers. In this study, the role of STAT3 in murine MPM was examined. Inhibition of the Janus kinase 2 (JAK2)/STAT3 pathway with the selective inhibitor JSI-124 has an antitumor effect in murine MPM. Specifically, we demonstrated that JSI-124 inhibits murine MPM cell growth and induces apoptotic and autophagic cell death. Exposure of RN5 and AB12 cells to JSI-124 resulted in apoptosis via the Bcl-2 family of proteins. JSI-124 triggered autophagosome formation, accumulation, and conversion of LC3I to LC3II. Autophagy inhibitors, Chloroquine (CQ) and Bafilomycin A1 (Baf-A1), inhibited autophagy and sensitized RN5 and AB12 cells to JSI-124-induced apoptosis. Our data indicate that JSI-124 is a promising therapeutic agent for MPM treatment.
Collapse
Affiliation(s)
- Chengke Zhang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Qifeng Sun
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Jiangfeng Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Ning Jiang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yingtao Hao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Junwen Luo
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Saraf Karim
- Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Licun Wu
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Chuanliang Peng
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- Key Laboratory of Thoracic Cancer, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
9
|
Molenda S, Sikorska A, Florczak A, Lorenc P, Dams-Kozlowska H. Oligonucleotide-Based Therapeutics for STAT3 Targeting in Cancer-Drug Carriers Matter. Cancers (Basel) 2023; 15:5647. [PMID: 38067351 PMCID: PMC10705165 DOI: 10.3390/cancers15235647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 09/08/2024] Open
Abstract
High expression and phosphorylation of signal transducer and transcription activator 3 (STAT3) are correlated with progression and poor prognosis in various types of cancer. The constitutive activation of STAT3 in cancer affects processes such as cell proliferation, apoptosis, metastasis, angiogenesis, and drug resistance. The importance of STAT3 in cancer makes it a potential therapeutic target. Various methods of directly and indirectly blocking STAT3 activity at different steps of the STAT3 pathway have been investigated. However, the outcome has been limited, mainly by the number of upstream proteins that can reactivate STAT3 or the relatively low specificity of the inhibitors. A new branch of molecules with significant therapeutic potential has emerged thanks to recent developments in the regulatory function of non-coding nucleic acids. Oligonucleotide-based therapeutics can silence target transcripts or edit genes, leading to the modification of gene expression profiles, causing cell death or restoring cell function. Moreover, they can reach untreatable targets, such as transcription factors. This review briefly describes oligonucleotide-based therapeutics that found application to target STAT3 activity in cancer. Additionally, this review comprehensively summarizes how the inhibition of STAT3 activity by nucleic acid-based therapeutics such as siRNA, shRNA, ASO, and ODN-decoy affected the therapy of different types of cancer in preclinical and clinical studies. Moreover, due to some limitations of oligonucleotide-based therapeutics, the importance of carriers that can deliver nucleic acid molecules to affect the STAT3 in cancer cells and cells of the tumor microenvironment (TME) was pointed out. Combining a high specificity of oligonucleotide-based therapeutics toward their targets and functionalized nanoparticles toward cell type can generate very efficient formulations.
Collapse
Affiliation(s)
- Sara Molenda
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Agata Sikorska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Anna Florczak
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Patryk Lorenc
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Hanna Dams-Kozlowska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| |
Collapse
|
10
|
Fernandes R, Costa C, Fernandes R, Barros AN. Inflammation in Prostate Cancer: Exploring the Promising Role of Phenolic Compounds as an Innovative Therapeutic Approach. Biomedicines 2023; 11:3140. [PMID: 38137361 PMCID: PMC10740737 DOI: 10.3390/biomedicines11123140] [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: 10/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Prostate cancer (PCa) remains a significant global health concern, being a major cause of cancer morbidity and mortality worldwide. Furthermore, profound understanding of the disease is needed. Prostate inflammation caused by external or genetic factors is a central player in prostate carcinogenesis. However, the mechanisms underlying inflammation-driven PCa remain poorly understood. This review dissects the diagnosis methods for PCa and the pathophysiological mechanisms underlying the disease, clarifying the dynamic interplay between inflammation and leukocytes in promoting tumour development and spread. It provides updates on recent advances in elucidating and treating prostate carcinogenesis, and opens new insights for the use of bioactive compounds in PCa. Polyphenols, with their noteworthy antioxidant and anti-inflammatory properties, along with their synergistic potential when combined with conventional treatments, offer promising prospects for innovative therapeutic strategies. Evidence from the use of polyphenols and polyphenol-based nanoparticles in PCa revealed their positive effects in controlling tumour growth, proliferation, and metastasis. By consolidating the diverse features of PCa research, this review aims to contribute to increased understanding of the disease and stimulate further research into the role of polyphenols and polyphenol-based nanoparticles in its management.
Collapse
Affiliation(s)
- Raquel Fernandes
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Cátia Costa
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Rúben Fernandes
- FP-I3ID, Instituto de Investigação, Inovação e Desenvolvimento, FP-BHS, Biomedical and Health Sciences, Universidade Fernando Pessoa, 4249-004 Porto, Portugal;
- CECLIN, Centro de Estudos Clínicos, Hospital Fernando Pessoa, 4420-096 Gondomar, Portugal
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Novo Barros
- Centre for Research and Technology of Agro-Environmental and Biological Sciences, CITAB, Inov4Agro, University of Trás-os-Montes and Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal;
| |
Collapse
|
11
|
Jameel M, Fatma H, Nadtochii LA, Siddique HR. Molecular Insight into Prostate Cancer: Preventive Role of Selective Bioactive Molecules. Life (Basel) 2023; 13:1976. [PMID: 37895357 PMCID: PMC10608662 DOI: 10.3390/life13101976] [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: 08/21/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Prostate cancer (CaP) is one of the most prevalent male malignancies, accounting for a considerable number of annual mortalities. However, the prompt identification of early-stage CaP often faces delays due to diverse factors, including socioeconomic inequalities. The androgen receptor (AR), in conjunction with various other signaling pathways, exerts a central influence on the genesis, progression, and metastasis of CaP, with androgen deprivation therapy (ADT) serving as the primary therapeutic strategy. Therapeutic modalities encompassing surgery, chemotherapy, hormonal intervention, and radiotherapy have been formulated for addressing early and metastatic CaP. Nonetheless, the heterogeneous tumor microenvironment frequently triggers the activation of signaling pathways, culminating in the emergence of chemoresistance, an aspect to which cancer stem cells (CSCs) notably contribute. Phytochemicals emerge as reservoirs of bioactive agents conferring manifold advantages against human morbidity. Several of these phytochemicals demonstrate potential chemoprotective and chemosensitizing properties against CaP, with selectivity exhibited towards malignant cells while sparing their normal counterparts. In this context, the present review aims to elucidate the intricate molecular underpinnings associated with metastatic CaP development and the acquisition of chemoresistance. Moreover, the contributions of phytochemicals to ameliorating CaP initiation, progression, and chemoresistance are also discussed.
Collapse
Affiliation(s)
- Mohd Jameel
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| | - Homa Fatma
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| | - Liudmila A. Nadtochii
- Department of Microbiology, Saint Petersburg State Chemical & Pharmaceutical University, 197022 Saint Petersburg, Russia
| | - Hifzur R. Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| |
Collapse
|
12
|
Antropova EA, Khlebodarova TM, Demenkov PS, Volianskaia AR, Venzel AS, Ivanisenko NV, Gavrilenko AD, Ivanisenko TV, Adamovskaya AV, Revva PM, Kolchanov NA, Lavrik IN, Ivanisenko VA. Reconstruction of the regulatory hypermethylation network controlling hepatocellular carcinoma development during hepatitis C viral infection. J Integr Bioinform 2023; 20:jib-2023-0013. [PMID: 37978846 PMCID: PMC10757076 DOI: 10.1515/jib-2023-0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/31/2023] [Indexed: 11/19/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has been associated with hepatitis C viral (HCV) infection as a potential risk factor. Nonetheless, the precise genetic regulatory mechanisms triggered by the virus, leading to virus-induced hepatocarcinogenesis, remain unclear. We hypothesized that HCV proteins might modulate the activity of aberrantly methylated HCC genes through regulatory pathways. Virus-host regulatory pathways, interactions between proteins, gene expression, transport, and stability regulation, were reconstructed using the ANDSystem. Gene expression regulation was statistically significant. Gene network analysis identified four out of 70 HCC marker genes whose expression regulation by viral proteins may be associated with HCC: DNA-binding protein inhibitor ID - 1 (ID1), flap endonuclease 1 (FEN1), cyclin-dependent kinase inhibitor 2A (CDKN2A), and telomerase reverse transcriptase (TERT). It suggested the following viral protein effects in HCV/human protein heterocomplexes: HCV NS3(p70) protein activates human STAT3 and NOTC1; NS2-3(p23), NS5B(p68), NS1(E2), and core(p21) activate SETD2; NS5A inhibits SMYD3; and NS3 inhibits CCN2. Interestingly, NS3 and E1(gp32) activate c-Jun when it positively regulates CDKN2A and inhibit it when it represses TERT. The discovered regulatory mechanisms might be key areas of focus for creating medications and preventative therapies to decrease the likelihood of HCC development during HCV infection.
Collapse
Affiliation(s)
| | - Tamara M. Khlebodarova
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Pavel S. Demenkov
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | | | - Artur S. Venzel
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nikita V. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexandr D. Gavrilenko
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Timofey V. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Anna V. Adamovskaya
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Polina M. Revva
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106Magdeburg, Germany
| | - Vladimir A. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of RAS, Novosibirsk, Russia
- Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
13
|
Ota H, Sato H, Mizumoto S, Wakai K, Yoneda K, Yamamoto K, Nakanishi H, Ikeda JI, Sakamoto S, Ichikawa T, Yamada S, Takahashi S, Ikehara Y, Nishihara S. Switching mechanism from AR to EGFR signaling via 3-O-sulfated heparan sulfate in castration-resistant prostate cancer. Sci Rep 2023; 13:11618. [PMID: 37463954 DOI: 10.1038/s41598-023-38746-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/13/2023] [Indexed: 07/20/2023] Open
Abstract
Androgen deprivation therapy is given to suppress prostate cancer growth; however, some cells continue to grow hormone-independently as castration-resistant prostate cancer (CRPC). Sulfated glycosaminoglycans promote ligand binding to receptors as co-receptors, but their role in CRPC remains unknown. Using the human prostate cancer cell line C4-2, which can proliferate in hormone-dependent and hormone-independent conditions, we found that epidermal growth factor (EGF)-activated EGFR-ERK1/2 signaling via 3-O-sulfated heparan sulfate (HS) produced by HS 3-O-sulfotransferase 1 (HS3ST1) is activated in C4-2 cells under hormone depletion. Knockdown of HS3ST1 in C4-2 cells suppressed hormone-independent growth, and inhibited both EGF binding to the cell surface and activation of EGFR-ERK1/2 signaling. Gefitinib, an EGFR inhibitor, significantly suppressed C4-2 cell proliferation and growth of a xenografted C4-2 tumor in castrated mouse. Collectively, our study has revealed a mechanism by which cancer cells switch to hormone-independent growth and identified the key regulator as 3-O-sulfated HS.
Collapse
Affiliation(s)
- Hayato Ota
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo, Japan
| | - Hirokazu Sato
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Ken Wakai
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kei Yoneda
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuo Yamamoto
- Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hayao Nakanishi
- Laboratory of Pathology and Clinical Research, Aichi Cancer Center Aichi Hospital, Nagoya, Aichi, Japan
| | - Jun-Ichiro Ikeda
- Department of Diagnostic Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shinichi Sakamoto
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Yuzuru Ikehara
- Department of Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shoko Nishihara
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Tokyo, Japan.
- Glycan & Life System Integration Center (GaLSIC), Soka University, Tokyo, Japan.
| |
Collapse
|
14
|
Steiner I, Flores-Tellez TDNJ, Mevel R, Ali A, Wang P, Schofield P, Behan C, Forsythe N, Ashton G, Taylor C, Mills IG, Oliveira P, McDade SS, Zaiss DM, Choudhury A, Lacaud G, Baena E. Autocrine activation of MAPK signaling mediates intrinsic tolerance to androgen deprivation in LY6D prostate cancer cells. Cell Rep 2023; 42:112377. [PMID: 37060563 DOI: 10.1016/j.celrep.2023.112377] [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: 11/05/2021] [Revised: 12/12/2022] [Accepted: 03/23/2023] [Indexed: 04/16/2023] Open
Abstract
The emergence of castration-resistant prostate cancer remains an area of unmet clinical need. We recently identified a subpopulation of normal prostate progenitor cells, characterized by an intrinsic resistance to androgen deprivation and expression of LY6D. We here demonstrate that conditional deletion of PTEN in the murine prostate epithelium causes an expansion of transformed LY6D+ progenitor cells without impairing stem cell properties. Transcriptomic analyses of LY6D+ luminal cells identified an autocrine positive feedback loop, based on the secretion of amphiregulin (AREG)-mediated activation of mitogen-activated protein kinase (MAPK) signaling, increasing cellular fitness and organoid formation. Pharmacological interference with this pathway overcomes the castration-resistant properties of LY6D+ cells with a suppression of organoid formation and loss of LY6D+ cells in vivo. Notably, LY6D+ tumor cells are enriched in high-grade and androgen-resistant prostate cancer, providing clinical evidence for their contribution to advanced disease. Our data indicate that early interference with MAPK inhibitors can prevent progression of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Ivana Steiner
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Teresita Del N J Flores-Tellez
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Renaud Mevel
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Amin Ali
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Pengbo Wang
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Pieta Schofield
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Caron Behan
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Nicholas Forsythe
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL Northern Ireland, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Garry Ashton
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Catherine Taylor
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, M20 4BX Manchester, UK
| | - Ian G Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL Northern Ireland, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK; Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, OX3 9DU Oxford, UK; Department of Clinical Sciences and Centre for Cancer Biomarkers, University of Bergen, 7804 Bergen, Norway
| | - Pedro Oliveira
- Department of Pathology, The Christie NHS Foundation Trust, M20 4BX Manchester, UK
| | - Simon S McDade
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL Northern Ireland, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Dietmar M Zaiss
- Department of Immune Medicine, University Regensburg, Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, and Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany
| | - Ananya Choudhury
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, M20 4BX Manchester, UK; The University of Manchester, Manchester Cancer Research Centre, M20 4BX Manchester, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK
| | - Esther Baena
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG Macclesfield, UK.
| |
Collapse
|
15
|
Wu W, Tan K, Xu W, Liu J, Lv Z. Pathogenicity of p.Phe147del in RET in familial Hirschsprung's disease. Am J Transl Res 2023; 15:2690-2702. [PMID: 37193168 PMCID: PMC10182518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/15/2023] [Indexed: 05/18/2023]
Abstract
OBJECTIVE The study aimed to explore the pathogenicity of RET p.Phe147del in a Hirschsprung'irschspru (HSCR) family and facilitate the deeper understanding of HSCR families. METHODS Whole-exome sequencing (WES) was performed to decipher a HSCR family. We used a "GlycoEP" tool to analyze RET protein glycosylation. A series of molecular biological approaches including mutated plasmid construction, cell transfection, polymerase chain reaction, immunofluorescence and immunoblotting were introduced to determine the mutation status and altered expression of RET as well as its related genes or proteins. MG132 was applied to analyze the mechanism of mutated RET. RESULTS WES and Sanger results revealed that p.Phe147del in-frame mutation (IM) was a potential pathogenetic factor for familial HSCR. Moreover, the IM led to disrupted N-glycosylation of RET accompanied with protein structural change, resulting in the decreased transcriptional and protein level of RET, CCND1, VEGF and BCL2, and the decreased protein level of phosphorylated ERK and STAT3. Further studies revealed that the IM-evoked RET decline was reversed by inhibiting proteosome in a dose dependent manner, thus suggesting that the decrease in intracellular RET protein levels interrupted the transportation of RET protein from the cytoplasm to the cell surface. CONCLUSION The newly found p.Phe147del IM of RET is pathogenic to familial HSCR and it disrupts RET structure and abundance via the proteasome pathway, representing evidence for the early prevention, clinical diagnosis and treatment of HSCR.
Collapse
Affiliation(s)
- Wei Wu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Kezhe Tan
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Weijue Xu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Jiangbin Liu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University Shanghai, China
| | - Zhibao Lv
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
16
|
Verma P, Shukla N, Kumari S, Ansari M, Gautam NK, Patel GK. Cancer stem cell in prostate cancer progression, metastasis and therapy resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188887. [PMID: 36997008 DOI: 10.1016/j.bbcan.2023.188887] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/18/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023]
Abstract
Prostate cancer (PCa) is the most diagnosed malignancy in the men worldwide. Cancer stem cells (CSCs) are the sub-population of cells present in the tumor which possess unique properties of self-renewal and multilineage differentiation thus thought to be major cause of therapy resistance, disease relapse, and mortality in several malignancies including PCa. CSCs have also been shown positive for the common stem cells markers such as ALDH EZH2, OCT4, SOX2, c-MYC, Nanog etc. Therefore, isolation and characterization of CSCs specific markers which may discriminate CSCs and normal stem cells are critical to selectively eliminate CSCs. Rapid advances in the field offers a theoretical explanation for many of the enduring uncertainties encompassing the etiology and an optimism for the identification of new stem-cell targets, development of reliable and efficient therapies in the future. The emerging reports have also provided unprecedented insights into CSCs plasticity, quiescence, renewal, and therapeutic response. In this review, we discuss the identification of PCa stem cells, their unique properties, stemness-driving pathways, new diagnostics, and therapeutic interventions.
Collapse
|
17
|
Ji Y, Zhang R, Han X, Zhou J. Targeting the N-terminal domain of the androgen receptor: The effective approach in therapy of CRPC. Eur J Med Chem 2023; 247:115077. [PMID: 36587421 DOI: 10.1016/j.ejmech.2022.115077] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022]
Abstract
The androgen receptor (AR) is dominant in prostate cancer (PCa) pathology. Current therapeutic agents for advanced PCa include androgen synthesis inhibitors and AR antagonists that bind to the hormone binding pocket (HBP) at the ligand binding domain (LBD). However, AR amplification, AR splice variants (AR-Vs) expression, and intra-tumoral de novo synthesis of androgens result in the reactivation of AR signalling. The AR N-terminal domain (NTD) plays an essential role in AR transcriptional activity. The AR inhibitor targeting NTD could potentially block the activation of both full-length AR and AR-Vs, thus overcoming major resistance mechanisms to current treatments. This review discusses the progress of research in various NTD inhibitors and provides new insight into the development of AR-NTD inhibitors.
Collapse
Affiliation(s)
- Yang Ji
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Rongyu Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Xiaoli Han
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China.
| |
Collapse
|
18
|
Yang F, Bian Z, Xu P, Sun S, Huang Z. MicroRNA-204-5p: A pivotal tumor suppressor. Cancer Med 2023; 12:3185-3200. [PMID: 35908280 PMCID: PMC9939231 DOI: 10.1002/cam4.5077] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/08/2022] [Accepted: 07/03/2022] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of non-coding single-stranded RNA molecules with a length of approximately 18-25 nt nucleotides that regulate gene expression post-transcriptionally. MiR-204-5p originates from the sixth intron of the transient receptor potential cation channel subfamily M member 3 (TRPM3) gene. MiR-204-5p is frequently downregulated in various cancer types and is related to the clinicopathological characteristics and prognosis of cancer patients. So far, many studies have determined that miR-204-5p functions as a tumor suppressor for its extensive and powerful capacity to inhibit tumor proliferation, metastasis, autophagy, and chemoresistance in multiple cancer types. MiR-204-5p appears to be a promising prognostic biomarker and a therapeutic target for human cancers. This review summarized the latest advances on the role of miR-204-5p in human cancers.
Collapse
Affiliation(s)
- Fan Yang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Zehua Bian
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Peiwen Xu
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Shengbai Sun
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Zhaohui Huang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsuChina
- Laboratory of Cancer Epigenetics, Wuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| |
Collapse
|
19
|
Inflammation and Prostate Cancer: Pathological Analysis from Pros-IT CNR 2. Cancers (Basel) 2023; 15:cancers15030630. [PMID: 36765589 PMCID: PMC9913270 DOI: 10.3390/cancers15030630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Extensive research effort has been devoted to investigating the link between inflammation and PCa. However, this relationship remains unclear and controversial. The aim of our multi-center study was to investigate this association by histologically evaluating the distribution of PI and PCA in prostate biopsy cores from patients of eight referral centers in Italy. RESULTS We evaluated 2220 cores from 197 patients; all the frustules were re-evaluated by dedicated pathologists retrospectively. Pathologists assigned IRANI scores and determined the positions of PIs; pathologists also re-evaluated the presence of PCa and relative ISUP grade. PCa was recorded in 749/2220 (33.7%). We divided this sample into a PCa PI group (634/749 cores [84.7%]) and a non-PCa + PI group (1157/1471 cores [78.7%]). We observed a statistically significant difference in the presence of inflammation among cores with cancer (p < 0.01). Moreover, periglandular inflammation was higher in the cores with neoplasia, while stromal inflammation was higher in cores without neoplasia (38.5% vs. 31.1% and 55.4% vs. 63.5% p < 0.01). CONCLUSIONS In our experience, there is evidence of an association between PI and PCa at a tissue level. Further studies are needed to confirm our findings and to identify patients who might benefit from target therapies to prevent PCa occurrence and/or progression.
Collapse
|
20
|
Sanmukh SG, Dos Santos NJ, Nascimento Barquilha C, De Carvalho M, Pintor Dos Reis P, Delella FK, Carvalho HF, Latek D, Fehér T, Felisbino SL. Bacterial RNA virus MS2 exposure increases the expression of cancer progression genes in the LNCaP prostate cancer cell line. Oncol Lett 2023; 25:86. [PMID: 36760518 PMCID: PMC9878357 DOI: 10.3892/ol.2023.13672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/02/2022] [Indexed: 01/19/2023] Open
Abstract
Bacteriophages effectively counteract diverse bacterial infections, and their ability to treat most types of cancer has been explored using phage engineering or phage-virus hybrid platforms. In the present study, it was demonstrated that the bacteriophage MS2 can affect the expression of genes associated with the proliferation and survival of LNCaP prostate epithelial cells. LNCaP cells were exposed to bacteriophage MS2 at a concentration of 1×107 plaque forming units/ml for 24-48 h. After exposure, various cellular parameters, including cell viability, morphology, and changes in gene expression, were examined. MS2 affected cell viability adversely, reducing viability by 25% in the first 4 h of treatment; however, cell viability recovered within 24-48 h. Similarly, the AKT, androgen receptor, integrin α5, integrin β1, MAPK1, MAPK3, STAT3, and peroxisome proliferator-activated receptor-γ coactivator 1α genes, which are involved in various normal cellular processes and tumor progression, were significantly upregulated, whereas the expression levels of HSP90, ITGB5, ITGB3, HSP27, ITGAV, and PI3K genes were unchanged. Therefore, based on viability and gene expression changes, bacteriophage MS2 severely impaired LNCaP cells by reducing anchorage-dependent survival and androgen signaling. A caveolin-mediated endocytosis mechanism for MS2-mediated signaling in prostate cancer cells was proposed based on reports involving bacteriophages T4, M13, and MS2, and their interactions with LNCaP and PC3 cell lines.
Collapse
Affiliation(s)
- Swapnil Ganesh Sanmukh
- Laboratory of Extracellular Matrix Biology, Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, Botucatu, São Paulo 18618-689, Brazil,Synthetic and Systems Biology Unit, Biological Research Center, Eötvös Loránd Research Network, 6726 Szeged, Hungary,Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| | - Nilton José Dos Santos
- Laboratory of Extracellular Matrix Biology, Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, Botucatu, São Paulo 18618-689, Brazil,Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo 13083-970, Brazil
| | - Caroline Nascimento Barquilha
- Laboratory of Extracellular Matrix Biology, Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, Botucatu, São Paulo 18618-689, Brazil,Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo 13083-970, Brazil
| | - Márcio De Carvalho
- Department of Surgery and Orthopedics, Faculty of Medicine, Sao Paulo State University, Botucatu, São Paulo 18618-687, Brazil
| | - Patricia Pintor Dos Reis
- Department of Surgery and Orthopedics, Faculty of Medicine, Sao Paulo State University, Botucatu, São Paulo 18618-687, Brazil
| | - Flávia Karina Delella
- Laboratory of Extracellular Matrix Biology, Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, Botucatu, São Paulo 18618-689, Brazil
| | - Hernandes F. Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo 13083-970, Brazil
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| | - Tamás Fehér
- Synthetic and Systems Biology Unit, Biological Research Center, Eötvös Loránd Research Network, 6726 Szeged, Hungary
| | - Sérgio Luis Felisbino
- Laboratory of Extracellular Matrix Biology, Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, Botucatu, São Paulo 18618-689, Brazil,Correspondence to: Professor Sérgio Luis Felisbino, Laboratory of Extracellular Matrix Biology, Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Sao Paulo State University, 250 Antônio Celso Wagner Zanin, Botucatu, São Paulo 18618-689, Brazil, E-mail:
| |
Collapse
|
21
|
Abstract
PURPOSE Brain-derived neurotrophic factor (BDNF) belongs to the family of neurotrophic factors that can potentially increase cancer cell growth, survival, proliferation, anoikis, and migration by tyrosine kinase receptors TrkB and the p75NTR death receptor. The activation of BDNF/TrkB pathways leads to several downstream signaling pathways, including PI3K/Akt, Jak/STAT, PLCγ, Ras-Raf-MEK-ERK, NF-kB, and transactivation of EGFR. The current review aimed to provide an overview of the role of BDNF and its signaling in cancer. METHODS We searched a major medical database, PubMed, to identify eligible studies for a narrative synthesis. RESULTS Pathological examinations demonstrate BDNF overexpression in human cancer, notably involving the prostate, lung, breast, and underlying tissues, associated with a higher death rate and poor prognosis. Therefore, measurement of BDNF, either for identifying the disease or predicting response to therapy, can be helpful in cancer patients. Expression profiling studies have recognized the role of microRNAs (miR) in modulating BDNF/TrkB pathways, such as miR-101, miR-107, miR-134, miR-147, miR-191, miR-200a/c, miR-204, miR-206, miR-210, miR-214, miR-382, miR-496, miR-497, miR-744, and miR-10a-5p, providing a potential biological mechanism by which targeted therapies may correlate with decreased BDNF expression in cancers. Clinical studies investigating the use of agents targeting BDNF receptors and related signaling pathways and interfering with the related oncogenic effect, including Entrectinib, Larotrectinib, Cabozantinib, Repotrectinib, Lestaurtinib, and Selitrectinib, are in progress. CONCLUSION The aberrant signaling of BDNF is implicated in various cancers. Well-designed clinical trials are needed to clarify the BDNF role in cancer progression and target it as a therapeutic method.
Collapse
|
22
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
23
|
Upregulation of PARG in prostate cancer cells suppresses their malignant behavior and downregulates tumor-promoting genes. Biomed Pharmacother 2022; 153:113504. [PMID: 36076593 DOI: 10.1016/j.biopha.2022.113504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/03/2023] Open
Abstract
Post-translational modification of nuclear proteins through the addition of poly(ADP-ribose) (pADPr) moieties is upregulated in many metastatic cancers, where the high levels of pADPr have often been associated with poor cancer prognosis. Although the inhibitors of poly(ADP-ribose) polymerases (PARPs) have been utilized as potent anti-cancer agents, their efficacy in clinical trials varied among patient groups and has often been unpredictable. Such outcome cannot be interpreted solely by the inability to keep PARP-driven DNA repair in check. The focus of studies on PARP-driven tumorigenesis have recently been shifted toward PARP-dependent regulation of transcription. Here we utilized the controlled overexpression of poly(ADP-ribose) glycohydrolase (PARG), a sole pADPr-degrading enzyme, to investigate pADPr-dependent gene regulation in prostate cancer PC-3 cells. We demonstrated that PARG upregulation reduces pADPr levels and inhibits the expression of genes in key tumor-promoted pathways, including TNFα/NF-kB, IL6/STAT3, MYC, and KRAS signaling, the genes involved in inflammation response, especially chemokines, and endothelial-mesenchymal transition. The observed effect of PARG on transcription was consistent across all tested prostate cancer cell lines and correlates with PARG-induced reduction of clonogenic potential of PC-3 cells in vitro and a significant growth inhibition of PC-3-derived tumors in nude mice in vivo.
Collapse
|
24
|
Hellsten R, Stiehm A, Palominos M, Persson M, Bjartell A. The STAT3 inhibitor GPB730 enhances the sensitivity to enzalutamide in prostate cancer cells. Transl Oncol 2022; 24:101495. [PMID: 35917644 PMCID: PMC9344336 DOI: 10.1016/j.tranon.2022.101495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 10/25/2022] Open
Abstract
Enzalutamide is a second-generation anti-androgen which has shown increased survival in patients with metastatic prostate cancer. However, some patients do not respond to this therapy or will develop resistance to treatment over time. Signal Transducer and Activator of Transcription 3 (STAT3) is known to be involved in castration-resistant prostate cancer and to interact with androgen receptor (AR)-signaling. This study aims to investigate the combination enzalutamide and the small molecule STAT3 inhibitor GPB730 for enhanced therapeutic effect in advanced prostate cancer in vitro. The prostate cancer cell lines LNCaP (androgen dependent) and C4-2 (androgen insensitive) were used. The effect of enzalutamide and GPB730, alone and in combination, was investigated on viability and IC50 values calculated. Enzalutamide and GPB730 treated LNCaP and C4-2 cells were subjected to western blot and QPCR analyses in order to investigate the expression of AR, STAT3 and down-stream targets. C4-2 were less sensitive to growth inhibition by enzalutamide than LNCaP cells. GPB730 enhanced the growth inhibitory effect of enzalutamide in LNCaP and C4-2 cells. The addition of GPB730 to enzalutamide decreased the IC50 values for enzalutamide by 3.3-fold for LNCaP and by 12-fold for C4-2. In C4-2 cells, GPB730 alone decreased PSA expression and enhanced the enzalutamide induced decrease in NKX3.1 expression. GPB730 and enzalutamide in combination enhanced inhibition of c-myc and survivin expression. This study suggests that enzalutamide may be combined with the STAT3 inhibitor GPB730 in order to enhance the efficacy of enzalutamide, offering a new therapeutic approach in advanced prostate cancer.
Collapse
Affiliation(s)
- Rebecka Hellsten
- Department of Translational Medicine, Lund University, Scheelevägen 8, Building 404:A3, Lund SE-223 63, Sweden.
| | - Anna Stiehm
- Department of Translational Medicine, Lund University, Scheelevägen 8, Building 404:A3, Lund SE-223 63, Sweden
| | - Macarena Palominos
- Department of Translational Medicine, Lund University, Scheelevägen 8, Building 404:A3, Lund SE-223 63, Sweden
| | - Margareta Persson
- Department of Laboratory Medicine, Lund University, Scheelevägen 8, Building 404:A3, Lund SE-223 63, Sweden
| | - Anders Bjartell
- Department of Translational Medicine, Lund University, Scheelevägen 8, Building 404:A3, Lund SE-223 63, Sweden; Department of Urology, Skåne University Hospital, Jan Waldenströms gata 5, Malmö SE-205 02, Sweden
| |
Collapse
|
25
|
Yehya A, Ghamlouche F, Zahwe A, Zeid Y, Wakimian K, Mukherji D, Abou-Kheir W. Drug resistance in metastatic castration-resistant prostate cancer: an update on the status quo. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:667-690. [PMID: 36176747 PMCID: PMC9511807 DOI: 10.20517/cdr.2022.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/12/2022] [Indexed: 12/04/2022]
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related morbidity and mortality in men globally. Despite improvements in the diagnosis and treatment of PCa, a significant proportion of patients with high-risk localized disease and all patients with advanced disease at diagnosis will experience progression to metastatic castration-resistant prostate cancer (mCRPC). Multiple drugs are now approved as the standard of care treatments for patients with mCRPC that have been shown to prolong survival. Although the majority of patients will respond initially, primary and secondary resistance to these therapies make mCRPC an incurable disease. Several molecular mechanisms underlie the development of mCRPC, with the androgen receptor (AR) axis being the main driver as well as the key drug target. Understanding resistance mechanisms is crucial for discovering novel therapeutic strategies to delay or reverse the progression of the disease. In this review, we address the diverse mechanisms of drug resistance in mCRPC. In addition, we shed light on emerging targeted therapies currently being tested in clinical trials with promising potential to overcome mCRPC-drug resistance.
Collapse
Affiliation(s)
- Amani Yehya
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- Equally contributing authors
| | - Fatima Ghamlouche
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- Equally contributing authors
| | - Amin Zahwe
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- Equally contributing authors
| | - Yousef Zeid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Kevork Wakimian
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Deborah Mukherji
- Division of Hematology/Oncology, Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
26
|
Hua Y, Yuan X, Shen YH, Wang J, Azeem W, Yang S, Gade A, Lellahi SM, Øyan AM, Ke X, Zhang WD, Kalland KH. Novel STAT3 Inhibitors Targeting STAT3 Dimerization by Binding to the STAT3 SH2 Domain. Front Pharmacol 2022; 13:836724. [PMID: 35712699 PMCID: PMC9196127 DOI: 10.3389/fphar.2022.836724] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Our drug discovery model has identified two novel STAT3 SH2 domain inhibitors 323–1 and 323–2 (delavatine A stereoisomers) in a series of experiments. In silico computational modeling, drug affinity responsive target stability (DARTS), and fluorescence polarization (FP) assays altogether determined that 323–1 and 323–2 directly target the STAT3 SH2 domain and inhibited both phosphorylated and non-phosphorylated STAT3 dimerization. Computational docking predicted that compound 323s bind to three subpockets of the STAT3 SH2 domain. The 323s inhibition of STAT3 dimerization was more potent than the commercial STAT3 SH2 domain inhibitor S3I-201 in the co-immunoprecipitation assay, correlating with computational docking data. The fluorescence polarization assay further confirmed that the compound 323s target the STAT3 SH2 domain by competitively abrogating the interaction between STAT3 and the SH2-binding peptide GpYLPQTV. Compared with S3I-201, the 323 compounds exhibited stronger inhibition of STAT3 and reduced the level of IL-6-stimulated phosphorylation of STAT3 (Tyr705) in LNCaP cells over the phosphorylation of STAT1 (Tyr701) induced by IFN-ɣ in PC3 cells or the phosphorylation of STAT1 (Ser727) in DU145 cells. Both compounds downregulated STAT3 target genes MCL1 and cyclin D1. Thus, the two compounds are promising lead compounds for the treatment of cancers with hyper-activated STAT3.
Collapse
Affiliation(s)
- Yaping Hua
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- *Correspondence: Yaping Hua, ; Wei-dong Zhang, ; Karl-Henning Kalland,
| | - Xing Yuan
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yun-heng Shen
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jinxin Wang
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Waqas Azeem
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Shuo Yang
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Alexandra Gade
- Centre for Molecular Medicine Norway, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Seyed Mohammad Lellahi
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Anne Margrete Øyan
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xisong Ke
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei-dong Zhang
- Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yaping Hua, ; Wei-dong Zhang, ; Karl-Henning Kalland,
| | - Karl-Henning Kalland
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
- *Correspondence: Yaping Hua, ; Wei-dong Zhang, ; Karl-Henning Kalland,
| |
Collapse
|
27
|
Zhang Q, Zhou XM, Wei SZ, Cui DS, Deng KL, Liang G, Luo Y, Luo B, Liang XJ. STAT3 as a target for sensitizing prostate cancer cells to irradiation. JOURNAL OF RADIATION RESEARCH 2022; 63:174-182. [PMID: 34970978 PMCID: PMC8944309 DOI: 10.1093/jrr/rrab117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Radioresistance of prostate cancer (PCa) is a major factor leading to local failure of radiotherapy. STAT3 is an oncogenic protein that was recently found to be activated in PCa tumors. This study aimed to investigate the radiosensitization effect of targeting STAT3 in PCa tumors. Here, the radiosensitization effect of STAT3 blockade was investigated by clonogenic assay, flow cytometry and western blot analysis in human PCa cells in vitro and in vivo. We demonstrated that STAT3 blockade with a STAT3 inhibitor or siRNA increased the radiosensitivity of PCa cells and that radiation together with STAT3 blockade induced more apoptosis and double-strand breaks (DSBs) than radiation alone in LNCaP cells. In addition, radiation induced STAT3 activation and survivin expression in PCa cells, which was inhibited by STAT3 blockade. Transfection with survivin cDNA attenuated the radiosensitization effect of STAT3 blockade. These effects were further confirmed by in vivo studies, which showed that the STAT3 inhibitor enhanced the treatment efficacy of radiation on LNCaP xenografts with decreased STAT3 activation and survivin expression. These findings suggest that STAT3 blockade radiosensitizes PCa cells through regulation of survivin. Thus, our study has revealed STAT3 as a potential sensitizer for irradiation in PCa cells. Its clinical application as an adjuvant in radiotherapy of PCa should be explored in the future.
Collapse
Affiliation(s)
| | | | - Shao-Zhong Wei
- Department of Urology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dian-Sheng Cui
- Department of Urology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang-Li Deng
- Department of Urology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gai Liang
- Department of Radiotherapy Center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Luo
- Department of Radiotherapy Center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Luo
- Corresponding author. No. 116 Zhuodaoquan South Road, Wuhan, Hubei Province, 430000, People’s Republic of China. Telephone: 86-27-87287963;
| | - Xin-Jun Liang
- Department of Abdominal Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Araki T, Watanabe Y, Okada Y, Murakami H, Ogo N, Asai A. Identification of serum and glucocorticoid-regulated kinase 1 as a regulator of signal transducer and activator of transcription 3 signaling. Exp Cell Res 2022; 413:113079. [PMID: 35202674 DOI: 10.1016/j.yexcr.2022.113079] [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: 10/18/2021] [Revised: 12/20/2021] [Accepted: 02/11/2022] [Indexed: 11/18/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays key roles in cancer cell proliferation, invasion, and immunosuppression. In many human cancer cells, STAT3 is hyperactivated, which leads to tumor progression and drug resistance, and therefore STAT3 and its modulators are considered effective drug targets. However, the complex regulatory mechanisms of STAT3 have made it difficult to develop potent anticancer drugs that suppress its activity. Here, we report serum and glucocorticoid-regulated kinase 1 (SGK1) as a novel regulator of STAT3 signaling and an effective target for combination therapy with Janus kinase (JAK) inhibitors. We screened small molecules using a gain-of-function mutant of STAT3 resistant to JAK inhibition and found that an SGK1 inhibitor suppressed the constitutive activation of STAT3. Importantly, our results revealed that SGK1 also mediated the activation of wild-type STAT3. Further examination suggested that the tuberous sclerosis complex 2 and mammalian target of rapamycin signaling pathway were involved in STAT3 activation by SGK1. Finally, we demonstrated that SGK1 inhibition enhanced the inhibitory effect of a JAK inhibitor on STAT3 phosphorylation and cancer cell proliferation. Our findings provide new insights into the molecular mechanisms of STAT3 activation and suggest SGK1 as a potential target for STAT3-targeted combination cancer therapy.
Collapse
Affiliation(s)
- Toshihiro Araki
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan; Discovery Technology Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Muraoka-Higashi, Fujisawa, Kanagawa, Japan
| | - Yuuki Watanabe
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan; Public Affairs and Policy Department, Mitsubishi Tanabe Pharma Corporation, Marunouchi, Chiyoda-ku, Tokyo, Japan
| | - Yusuke Okada
- Sohyaku Project Planning & Management Department, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Marunouchi, Chiyoda-ku, Tokyo, Japan
| | - Hisashi Murakami
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, Shizuoka, Japan.
| |
Collapse
|
29
|
Association of STAT3, PTPRT, TNK2-AS1, LINC-ROR Genes Expression Level with Prostate Cancer and Benign Prostatic Hyperplasia. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2022. [DOI: 10.5812/ijcm.120188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Prostate cancer (PCa) and benign prostate hyperplasia (BPH) are highly prevalent heterogeneous disorders among men. Since angiogenesis is the key step in cancer progression, the deregulation of genes involved in this process may play a role in cancer development. Objectives: We evaluated the expression level of 4 angiogenesis-related genes including signal transducer and activator of transcription 3 (STAT3), protein tyrosine phosphatase receptor type T (PTPRT), TNK2 antisense RNA 1 (TNK2-AS1), and long intergenic non-protein coding rna-regulator of reprogramming (LINC-ROR) in patients with PCa and BPH. Methods: The expression level of STAT3, PTPRT, TNK2-AS1, and LINC-ROR genes in tumoral and adjacent non-cancerous tissue (ANCT) samples of 50 PCa patients and tissue samples from 50 BPH patients were evaluated, using the real-time PCR method. The statistical analysis was performed to evaluate the association between genes expression and clinicopathological characteristics of patients with PCa. Results: The expression level of STAT3 and LINC-ROR was upregulated in tumoral tissues compared to ANCTs (P < 0.0001 for both). Only the expression level of STAT3 in PCa was higher than in BPH tissues (P = 0.001). The elevated expression of STAT3 was associated with the higher grade group of the tumor (P = 0.03). Also, the high expression level of PTPRT and LINC-ROR genes was associated with a higher stage of cancer in patients with PCa (P = 0.002, P = 0.0001 respectively). The STAT3 gene transcript level had an excellent diagnostic power for discrimination between tumoral tissue and the ANCTs with an area under the curve (AUC) of 0.93. Conclusions: The higher expression of STAT3 and LINC-ROR suggested a role in the pathogenesis of PCa in higher stages. Also, STAT3 expression level could be suggested as a potential biomarker for PCa in combination with PSA level.
Collapse
|
30
|
Nuclear expression of pSTAT3Tyr705 and pSTAT3Ser727 in the stromal compartment of localized hormone-naïve prostate cancer. Pathol Res Pract 2022; 232:153811. [DOI: 10.1016/j.prp.2022.153811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022]
|
31
|
Mitsogiannis I, Tzelves L, Dellis A, Issa H, Papatsoris A, Moussa M. Prostate cancer immunotherapy. Expert Opin Biol Ther 2022; 22:577-590. [PMID: 35037527 DOI: 10.1080/14712598.2022.2027904] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Medical treatment for prostate cancer (PC) targets hormonal pathways used by malignant cells. Research advances aided in gaining knowledge about implicated molecular pathways and opened the way for establishment of new types of therapies by modifying immunological mechanisms. The aim of this review is to present completed and ongoing research projects regarding PC immunotherapy. AREAS COVERED A literature search was conducted in PubMed/MEDLINE, Scopus, Cochrane Central Register of Controlled Trials, and https://www.clinicaltrials.gov/ from inception until 07/2021, to identify completed or ongoing Phase III trials regarding several immunotherapies against PC. Studies on vaccine therapies, CTLA-4 inhibitors, PD-1/PD-L1 inhibitors, PARP inhibitors, PSMA-targeted therapies, and tyrosine kinase inhibitors were considered eligible. EXPERT OPINION Although many molecules are being tested against PC cells, only sipuleucel-T has gain approval in the USA. The main reason for this delay in establishing immunotherapy as a standard option for managing PC is the heterogeneity and tumor immune microenvironment complexities. Ipilimumab and olaparib were proved to prolong overall survival significantly against placebo, but a lot of research is going on to identify which patients and at what stage of disease will benefit the most before incorporating them in clinical practice. More recent options such as PSMA-targeted treatments are currently evaluated. ARTICLE HIGHLIGHTS Intense research performed on immunotherapy for prostate cancer.Vaccine therapy with sipuleucel-T, the only approved immunotherapy for prostate cancer.Ipilimumab shows survival benefits.Olaparib shows survival benefits.Findings should be confirmed on further trials to identify target population characteristics and proper disease stage.Immunotherapy is not yet a standard due to tumor environment complex interaction between immune system and malignant cells.
Collapse
Affiliation(s)
- Iraklis Mitsogiannis
- 2nd Department of Urology, School of Medicine, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Lazaros Tzelves
- 2nd Department of Urology, School of Medicine, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Dellis
- 2nd Department of Urology, School of Medicine, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece.,Department of Surgery, School of Medicine, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Hussein Issa
- Department of Urology, Al Zahraa Hospital, University Medical Center, Lebanese University, Beirut, Lebanon
| | - Athanasios Papatsoris
- 2nd Department of Urology, School of Medicine, Sismanoglio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Mohammad Moussa
- Department of Urology, Al Zahraa Hospital, University Medical Center, Lebanese University, Beirut, Lebanon
| |
Collapse
|
32
|
The Metastasis Suppressor NDRG1 Directly Regulates Androgen Receptor Signaling in Prostate Cancer. J Biol Chem 2021; 297:101414. [PMID: 34785213 PMCID: PMC8668986 DOI: 10.1016/j.jbc.2021.101414] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
N-myc-downregulated gene 1 (NDRG1) has potent anticancer effects and inhibits cell growth, survival, metastasis, and angiogenesis. Previous studies suggested that NDRG1 is linked to the androgen signaling network, but this mechanistic relationship is unclear. Considering the crucial role of the androgen receptor (AR) in prostate cancer (PCa) progression, here we examined for the first time the effect of NDRG1 on AR expression, activation, and downstream signaling in LNCaP, 22Rv1, and C4-2B PCa cell types. We demonstrate that NDRG1 effectively promotes interaction of AR with the chaperone HSP90, which in turn stabilizes the AR while decreasing its androgen-mediated activation. The expression of NDRG1 suppressed: (1) AR activation, as measured by p-ARSer213 and p-ARSer81; (2) expression of a major AR transcriptional target, prostate-specific antigen (PSA); and (3) AR transcriptional activity, probably via inhibiting the c-Jun-AR interaction by reducing c-Jun phosphorylation (p-c-JunSer63). NDRG1 was also demonstrated to inhibit multiple key molecules involved in androgen-dependent and -independent signaling (namely EGFR, HER2, HER3, PI3K, STAT3, and NF-κB), which promote the development of castration-resistant prostate cancer. We also identified the cysteine-rich secretory protein/antigen 5/pathogenesis related-1 (CAP) domain of NDRG1 as vital for inhibition of AR activity. Examining NDRG1 and p-NDRG1 in PCa patient specimens revealed a significant negative correlation between NDRG1 and PSA levels in prostatectomy patients that went on to develop metastasis. These results highlight a vital role for NDRG1 in androgen signaling and its potential as a key therapeutic target and biomarker in PCa.
Collapse
|
33
|
Singh KB, Hahm ER, Singh SV. Leelamine suppresses cMyc expression in prostate cancer cells in vitro and inhibits prostate carcinogenesis in vivo. JOURNAL OF CANCER METASTASIS AND TREATMENT 2021; 7. [PMID: 34660908 DOI: 10.20517/2394-4722.2021.08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aim Leelamine (LLM) inhibits growth of human prostate cancer cells but the underlying mechanism is not fully understood. The present study was undertaken to determine the effect of LLM on cMyc, which is overexpressed in a subset of human prostate cancers. Methods The effect of LLM on cMyc expression and activity was determined by western blotting/confocal microscopy and luciferase reporter assay, respectively. A transgenic mouse model of prostate cancer (Hi-Myc) was used to determine chemopreventive efficacy of LLM. Results Exposure of androgen sensitive (LNCaP) and castration-resistant (22Rv1) human prostate cancer cells to LLM resulted in downregulation of protein and mRNA levels of cMyc. Overexpression of cMyc partially attenuated LLM-mediated inhibition of colony formation, cell viability, and cell migration in 22Rv1 and/or PC-3 cells. LLM treatment decreased protein levels of cMyc targets (e.g., lactate dehydrogenase), however, overexpression of cMyc did not attenuate these effects. A trend for a decrease in expression level of cMyc protein was discernible in 22Rv1 xenografts from LLM-treated mice compared with control mice. The LLM treatment (10 mg/kg body weight, 5 times/week) was well-tolerated by Hi-Myc transgenic mice. The incidence of high-grade prostatic intraepithelial neoplasia, adenocarcinoma in situ, and microinvasion was lower in LLM-treated Hi-Myc mice but the difference was not statistically significant. Conclusion The present study reveals that LLM inhibits cMyc expression in human prostate cancer cells in vitro but concentrations higher than 10 mg/kg may be required to achieve chemoprevention of prostate cancer.
Collapse
Affiliation(s)
- Krishna B Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
34
|
Ebersbach C, Beier AMK, Thomas C, Erb HHH. Impact of STAT Proteins in Tumor Progress and Therapy Resistance in Advanced and Metastasized Prostate Cancer. Cancers (Basel) 2021; 13:4854. [PMID: 34638338 PMCID: PMC8508518 DOI: 10.3390/cancers13194854] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/17/2022] Open
Abstract
Signal transducers and activators of transcription (STATs) are a family of transcription factors involved in several biological processes such as immune response, cell survival, and cell growth. However, they have also been implicated in the development and progression of several cancers, including prostate cancer (PCa). Although the members of the STAT protein family are structurally similar, they convey different functions in PCa. STAT1, STAT3, and STAT5 are associated with therapy resistance. STAT1 and STAT3 are involved in docetaxel resistance, while STAT3 and STAT5 are involved in antiandrogen resistance. Expression of STAT3 and STAT5 is increased in PCa metastases, and together with STAT6, they play a crucial role in PCa metastasis. Further, expression of STAT3, STAT5, and STAT6 was elevated in advanced and high-grade PCa. STAT2 and STAT4 are currently less researched in PCa. Since STATs are widely involved in PCa, they serve as potential therapeutic targets. Several inhibitors interfering with STATs signaling have been tested unsuccessfully in PCa clinical trials. This review focuses on the respective roles of the STAT family members in PCa, especially in metastatic disease and provides an overview of STAT-inhibitors evaluated in clinical trials.
Collapse
Affiliation(s)
- Celina Ebersbach
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.)
- Mildred Scheel Early Career Center, Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Alicia-Marie K. Beier
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.)
- Mildred Scheel Early Career Center, Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Christian Thomas
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.)
| | - Holger H. H. Erb
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany; (C.E.); (A.-M.K.B.); (C.T.)
| |
Collapse
|
35
|
Ma M, Zhao Z, Liang Q, Shen H, Zhao Z, Chen Z, He R, Feng S, Cao D, Gan G, Ye H, Qiu W, Deng J, Ming F, Jia J, Sun C, Li J, Zhang L. Overexpression of pEGF improved the gut protective function of Clostridium butyricum partly through STAT3 signal pathway. Appl Microbiol Biotechnol 2021; 105:5973-5991. [PMID: 34396488 DOI: 10.1007/s00253-021-11472-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/25/2022]
Abstract
Clostridium butyricum (C. butyricum) is a probiotic that could promote animal growth and protect gut health. So far, current studies mainly keep up with the basic biological functions of C. butyricum, missing the effective strategy to further improve its protective efficiency. A recent report about C. butyricum alleviating intestinal injury through epidermal growth factor receptor (EGFR) inspired us to bridge this gap by porcine epidermal growth factor (EGF) overexpression. Lacking a secretory overexpression system, we constructed the recombinant strains overexpressing pEGF in C. butyricum for the first time and obtained 4 recombinant strains for highly efficient secretion of pEGF (BC/pPD1, BC/pSPP, BC/pGHF, and BC/pDBD). Compared to the wild-type strain, we confirmed that the expression level ranges of the intestinal development-related genes (Claudin-1, GLUT-2, SUC, GLP2R, and EGFR) and anti-inflammation-related gene (IL-10) in IPECs were upregulated under recombinant strain stimulation, and the growth of Staphylococcus aureus and Salmonella typhimurium was significantly inhibited as well. Furthermore, a particular inhibitor (stattic) was used to block STAT3 tyrosine phosphorylation, resulting in the downregulation on antibacterial effect of recombinant strains. This study demonstrated that the secretory overexpression of pEGF in C. butyricum could upregulate the expression level of EGFR, consequently improving the intestinal protective functions of C. butyricum partly following STAT3 signal activation in IPECs and making it a positive loop. These findings on the overexpression strains pointed out a new direction for further development and utilization of C. butyricum. KEY POINTS: • By 12 signal peptide screening in silico, 4 pEGF overexpression strains of C. butyricum/pMTL82151-pEGF for highly efficient secretion of pEGF were generated for the first time. • The secretory overexpression of pEGF promoted the intestinal development, antimicrobial action, and anti-inflammatory function of C. butyricum. • The overexpressed pEGF upregulated the expression level of EGFR and further magnified the gut protective function of recombinant strains which in turn partly depended on STAT3 signal pathway in IPECs.
Collapse
Affiliation(s)
- Miaopeng Ma
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Zitong Zhao
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Qianyi Liang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Haokun Shen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Zengjue Zhao
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Zhiyang Chen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Rongxiao He
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Saixiang Feng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Ding Cao
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Guanhua Gan
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Hejia Ye
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Weihong Qiu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Jinbo Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Feiping Ming
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Junhao Jia
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Chongjun Sun
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Jiayi Li
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China
| | - Linghua Zhang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Wushan Road, Tianhe District, Guangzhou, 510642, Guangdong, China.
| |
Collapse
|
36
|
Leonel ECR, Ruiz TFR, Bedolo CM, Campos SGP, Taboga SR. Inflammatory repercussions in female steroid responsive glands after perinatal exposure to bisphenol A and 17-β estradiol. Cell Biol Int 2021; 45:2264-2274. [PMID: 34288236 DOI: 10.1002/cbin.11665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/18/2022]
Abstract
The mammary gland (MG) and female prostate are plastic reproductive organs which are highly responsive to hormones. Thus, endocrine disruptors, such as bisphenol A (BPA) and exogenous estrogens, negatively affect glandular homeostasis. In addition to previously described alterations, changes in inflammatory markers expression also trigger the development of a microenvironment that contributes to tumor progression. The current work aimed to evaluate the inflammatory responses of the MG and prostate gland to BPA (50 µg/kg) and 17-β estradiol (35 µg/kg) exposure during the perinatal window of susceptibility. The results showed that at 6 months of age there was an increase in the number of phospho-STAT3 (P-STAT3) positive cells in the female prostate from animals perinatally exposed to 50 µg/kg BPA daily. In addition, the number of macrophages increased in these animals in comparison with nonexposed animals, as shown by the F4/80 marker. Despite an increase in the incidence of lobuloalveolar and intraductal hyperplasia, the MG did not show any difference in the expression of the four inflammatory markers evaluated: tumor necrosis factor-α, COX-2, P-STAT3, and F4/80. Analysis of both glands from the same animal led to the conclusion that exposure to endocrine disruptors during the perinatal window of susceptibility leads to different inflammatory responses in different reproductive organs. As the prostate is more susceptible to these inflammatory mechanisms, it is reasonable to affirm that possible neoplastic alterations in this organ are related to changes in the inflammatory pattern of the stroma, a characteristic that is not evident in the MG.
Collapse
Affiliation(s)
- Ellen Cristina Rivas Leonel
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil.,Department of Histology, Embriology, and Cell Biology, Institute of Biological Sciences (ICB III), Federal University of Goiás (UFG), Goiânia, Goiás, Brazil
| | - Thalles Fernando Rocha Ruiz
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Carolina Marques Bedolo
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Silvana Gisele Pegorin Campos
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biology, Humanities, and Exact Sciences, Institute of Biosciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
37
|
Interplay of Epidermal Growth Factor Receptor and Signal Transducer and Activator of Transcription 3 in Prostate Cancer: Beyond Androgen Receptor Transactivation. Cancers (Basel) 2021; 13:cancers13143452. [PMID: 34298665 PMCID: PMC8307975 DOI: 10.3390/cancers13143452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 01/16/2023] Open
Abstract
Prostate cancer (PCa) is one of the most common cancers in the world and causes thousands of deaths every year. Conventional therapy for PCa includes surgery and androgen deprivation therapy (ADT). However, about 10-20% of all PCa cases relapse; there is also the further development of castration resistant adenocarcinoma (CRPC-Adeno) or neuroendocrine (NE) PCa (CRPC-NE). Due to their androgen-insensitive properties, both CRPC-Adeno and CRPC-NE have limited therapeutic options. Accordingly, this study reveals the inductive mechanisms of CRPC (for both CRPC-Adeno and CRPC-NE) and fulfils an urgent need for the treatment of PCa patients. Although previous studies have illustrated the emerging roles of epidermal growth factor receptors (EGFR), signal transducer, and activator of transcription 3 (STAT3) signaling in the development of CRPC, the regulatory mechanisms of this interaction between EGFR and STAT3 is still unclear. Our recent studies have shown that crosstalk between EGFR and STAT3 is critical for NE differentiation of PCa. In this review, we have collected recent findings with regard to the involvement of EGFR and STAT3 in malignancy progression and discussed their interactions during the development of therapeutic resistance for PCa.
Collapse
|
38
|
Crowley F, Sterpi M, Buckley C, Margetich L, Handa S, Dovey Z. A Review of the Pathophysiological Mechanisms Underlying Castration-resistant Prostate Cancer. Res Rep Urol 2021; 13:457-472. [PMID: 34235102 PMCID: PMC8256377 DOI: 10.2147/rru.s264722] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Androgen deprivation therapy or ADT is one of the cornerstones of management of locally advanced or metastatic prostate cancer, alongside radiation therapy. However, despite early response, most advanced prostate cancers progress into an androgen unresponsive or castrate resistant state, which hitherto remains an incurable entity and the second leading cause of cancer-related mortality in men in the US. Recent advances have uncovered multiple complex and intermingled mechanisms underlying this transformation. While most of these mechanisms revolve around androgen receptor (AR) signaling, novel pathways which act independently of the androgen axis are also being discovered. The aim of this article is to review the pathophysiological mechanisms that help bypass the apoptotic effects of ADT to create castrate resistance. The article discusses castrate resistance mechanisms under two categories: 1. Direct AR dependent pathways such as amplification or gain of function mutations in AR, development of functional splice variants, posttranslational regulation, and pro-oncogenic modulation in the expression of coactivators vs corepressors of AR. 2. Ancillary pathways involving RAS/MAP kinase, TGF-beta/SMAD pathway, FGF signaling, JAK/STAT pathway, Wnt-Beta catenin and hedgehog signaling as well as the role of cell adhesion molecules and G-protein coupled receptors. miRNAs are also briefly discussed. Understanding the mechanisms involved in the development and progression of castration-resistant prostate cancer is paramount to the development of targeted agents to overcome these mechanisms. A number of targeted agents are currently in development. As we strive for more personalized treatment across oncology care, treatment regimens will need to be tailored based on the type of CRPC and the underlying mechanism of castration resistance.
Collapse
Affiliation(s)
- Fionnuala Crowley
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Michelle Sterpi
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Conor Buckley
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Lauren Margetich
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Shivani Handa
- Department of Internal Medicine, Icahn School of Medicine, Mount Sinai Morningside and West, New York, NY, USA
| | - Zach Dovey
- Department of Urology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
39
|
Bao X, Zhu J, Ren C, Zhao A, Zhang M, Zhu Z, Lu X, Zhang Y, Li X, Sima X, Li J, Zhang Q, Ma B. β-elemonic acid inhibits growth and triggers apoptosis in human castration-resistant prostate cancer cells through the suppression of JAK2/STAT3/MCL-1 and NF-ĸB signal pathways. Chem Biol Interact 2021; 342:109477. [PMID: 33878321 DOI: 10.1016/j.cbi.2021.109477] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/02/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022]
Abstract
Castration-resistant prostate cancer (CRPC) has become a significant problem in the current treatment of prostate cancer (PCa) with the characteristics of high metastatic potential, resistance and easy recurrence. The abnormal activation of JAK2/STAT3/MCL-1 and NF-κB has been confirmed as the main reason for the development of CRPC. We previously found that β-elemonic acid (β-EA) as a natural triterpene has potential anti-inflammatory and anti-osteosarcoma effects with lower toxicity. But it remains unknown whether it had effects on CRPC. The present research in vitro and in vivo systematically investigates anti-cancer effects and mechanisms of β-EA on human CRPC. β-EA treatment resulted in apoptotic cell death in human PCa cells by mitochondrial apoptotic pathways (including up-regulation of cleaved caspase-3, cleaved PARP, and Bax or down-regulation of Bcl-2). Besides, β-EA at relatively lower levels inhibited colony-forming, the migration and invasion potential of PCa cells, indicating its anti-proliferation and anti-metastasis activities. After exploring the potential mechanism, our results suggested that it subsequently inhibited the activation of JAK2/STAT3/MCL-1 and NF-κB signaling pathway by the administration of β-EA. The silencing of NF-κB/p65, JAK2 and STAT3, respectively, increased the sensitivity of the PCa cells to β-EA induced apoptosis. Moreover, β-EA exhibited a strong affinity with its essential proteins JAK2, RELA/p65, NF-κBIα/IκBα by molecular docking analysis. Importantly, β-EA retards tumor growth in a murine xenograft model, consistent with our study in vitro. Taken together, findings from this study reveal for the first time the potential role and mechanisms of β-EA on CRPC.
Collapse
Affiliation(s)
- Xiaowen Bao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Jianwei Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Chaoxing Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Ang Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Mingya Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Zhiming Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Xuanzhao Lu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yuning Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
| | - Xinyu Sima
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Jiaqi Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
40
|
Akhter MS, Uddin MA, Kubra KT, Barabutis N. Elucidation of the Molecular Pathways Involved in the Protective Effects of AUY-922 in LPS-Induced Inflammation in Mouse Lungs. Pharmaceuticals (Basel) 2021; 14:ph14060522. [PMID: 34072430 PMCID: PMC8226636 DOI: 10.3390/ph14060522] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) cause thousands of deaths every year and are associated with high mortality rates (~40%) due to the lack of efficient therapies. Understanding the molecular mechanisms associated with those diseases will most probably lead to novel therapeutics. In the present study, we investigated the effects of the Hsp90 inhibitor AUY-922 in the major inflammatory pathways of mouse lungs. Mice were treated with LPS (1.6 mg/kg) via intratracheal instillation for 24 h and were then post-treated intraperitoneally with AUY-922 (10 mg/kg). The animals were examined 48 h after AUY-922 injection. LPS activated the TLR4-mediated signaling pathways, which in turn induced the release of different inflammatory cytokines and chemokines. AUY-922 suppressed the LPS-induced inflammation by inhibiting major pro-inflammatory pathways (e.g., JAK2/STAT3, MAPKs), and downregulated the IL-1β, IL-6, MCP-1 and TNFα. The expression levels of the redox regulator APE1/Ref1, as well as the DNA-damage inducible kinases ATM and ATR, were also increased after LPS treatment. Those effects were counteracted by AUY-922. Interestingly, this Hsp90 inhibitor abolished the LPS-induced pIRE1α suppression, a major component of the unfolded protein response. Our study elucidates the molecular pathways involved in the progression of murine inflammation and supports our efforts on the development of new therapeutics against lung inflammatory diseases and sepsis.
Collapse
|
41
|
Pihlstrøm N, Jin Y, Nenseth Z, Kuzu OF, Saatcioglu F. STAMP2 Expression Mediated by Cytokines Attenuates Their Growth-Limiting Effects in Prostate Cancer Cells. Cancers (Basel) 2021; 13:cancers13071579. [PMID: 33808059 PMCID: PMC8036285 DOI: 10.3390/cancers13071579] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Prostate cancer (PCa) is the most common non-skin cancer and one of the leading causes of cancer death in men. Despite significant developments in therapy options with improved survival, no curative treatment is currently available. We have previously identified six transmembrane protein of prostate 2 (STAMP2) as an important factor for PCa growth and survival. We now show that STAMP2 expression is regulated by inflammatory signaling, which has recently been implicated in PCa. Two proinflammatory cytokines, interleukin 6 and interleukin 1 beta, synergize with each other to induce STAMP2 expression. Interestingly, STAMP2 knockdown increased the sensitivity of PCa cells to cytokine treatment. Thus, STAMP2 that acts as a survival factor in PCa, is both independently and synergistically regulated by inflammatory signaling that may affect disease progression. Abstract Inflammatory events and dysregulated cytokine expression are implicated in prostate cancer (PCa), but the underlying molecular mechanisms are poorly understood at present. We have previously identified six transmembrane protein of the prostate 2 (STAMP2, also known as STEAP4) as an androgen-regulated gene, as well as a key regulator of PCa growth and survival. STAMP2 is also regulated by, and participates in, inflammatory signaling in other tissues and pathologies. Here, we show that the proinflammatory cytokines interleukin 6 (IL-6) and Interleukin 1 beta (IL-1β) significantly increase and strongly synergize in promoting STAMP2 expression in PCa cells. The two cytokines increase androgen-induced STAMP2 expression, but not expression of other known androgen target genes, suggesting a unique interplay of androgens and cytokines in regulating STAMP2 expression. Interestingly, STAMP2 knockdown significantly increased the ability of IL-6 and IL-1β to inhibit PCa cell growth in vitro. These results suggest that STAMP2 may represent a unique node through which inflammatory events mediate their effects on PCa growth and survival.
Collapse
Affiliation(s)
- Nicklas Pihlstrøm
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Yang Jin
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Zeynep Nenseth
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Omer F. Kuzu
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
- Correspondence: (O.F.K.); (F.S.); Tel.: +47-22-854-569 (F.S.); Fax: +47-22-857-207 (F.S.)
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0188 Oslo, Norway
- Correspondence: (O.F.K.); (F.S.); Tel.: +47-22-854-569 (F.S.); Fax: +47-22-857-207 (F.S.)
| |
Collapse
|
42
|
Ehsani M, David FO, Baniahmad A. Androgen Receptor-Dependent Mechanisms Mediating Drug Resistance in Prostate Cancer. Cancers (Basel) 2021; 13:1534. [PMID: 33810413 PMCID: PMC8037957 DOI: 10.3390/cancers13071534] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is a main driver of prostate cancer (PCa) growth and progression as well as the key drug target. Appropriate PCa treatments differ depending on the stage of cancer at diagnosis. Although androgen deprivation therapy (ADT) of PCa is initially effective, eventually tumors develop resistance to the drug within 2-3 years of treatment onset leading to castration resistant PCa (CRPC). Castration resistance is usually mediated by reactivation of AR signaling. Eventually, PCa develops additional resistance towards treatment with AR antagonists that occur regularly, also mostly due to bypass mechanisms that activate AR signaling. This tumor evolution with selection upon therapy is presumably based on a high degree of tumor heterogenicity and plasticity that allows PCa cells to proliferate and develop adaptive signaling to the treatment and evolve pathways in therapy resistance, including resistance to chemotherapy. The therapy-resistant PCa phenotype is associated with more aggressiveness and increased metastatic ability. By far, drug resistance remains a major cause of PCa treatment failure and lethality. In this review, various acquired and intrinsic mechanisms that are AR‑dependent and contribute to PCa drug resistance will be discussed.
Collapse
Affiliation(s)
| | | | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07740 Jena, Germany; (M.E.); (F.O.D.)
| |
Collapse
|
43
|
Zafar E, Maqbool MF, Iqbal A, Maryam A, Shakir HA, Irfan M, Khan M, Li Y, Ma T. A comprehensive review on anticancer mechanism of bazedoxifene. Biotechnol Appl Biochem 2021; 69:767-782. [PMID: 33759222 DOI: 10.1002/bab.2150] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Cancer is counted as a second leading cause of death among nontransmissible diseases. Identification of novel anticancer drugs is therefore necessary for the effective treatment of cancer. Conventional drug discovery is time consuming and expensive process. Unlike conventional drug discovery, drug repositioning offers a novel strategy for urgent drug discovery since it is a cost-effective and faster process. Bazedoxifene (BZA) is a synthetic selective estrogen receptor modulator, approved by the United States Food and Drug Administration for the treatment of osteoporosis in postmenopausal women. BZA is now being studied for its anticancer activity in various cancers including breast cancer, liver cancer, pancreatic cancer, colon cancer, head and neck cancer, medulloblastoma, brain cancer, and gastrointestinal cancer. Studies have reported that BZA is effective in reducing cancer progression through multiple mechanisms. BZA could effectively inhibit STAT3, PI3K/AKT, and MAPK signaling pathways and induce apoptosis. In addition to its anticancer activity as monotherapy, BZA has been shown to enhance the chemotherapeutic efficacy of clinical drugs such as paclitaxel, cisplatin, palbociclib, and oxaliplatin in multiple neoplasms. This review mainly focused on the anticancer activity, cellular targets, and anticancer mechanism of BZA, which may help the further design and conduct of research and repositioning it for oncological clinic trials.
Collapse
Affiliation(s)
- Erum Zafar
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | | | - Asia Iqbal
- Department of Wild Life and Ecology, University of Veternary and Animal Sciences, Ravi Campus, Patoki, Pakistan
| | - Amara Maryam
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Hafiz Abdullah Shakir
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Muhammad Khan
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The olive tree (Olea europaea L.) has featured as a significant part of medicinal history, used to treat a variety of ailments within folk medicine. The Mediterranean diet, which is rich in olive products, is testament to Olea europaeas positive effects on health, associated with reduced incidences of cancer and cardiovascular disease. This review aims to summarise the current literature regarding the therapeutic potential of Olea europaea products in cancer, detailing the possible compounds responsible for its chemotherapeutic effects. RECENT FINDINGS Much of the existing research has focused on the use of cell culture models of disease, demonstrating Olea europaea extracts, and specific compounds within these extracts, have efficacy in a range of in vitro and in vivo cancer models. The source of Olea europaeas cytotoxicity is yet to be fully defined; however, compounds such as oleuropein and verbascoside have independent cytotoxic effects on animal models of cancer. Initial results from animal models are promising but need to be translated to a clinical setting. Treatments utilising these compounds are likely to be well tolerated and represent a promising direction for future research.
Collapse
Affiliation(s)
- Chrystalla Antoniou
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Jonathon Hull
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
45
|
Das J, Barman Mandal S. Classification of Homo sapiens gene behavior using linear discriminant analysis fused with minimum entropy mapping. Med Biol Eng Comput 2021; 59:673-691. [PMID: 33595791 DOI: 10.1007/s11517-021-02324-y] [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: 07/02/2020] [Accepted: 01/18/2021] [Indexed: 11/25/2022]
Abstract
Classification of Homo sapiens gene behavior employing computational biology is a recent research trend. But monitoring gene activity profile and genetic behavior from the alphabetic DNA sequence using a non-invasive method is a tremendous challenge in functional genomics. The present paper addresses such issue and attempts to differentiate Homo sapiens genes using linear discriminant analysis (LDA) method. Annotated protein coding sequences of Homo sapiens genes, collected from NCBI, are taken as test samples. Minimum entropy-based mapping (MEM) technique assists to extract highest information from the numerical DNA sequences. The proposed LDA technique has successfully classified Homo sapiens genes based on the following features: composition of hydrophilic amino acids, dominance of arginine amino acid, and magnitude and size of individual amino acids. The proposed algorithm is successfully tested on 84 Homo sapiens healthy and cancer genes of the prostate and breast cells. Classification performance of the proposed LDA technique is judged by sensitivity (89.12%), specificity (91.9%), accuracy (90.87%), F1 score (92.03%), Matthews' correlation coefficients (81.04%), and miss rate (9.12%), and it outperforms other four existing classifiers. The results are cross-validated through Rayleigh PDF and mutual information technique. Fisher test, 2-sample T-test, and relative entropy test are considered to verify the efficacy of the present classifier.
Collapse
Affiliation(s)
- Joyshri Das
- Institute of Radio Physics & Electronics, University of Calcutta, Kolkata, India
| | - Soma Barman Mandal
- Institute of Radio Physics & Electronics, University of Calcutta, Kolkata, India
| |
Collapse
|
46
|
Fan X, Zhou J, Bi X, Liang J, Lu S, Yan X, Luo L, Yin Z. L-theanine suppresses the metastasis of prostate cancer by downregulating MMP9 and Snail. J Nutr Biochem 2020; 89:108556. [PMID: 33249185 DOI: 10.1016/j.jnutbio.2020.108556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/26/2020] [Accepted: 11/21/2020] [Indexed: 12/18/2022]
Abstract
Prostate cancer (PCa) is a very prevalent male-specific malignancy; most PCa patients eventually die as a result of metastasis. L-theanine (C7H14N2O3), a nonprotein amino acid derivative from green tea leaves, has been demonstrated to act as an anticarcinogen through proapoptotic and antiproliferative effects. However, the antimetastatic effect of L-theanine in tumor cells and its underlying mechanism are still unclear. Here, we found that L-theanine could suppress invasion, migration, and increase cell-cell adhesion of prostate cancer cells in vitro and in vivo. We also found that L-theanine could inhibit the epithelial-mesenchymal transition process in PCa. Our study revealed that L-theanine could downregulate MMP9, N-cadherin, Vimentin, Snail, and upregulate E-cadherin. Furthermore, L-theanine suppressed the transcription of MMP9 and Snail by significantly inhibiting the ERK/NF-κB signaling pathway and the binding activity of p65 to the promoter regions of MMP9 and Snail. All of these findings suggest that L-theanine has therapeutic potential for metastatic PCa and may be considered a promising candidate for antimetastatic therapy of prostate cancer.
Collapse
Affiliation(s)
- Xirui Fan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaowen Bi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Juanjuan Liang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Shuai Lu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Xintong Yan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, People's Republic of China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
47
|
Carceles-Cordon M, Kelly WK, Gomella L, Knudsen KE, Rodriguez-Bravo V, Domingo-Domenech J. Cellular rewiring in lethal prostate cancer: the architect of drug resistance. Nat Rev Urol 2020; 17:292-307. [PMID: 32203305 PMCID: PMC7218925 DOI: 10.1038/s41585-020-0298-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Over the past 5 years, the advent of combination therapeutic strategies has substantially reshaped the clinical management of patients with advanced prostate cancer. However, most of these combination regimens were developed empirically and, despite offering survival benefits, are not enough to halt disease progression. Thus, the development of effective therapeutic strategies that target the mechanisms involved in the acquisition of drug resistance and improve clinical trial design are an unmet clinical need. In this context, we hypothesize that the tumour engineers a dynamic response through the process of cellular rewiring, in which it adapts to the therapy used and develops mechanisms of drug resistance via downstream signalling of key regulatory cascades such as the androgen receptor, PI3K-AKT or GATA2-dependent pathways, as well as initiation of biological processes to revert tumour cells to undifferentiated aggressive states via phenotype switching towards a neuroendocrine phenotype or acquisition of stem-like properties. These dynamic responses are specific for each patient and could be responsible for treatment failure despite multi-target approaches. Understanding the common stages of these cellular rewiring mechanisms to gain a new perspective on the molecular underpinnings of drug resistance might help formulate novel combination therapeutic regimens.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - W Kevin Kelly
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard Gomella
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karen E Knudsen
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Urology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Veronica Rodriguez-Bravo
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Josep Domingo-Domenech
- Medical Oncology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Cancer Biology Department, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
48
|
de Araujo Junior RF, Eich C, Jorquera C, Schomann T, Baldazzi F, Chan AB, Cruz LJ. Ceramide and palmitic acid inhibit macrophage-mediated epithelial-mesenchymal transition in colorectal cancer. Mol Cell Biochem 2020; 468:153-168. [PMID: 32222879 PMCID: PMC7145792 DOI: 10.1007/s11010-020-03719-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/13/2020] [Indexed: 02/06/2023]
Abstract
Accumulating evidence indicates that ceramide (Cer) and palmitic acid (PA) possess the ability to modulate switching of macrophage phenotypes and possess anti-tumorigenic effects; however, the underlying molecular mechanisms are largely unknown. The aim of the present study was to investigate whether Cer and PA could induce switching of macrophage polarization from the tumorigenic M2- towards the pro-inflammatory M1-phenotype, and whether this consequently altered the potential of colorectal cancer cells to undergo epithelial–mesenchymal transition (EMT), a hallmark of tumor progression. Our study showed that Cer- and PA-treated macrophages increased expression of the macrophage 1 (M1)-marker CD68 and secretion of IL-12 and attenuated expression of the macrophage 2 (M2)-marker CD163 and IL-10 secretion. Moreover, Cer and PA abolished M2 macrophage-induced EMT and migration of colorectal cancer cells. At the molecular level, this coincided with inhibition of SNAI1 and vimentin expression and upregulation of E-cadherin. Furthermore, Cer and PA attenuated expression levels of IL-10 in colorectal cancer cells co-cultured with M2 macrophages and downregulated STAT3 and NF-κB expression. For the first time, our findings suggest the presence of an IL-10-STAT3-NF-κB signaling axis in colorectal cancer cells co-cultured with M2 macrophages, mimicking the tumor microenvironment. Importantly, PA and Cer were powerful inhibitors of this signaling axis and, consequently, EMT of colorectal cancer cells. These results contribute to our understanding of the immunological mechanisms that underlie the anti-tumorigenic effects of lipids for future combination with drugs in the therapy of colorectal carcinoma.
Collapse
Affiliation(s)
- Raimundo Fernandes de Araujo Junior
- Department of Morphology, Federal University of Rio Grande do Norte, Natal, RN, 59072-970, Brazil. .,Post-Graduation Programme in Structural and Functional Biology, Federal University of Rio Grande do Norte, Natal, RN, 59072-970, Brazil. .,Post-Graduation Programme in Health Science, Federal University of Rio Grande do Norte, Natal, RN, 59072-970, Brazil. .,Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
| | - Christina Eich
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Carla Jorquera
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Timo Schomann
- Percuros B.V., 2333 CL, Leiden, The Netherlands.,Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Fabio Baldazzi
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Alan B Chan
- Percuros B.V., 2333 CL, Leiden, The Netherlands.,Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
49
|
Rossini A, Giussani M, Ripamonti F, Aiello P, Regondi V, Balsari A, Triulzi T, Tagliabue E. Combined targeting of EGFR and HER2 against prostate cancer stem cells. Cancer Biol Ther 2020; 21:463-475. [PMID: 32089070 DOI: 10.1080/15384047.2020.1727702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Progression of prostate cancer has been associated with EGFR and HER2 activation and to tumor-initiating cells contribution toward chemotherapy resistance. We investigated the efficacy of a dual intervention against EGFR and HER2 to deplete the tumor-initiating cells, optimize the chemotherapy management and prevent the progression of castration-resistant prostate cancer (CRPC) cells. Using DU145, PC3, and 22Rv1 CRPC cell lines, biochemical analysis revealed activation of EGFR, HER2, MAPK, and STAT3 in DU145 and 22Rv1, and AKT and SRC in DU145 and PC-3. pSTAT3 nuclear staining was observed in DU145 xenografts and in 12 out of 14 CRPC specimens. The in vivo dual targeting of ErbB receptors with Cetuximab and Trastuzumab combined with chemotherapy caused an effective antitumor response in DU145 xenografted mice displaying STAT3 activation; conversely PC-3 bearing mice experienced tumor relapse. The potentiating of in vivo cytotoxic effect in DU145 model was accompanied by a significant decrease of prostatosphere-forming capacity assessed in vitro on residual tumor cells. Additionally, combined treatment in vitro with Cetuximab, Trastuzumab and chemotherapy negatively affected DU145 and 22Rv1 sphere formation, suggesting the critical function of ErbB receptors for tumor-initiating cells proliferation; no effect on PC-3 clonogenic potential was observed, indicating that other receptors than EGFR and HER2 may sustain PC3 tumor-initiating cells. These findings provided the preclinical evidence that the dual inhibition of EGFR and HER2 by targeting tumor-initiating cells may improve the efficacy of the current chemotherapy regimen, bringing benefits especially to castration-resistant patients with activated STAT3, and preventing disease progression.
Collapse
Affiliation(s)
- Anna Rossini
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marta Giussani
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Francesca Ripamonti
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Piera Aiello
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Andrea Balsari
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.,Dipartimento Di Scienze Biomediche per La Salute, Università Degli Studi Di Milano, Milan, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
50
|
Cytokines and Janus kinase/signal transducer and activator of transcription signaling in prostate cancer: overview and therapeutic opportunities. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.coemr.2020.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|