1
|
Luo Y, Deng X, Wei C, Liu Z, Song L, Han K, Li Y, Zhang J, Su S, Wang D. A novel bone metastasis-related gene signature for predicting prognosis, anti-androgen resistance, and drug choice in prostate cancer. J Bone Oncol 2025; 52:100673. [PMID: 40226044 PMCID: PMC11986555 DOI: 10.1016/j.jbo.2025.100673] [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: 09/27/2024] [Revised: 01/13/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Objective Prostate cancer (PCa) often metastasizes to the bone, posing a significant clinical challenge. This study aims to develop a bone metastasis-related risk model for PCa. Methods Bone metastasis-related genes (BMRGs) were identified through a combination of differential gene expression analysis and WGCNA using GSE32269 and GSE77930 datasets. Consensus clustering analysis was employed to determine the significance of these genes in molecular subtyping of PCa. LASSO-Cox regression analysis was utilized to construct the bone metastasis-related prognostic gene signature (BMRPS). The predictive performance of BMRPS was assessed using ROC curves, Kaplan-Meier survival curves, and a predictive nomogram. The immune landscape heterogeneity of subgroups was analyzed using CIBERSORT, ESTIMATE, and xCell algorithms. Drug sensitivity and molecular docking analysis were performed to identify drugs associated with BMRPS. Results Forty-four BMRGs associated with the prognosis of PCa were identified. Consensus clustering revealed the pivotal role of these genes in stratifying PCa into three distinct prognostic clusters. The BMRPS, consisting of 14 BMRGs, demonstrated excellent predictive accuracy for prognosis and served as an independent prognostic factor in PCa. BMRPS effectively predicted the overall survival of bone metastatic PCa and differentiated bone metastasis from other metastatic types. BMRPS showed a close correlation with the immune landscape and immunotherapeutic response biomarkers. Additionally, BMRPS was associated with anti-androgen resistance, and AZD8186 was identified as a potential BMRPS-related drug that holds promise for personalized treatment in PCa. Conclusion BMRPS facilitates the prediction of prognosis and resistance to anti-androgens in PCa. It also offers insights into the molecular mechanisms of bone metastasis and aids in drug selection for the treatment of PCa.
Collapse
Affiliation(s)
- Yu Luo
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoqi Deng
- Department of Nephrology, Zigong Fourth People’s Hospital, Zigong, Sichuan Province 643000, China
| | - Chengcheng Wei
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhangcheng Liu
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liangdong Song
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kun Han
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yunfan Li
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jindong Zhang
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuai Su
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Delin Wang
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
2
|
Meng C, Lin K, Shi W, Teng H, Wan X, DeBruine A, Wang Y, Liang X, Leo J, Chen F, Gu Q, Zhang J, Van V, Maldonado KL, Gan B, Ma L, Lu Y, Zhao D. Histone methyltransferase ASH1L primes metastases and metabolic reprogramming of macrophages in the bone niche. Nat Commun 2025; 16:4681. [PMID: 40394007 DOI: 10.1038/s41467-025-59381-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025] Open
Abstract
Bone metastasis is a major cause of cancer death; however, the epigenetic determinants driving this process remain elusive. Here, we report that histone methyltransferase ASH1L is genetically amplified and is required for bone metastasis in men with prostate cancer. ASH1L rewires histone methylations and cooperates with HIF-1α to induce pro-metastatic transcriptome in invading cancer cells, resulting in monocyte differentiation into lipid-associated macrophage (LA-TAM) and enhancing their pro-tumoral phenotype in the metastatic bone niche. We identified IGF-2 as a direct target of ASH1L/HIF-1α and mediates LA-TAMs' differentiation and phenotypic changes by reprogramming oxidative phosphorylation. Pharmacologic inhibition of the ASH1L-HIF-1α-macrophages axis elicits robust anti-metastasis responses in preclinical models. Our study demonstrates epigenetic alterations in cancer cells reprogram metabolism and features of myeloid components, facilitating metastatic outgrowth. It establishes ASH1L as an epigenetic driver priming metastasis and macrophage plasticity in the bone niche, providing a bona fide therapeutic target in metastatic malignancies.
Collapse
Affiliation(s)
- Chenling Meng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Shi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hongqi Teng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xinhai Wan
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anna DeBruine
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yin Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xin Liang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Javier Leo
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Feiyu Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qianlin Gu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vivien Van
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kiersten L Maldonado
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Di Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Moghbeli M. MicroRNAs as the critical regulators of bone metastasis during prostate tumor progression. Int J Biol Macromol 2025; 309:142912. [PMID: 40203904 DOI: 10.1016/j.ijbiomac.2025.142912] [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: 03/01/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Prostate cancer (PCa) is the most prevalent cancer among men globally. Although, there are various therapeutic methods for the localized or advanced cancers, there is still a high rate of mortality among PCa patients that is mainly associated with bone metastasis in advanced tumors. There are few options available for treating bone metastasis in PCa, which only provide symptom relief without curing the disease. Therefore, it is crucial to evaluate the molecular mechanisms associated with bone metastasis of PCa cells to suggest the novel diagnostic and therapeutic approaches that could lower the morbidity and mortality rates in PCa patients. MicroRNAs (miRNAs) are involved in regulation of various pathophysiological processes such as tumor growth and osteoblasts/osteoclasts formation. Since, miRNA deregulation has been also frequently observed in PCa patients with bone metastasis, we discussed the role of miRNAs in bone metastasis during PCa progression. It has been reported that miRNAs mainly reduced the ability of PCa tumor cells for the bone metastasis through the regulation of WNT, NF-kB, PI3K/AKT, and TGF-β signaling pathways. They also affected the EMT process, transcription factors, and structural proteins to regulate the bone metastasis during PCa progression. This review paves the way to suggest the miRNAs as the reliable markers not only for the non-invasive early diagnosis, but also for the targeted therapy of PCa tumors with bone metastasis.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Wani SA, Qudrat S, Zubair H, Iqbal Z, Gulzar B, Aziz S, Inayat A, Safi D, Kamran A. Role of osteoclast inhibitors in prostate cancer bone metastasis; a narrative review. J Oncol Pharm Pract 2025; 31:119-127. [PMID: 39169855 DOI: 10.1177/10781552241275943] [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: 08/23/2024]
Abstract
OBJECTIVE To study the role of Osteoclast inhibitors in advanced prostate cancer metastasis treatment and their efficacy in reducing skeletal related events. METHODS: DATA SOURCE A comprehensive search was done using search terms as "osteoclast inhibitors" "Bisphosphonates" "Zoledronic acid" " pamidronate" " Alendronate" "Denosumab" " Prostate cancer metastasis" in pubmed and Google scholar. Relevant articles were screened and collected . The collected articles were used to frame the review and data showing use of Osteoclast inhibitors In prostate cancer bone metastasis was collected. DATA SUMMARY Prostate cancer metastasizes most commonly to the skeleton thus leading to significant morbidity ranging from pain, pathological fractures to spinal cord compression and are the primary cause of patient disability and reduced quality of life.Initially, radiation therapy and radiopharmaceuticals were the mainstay of treatment however the role of Bisphosphonates and denosumab has become an integral part of therapy to manage metastatic prostate cancer. These agents significantly decrease skeletal related events and enhance patients quality of life. Emerging therapies like Radium-223 have also shown promise in reducing skeletal related events and also improving survival rates in patients with bone metastasis. Other treatment options which are being used are systemic agents like Docetaxel, cabazitaxel, hormonal therapies like abiraterone and enzalutamide. Immunotherapy with sipuleucel-T has demonstrated a reduction in mortality among prostate cancer patients with metastasis, highlighting the need for further research in this area. Ongoing studies are investigating novel agents that target both tumor cells and the bone microenvironment. CONCLUSION Osteoclast inhibitors are effective in reducing skeletal related events in advanced bone metastasis and improve the quality of life of patients.
Collapse
Affiliation(s)
- Shariq Ahmad Wani
- Department of Internal Medicine, Government medical college, Srinagar, India
| | - Salma Qudrat
- internal Medicine, Khyber teaching hospital, Peshawar, Pakistan
| | - Hina Zubair
- Internal Medicine, Rawalpindi medical university, Rawalpindi, Pakistan
| | - Zahra Iqbal
- Internal Medicine, Virginia commonwealth university, Richmond, USA
| | - Babar Gulzar
- Department of Internal Medicine, Government medical college, Srinagar, India
| | - Sundal Aziz
- Internal Medicine, Khyber medical university, Peshawar, Pakistan
| | - Arsalan Inayat
- Internal Medicine, HSHS St Mary's Hospital, Decatur, Illinois, USA
| | - Danish Safi
- Internal Medicine, WVU hospital, Morgantown, WV, USA
| | - Amir Kamran
- Internal Medicine, Charleston Area Medical center, Charleston, USA
| |
Collapse
|
5
|
Farahani N, Alimohammadi M, Raei M, Nabavi N, Aref AR, Hushmandi K, Daneshi S, Razzaghi A, Taheriazam A, Hashemi M. Exploring the dual role of endoplasmic reticulum stress in urological cancers: Implications for tumor progression and cell death interactions. J Cell Commun Signal 2024; 18:e12054. [PMID: 39691874 PMCID: PMC11647052 DOI: 10.1002/ccs3.12054] [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: 05/07/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 12/19/2024] Open
Abstract
The endoplasmic reticulum (ER) is crucial for maintaining calcium balance, lipid biosynthesis, and protein folding. Disruptions in ER homeostasis, often due to the accumulation of misfolded or unfolded proteins, lead to ER stress, which plays a significant role in various diseases, especially cancer. Urological cancers, which account for high male mortality worldwide, pose a persistent challenge due to their incurability and tendency to develop drug resistance. Among the numerous dysregulated biological mechanisms, ER stress is a key factor in the progression and treatment response of these cancers. This review highlights the dual role of aberrant ER stress activation in urologic cancers, affecting both tumor growth and therapeutic outcomes. While ER stress can support tumor growth through pro-survival autophagy, it primarily inhibits cancer progression via apoptosis and pro-death autophagy. Interestingly, ER stress can paradoxically aid cancer progression through mechanisms such as exosome-mediated immune evasion. Additionally, the review examines how pharmacological interventions, particularly with phytochemicals, can stimulate ER stress-mediated tumor suppression. Key regulators, including PERK, IRE1α, and ATF6, are discussed for their roles in upregulating CHOP levels and triggering apoptosis. In conclusion, a deeper understanding of ER stress in urological cancers not only clarifies the complex interactions between cellular stress and cancer progression but also provides new opportunities for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mina Alimohammadi
- Department of ImmunologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Mehdi Raei
- Health Research CenterLife Style InstituteBaqiyatallah University of Medical SciencesTehranIran
| | | | - Amir Reza Aref
- Department of SurgeryMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kiavash Hushmandi
- Nephrology and Urology Research CenterClinical Sciences InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - Salman Daneshi
- Department of Public HealthSchool of HealthJiroft University of Medical SciencesJiroftIran
| | - Alireza Razzaghi
- Social Determinants of Health Research CenterResearch Institute for Prevention of Non‐Communicable DiseasesQazvin University of Medical SciencesQazvinIran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of OrthopedicsFaculty of MedicineTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| |
Collapse
|
6
|
Mao W, Zhang H, Wang K, Geng J, Wu J. Research progress of MUC1 in genitourinary cancers. Cell Mol Biol Lett 2024; 29:135. [PMID: 39491020 PMCID: PMC11533421 DOI: 10.1186/s11658-024-00654-x] [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: 06/09/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
MUC1 is a highly glycosylated transmembrane protein with a high molecular weight. It plays a role in lubricating and protecting mucosal epithelium, participates in epithelial cell renewal and differentiation, and regulates cell adhesion, signal transduction, and immune response. MUC1 is expressed in both normal and malignant epithelial cells, and plays an important role in the diagnosis, prognosis prediction and clinical monitoring of a variety of tumors and is expected to be a new therapeutic target. This article reviews the structural features, expression regulation mechanism, and research progress of MUC1 in the development of genitourinary cancers and its clinical applications.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| | - Houliang Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Keyi Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Jiang Geng
- Department of Urology, Bengbu First People's Hospital, Bengbu, People's Republic of China.
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.
| | - Jianping Wu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
7
|
Wei X, Liang M, Deng M, Zheng J, Luo F, Ma Q. A switch from lysosomal degradation to secretory autophagy initiates osteogenic bone metastasis in prostate cancer. J Extracell Vesicles 2024; 13:e70002. [PMID: 39497621 PMCID: PMC11535520 DOI: 10.1002/jev2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 07/27/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
The identification of both autophagy-related material degradation and unconventional secretion has paved the way for significant breakthroughs linking autophagy to a plethora of physiological processes and disease conditions. However, the mechanisms that coordinate these two pathways remain elusive. Here, we demonstrate that a switch from the lysosomal degradation to a secretory autophagy pathway is governed by protein tyrosine phosphatase 1B (PTP1B, encoded by PTPN1). Dephosphorylation at two tyrosine residues of syntaxin17 (STX17) by PTP1B reduces autophagosome-lysosome fusion while switching the cells to a secretory autophagy pathway. Both PTP1B overexpression and tumour-derived extracellular vesicles (EVs) can activate the secretory autophagy pathway in osteoblasts. Moreover, we demonstrate that osteoblastic LC3+ EVs, generated via the secretory autophagy pathway, are the primary contributor to tumour-associated bone remodelling in prostate cancer. Depletion of tumour-derived EVs secretion or genetic ablation of osteoblastic PTP1B rescues aberrant bone remodelling and lesions, highlighting the relevance between LC3+ EVs and the formation of bone metastatic niche. Our results reveal the significance of tumour-regulated PTP1B in the fate decision of autophagosomes, and propose a role ofLC3+ EVs in shaping the bone metastatic niche.
Collapse
Affiliation(s)
- Xiaoyu Wei
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
| | - Mengmeng Liang
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
- Institute of Environment and Operational MedicineAcademy of Military Medicine Sciences, Academy of Military SciencesTianjinChina
| | - Min Deng
- Department of UrologyXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Ji Zheng
- Department of UrologyXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Fei Luo
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
| | - Qinyu Ma
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
- Institute of CancerXinqiao Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of ImmunotherapyChongqingChina
- Shigatse Branch, Xinqiao HospitalThird Military Medical UniversityShigatseChina
| |
Collapse
|
8
|
Dufrancais O, Verdys P, Plozza M, Métais A, Juzans M, Sanchez T, Bergert M, Halper J, Panebianco CJ, Mascarau R, Gence R, Arnaud G, Neji MB, Maridonneau-Parini I, Cabec VL, Boerckel JD, Pavlos NJ, Diz-Muñoz A, Lagarrigue F, Blin-Wakkach C, Carréno S, Poincloux R, Burkhardt JK, Raynaud-Messina B, Vérollet C. Moesin controls cell-cell fusion and osteoclast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593799. [PMID: 38798563 PMCID: PMC11118517 DOI: 10.1101/2024.05.13.593799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Cell-cell fusion is an evolutionarily conserved process that is essential for many functions, including fertilisation and the formation of placenta, muscle and osteoclasts, multinucleated cells that are unique in their ability to resorb bone. The mechanisms of osteoclast multinucleation involve dynamic interactions between the actin cytoskeleton and the plasma membrane that are still poorly characterized. Here, we found that moesin, a cytoskeletal linker protein member of the Ezrin/Radixin/Moesin (ERM) protein family, is activated during osteoclast maturation and plays an instrumental role in both osteoclast fusion and function. In mouse and human osteoclast precursors, moesin inhibition favors their ability to fuse into multinucleated osteoclasts. Accordingly, we demonstrated that moesin depletion decreases membrane-to-cortex attachment and enhances the formation of tunneling nanotubes (TNTs), F-actin-based intercellular bridges that we reveal here to trigger cell-cell fusion. Moesin also controls HIV-1- and inflammation-induced cell fusion. In addition, moesin regulates the formation of the sealing zone, the adhesive structure determining osteoclast bone resorption area, and thus controls bone degradation, via a β3-integrin/RhoA/SLK pathway. Supporting our results, moesin - deficient mice present a reduced density of trabecular bones and increased osteoclast abundance and activity. These findings provide a better understanding of the regulation of cell-cell fusion and osteoclast biology, opening new opportunities to specifically target osteoclast activity in bone disease therapy.
Collapse
|
9
|
Jin L, Zhu J, Yao L, Shen G, Xue BX, Tao W. Targeting SphK1/2 by SKI-178 inhibits prostate cancer cell growth. Cell Death Dis 2023; 14:537. [PMID: 37604912 PMCID: PMC10442381 DOI: 10.1038/s41419-023-06023-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/23/2023]
Abstract
Sphingosine kinases (SphK), including SphK1 and SphK2, are important enzymes promoting progression of prostate cancer. SKI-178 is a novel and highly potent SphK1/2 dual inhibitor. We here tested the potential anti-prostate cancer cell activity of SKI-178. Bioinformatics analyses and results from local tissues demonstrated that that both SphK1 and SphK2 are upregulated in human prostate cancer tissues. Ectopic overexpression of SphK1 and SphK2, by lentiviral constructs, promoted primary prostate cancer cell proliferation and migration. In primary human prostate cancer cells and immortalized cell lines, SKI-178 potently inhibited cell viability, proliferation, cell cycle progression and cell migration, causing robust cell death and apoptosis. SKI-178 impaired mitochondrial functions, causing mitochondrial depolarization, reactive oxygen species production and ATP depletion.SKI-178 potently inhibited SphK activity and induced ceramide production, without affecting SphK1/2 expression in prostate cancer cells. Further, SKI-178 inhibited Akt-mTOR activation and induced JNK activation in prostate cancer cells. Contrarily, a constitutively-active Akt1 construct or the pharmacological JNK inhibitors attenuated SKI-178-induced cytotoxicity in prostate cancer cells. In vivo, daily intraperitoneal injection of a single dose of SKI-178 potently inhibited PC-3 xenograft growth in nude mice. SphK inhibition, ceramide production, ATP depletion and lipid peroxidation as well as Akt-mTOR inactivation and JNK activation were detected in PC-3 xenograft tissues with SKI-178 administration. Together, targeting SphK1/2 by SKI-178 potently inhibited prostate cancer cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Lu Jin
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhu
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Linya Yao
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Yangzhou University, Kunshan, China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China.
| | - Bo-Xin Xue
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wei Tao
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
10
|
Li XF, Selli C, Zhou HL, Cao J, Wu S, Ma RY, Lu Y, Zhang CB, Xun B, Lam AD, Pang XC, Fernando A, Zhang Z, Unciti-Broceta A, Carragher NO, Ramachandran P, Henderson NC, Sun LL, Hu HY, Li GB, Sawyers C, Qian BZ. Macrophages promote anti-androgen resistance in prostate cancer bone disease. J Exp Med 2023; 220:213858. [PMID: 36749798 PMCID: PMC9948761 DOI: 10.1084/jem.20221007] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 11/14/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
Metastatic castration-resistant prostate cancer (PC) is the final stage of PC that acquires resistance to androgen deprivation therapies (ADT). Despite progresses in understanding of disease mechanisms, the specific contribution of the metastatic microenvironment to ADT resistance remains largely unknown. The current study identified that the macrophage is the major microenvironmental component of bone-metastatic PC in patients. Using a novel in vivo model, we demonstrated that macrophages were critical for enzalutamide resistance through induction of a wound-healing-like response of ECM-receptor gene expression. Mechanistically, macrophages drove resistance through cytokine activin A that induced fibronectin (FN1)-integrin alpha 5 (ITGA5)-tyrosine kinase Src (SRC) signaling cascade in PC cells. This novel mechanism was strongly supported by bioinformatics analysis of patient transcriptomics datasets. Furthermore, macrophage depletion or SRC inhibition using a novel specific inhibitor significantly inhibited resistant growth. Together, our findings elucidated a novel mechanism of macrophage-induced anti-androgen resistance of metastatic PC and a promising therapeutic approach to treat this deadly disease.
Collapse
Affiliation(s)
- Xue-Feng Li
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cigdem Selli
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Han-Lin Zhou
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
- BGI-Shenzhen, Shenzhen, China
- BGI-Henan, BGI-Shenzhen, Xinxiang, China
| | - Jian Cao
- Department of Urology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medicine School, Central South University, Changsha, China
| | - Shuiqing Wu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruo-Yu Ma
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Ye Lu
- BGI-Shenzhen, Shenzhen, China
- BGI-Henan, BGI-Shenzhen, Xinxiang, China
| | - Cheng-Bin Zhang
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Bijie Xun
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Alyson D. Lam
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Xiao-Cong Pang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Anu Fernando
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Zeda Zhang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Neil O. Carragher
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Prakash Ramachandran
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C. Henderson
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ling-Ling Sun
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hai-Yan Hu
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Gui-Bo Li
- BGI-Shenzhen, Shenzhen, China
- BGI-Henan, BGI-Shenzhen, Xinxiang, China
| | - Charles Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Charles Sawyers:
| | - Bin-Zhi Qian
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Correspondence to Bin-Zhi Qian:
| |
Collapse
|
11
|
Oya T, Ichikawa Y, Nakamura S, Tomita Y, Sasaki T, Inoue T, Sakuma H. Quantitative assessment of 99mTc-methylene diphosphonate bone SPECT/CT for assessing bone metastatic burden and its prognostic value in patients with castration-resistant prostate cancers: initial results in a single-center retrospective study. Ann Nucl Med 2023; 37:360-370. [PMID: 36947324 DOI: 10.1007/s12149-023-01833-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE To evaluate the prognostic value of the quantitative assessment of 99mTc-methylene diphosphonate (99mTc-MDP) bone SPECT/CT in castration-resistant prostate cancer (CRPC) patients with bone metastases. METHODS A total of 103 patients who underwent 99mTc-MDP bone SPECT/CT imaging from the neck to the proximal femur were included. First, in 65 patients without bone metastases, the normal range of standardized uptake value (SUV) of non-pathological bone was evaluated to determine an SUV threshold to reliably exclude most normal osseous activity. Then, in 38 CRPC patients with bone metastases, lesion uptake volume (LUV), which is the extracted volume of bone metastases exhibiting high accumulation above the SUV threshold, was calculated. The relation between LUV and prostate-related mortality was statistically evaluated. RESULTS Based on the SUV measurements of non-pathological bones, the optimal SUV threshold, which defines abnormal bone SPECT uptake, was determined to be 8. Median LUV was 39 mL (interquartile range 4.0-104.3 mL) in the CRPC subjects with bone metastases. Kaplan-Meier survival analysis showed a significant relation between prostate cancer-specific survival and LUV (cut-off value, 19.95 mL; P = 0.001). Multivariate analysis revealed LUV as an independent prognostic factor for the survival (P = 0.008, hazard ratio 23.424). Global chi-square test showed that LUV had significant incremental prognostic value in addition to prostate-specific antigen and the interval from progression to CRPC until bone SPECT/CT (P = 0.022). CONCLUSION Quantitative assessment of 99mTc-MDP bone SPECT images can provide valuable prognostic information in CRPC patients with bone metastases.
Collapse
Affiliation(s)
- Takashi Oya
- Department of Radiology, Mie University Hospital, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yasutaka Ichikawa
- Department of Radiology, Mie University Hospital, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Satsohi Nakamura
- Department of Radiology, Mie University Hospital, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yoya Tomita
- Department of Radiology, Mie University Hospital, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takeshi Sasaki
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Hajime Sakuma
- Department of Radiology, Mie University Hospital, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
12
|
Urabe F, Kosaka N, Yamamoto Y, Ito K, Otsuka K, Soekmadji C, Egawa S, Kimura T, Ochiya T. Metastatic prostate cancer-derived extracellular vesicles facilitate osteoclastogenesis by transferring the CDCP1 protein. J Extracell Vesicles 2023; 12:e12312. [PMID: 36880252 PMCID: PMC9989745 DOI: 10.1002/jev2.12312] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Bone metastases are still incurable and result in the development of clinical complications and decreased survival for prostate cancer patients. Recently, a number of studies have shown that extracellular vesicles (EVs) play important roles in tumour progression. Here, we show that EVs from metastatic prostate cancer cells promote osteoclast formation in the presence of receptor activator of NF-κB ligand (RANKL). EV characterization followed by functional siRNA screening identified CUB-domain containing protein 1 (CDCP1), a transmembrane protein, as an inducer of osteoclastogenesis. Additionally, CDCP1 expression on plasma-derived EVs was upregulated in bone metastatic prostate cancer patients. Our findings elucidate the effect of EVs from metastatic prostate cancer cells on osteoclast formation, which is promoted by CDCP1 located on EVs. Furthermore, our data suggested that CDCP1 expression on EVs might be useful to detect bone metastasis of prostate cancer.
Collapse
Affiliation(s)
- Fumihiko Urabe
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Nobuyoshi Kosaka
- Department of Molecular and Cellular MedicineInstitute of Medical Science, Tokyo Medical UniversityTokyoJapan
| | - Yusuke Yamamoto
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Kagenori Ito
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Kurataka Otsuka
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
- Department of Molecular and Cellular MedicineInstitute of Medical Science, Tokyo Medical UniversityTokyoJapan
- R&D Division, Kewpie Corporation Sengawa KewportTokyoJapan
| | - Carolina Soekmadji
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | - Shin Egawa
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
| | - Takahiro Kimura
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineInstitute of Medical Science, Tokyo Medical UniversityTokyoJapan
| |
Collapse
|
13
|
Nätkin R, Pennanen P, Syvälä H, Bläuer M, Kesseli J, Tammela TLJ, Nykter M, Murtola TJ. Adaptive and non-adaptive gene expression responses in prostate cancer during androgen deprivation. PLoS One 2023; 18:e0281645. [PMID: 36809527 PMCID: PMC9942993 DOI: 10.1371/journal.pone.0281645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
Androgen deprivation therapy is the cornerstone treatment of advanced prostate cancer. Eventually prostate cancer cells overcome androgen deprivation therapy, giving rise to castration resistant prostate cancer (CRPC) characterized by increased androgen receptor (AR) activity. Understanding the cellular mechanisms leading to CRPC is needed for development of novel treatments. We used long-term cell cultures to model CRPC; a testosterone-dependent cell line (VCaP-T) and cell line adapted to grow in low testosterone (VCaP-CT). These were used to uncover persistent and adaptive responses to testosterone level. RNA was sequenced to study AR-regulated genes. Expression level changed due to testosterone depletion in 418 genes in VCaP-T (AR-associated genes). To evaluate significance for CRPC growth, we compared which of them were adaptive i.e., restored expression level in VCaP-CT. Adaptive genes were enriched to steroid metabolism, immune response and lipid metabolism. The Cancer Genome Atlas Prostate Adenocarcinoma data were used to assess the association with cancer aggressiveness and progression-free survival. Expressions of 47 AR-associated or association gaining genes were statistically significant markers for progression-free survival. These included genes related to immune response, adhesion and transport. Taken together, we identified and clinically validated multiple genes being linked with progression of prostate cancer and propose several novel risk genes. Possible use as biomarkers or therapeutic targets should be studied further.
Collapse
Affiliation(s)
- Reetta Nätkin
- Faculty of Medicine and Health Technology, Prostate Cancer Research Center, Tampere University and Tays Cancer Center, Tampere, Finland
- * E-mail: (RN); (TJM)
| | - Pasi Pennanen
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Heimo Syvälä
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Merja Bläuer
- Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere Pancreas Laboratory and Department of Gastroenterology and Alimentary Tract Surgery, Tampere University, Tampere, Finland
| | - Juha Kesseli
- Faculty of Medicine and Health Technology, Prostate Cancer Research Center, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Teuvo L. J. Tammela
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Urology, Tays Cancer Center, Tampere, Finland
| | - Matti Nykter
- Faculty of Medicine and Health Technology, Prostate Cancer Research Center, Tampere University and Tays Cancer Center, Tampere, Finland
| | - Teemu J. Murtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Urology, Tays Cancer Center, Tampere, Finland
- * E-mail: (RN); (TJM)
| |
Collapse
|
14
|
Zhu L, Shi H, Wei H, Wang C, Shi S, Zhang F, Yan R, Liu Y, He T, Wang L, Cheng J, Duan H, Du H, Meng F, Zhao W, Gu X, Guo L, Ni Y, He Y, Guan T, Han A. An accurate prediction of the origin for bone metastatic cancer using deep learning on digital pathological images. EBioMedicine 2022; 87:104426. [PMID: 36577348 PMCID: PMC9803701 DOI: 10.1016/j.ebiom.2022.104426] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Determining the origin of bone metastatic cancer (OBMC) is of great significance to clinical therapeutics. It is challenging for pathologists to determine the OBMC with limited clinical information and bone biopsy. METHODS We designed a regional multiple-instance learning algorithm to predict the OBMC based on hematoxylin-eosin (H&E) staining slides alone. We collected 1041 cases from eight different hospitals and labeled 26,431 regions of interest to train the model. The performance of the model was assessed by ten-fold cross validation and external validation. Under the guidance of top3 predictions, we conducted an IHC test on 175 cases of unknown origins to compare the consistency of the results predicted by the model and indicated by the IHC markers. We also applied the model to identify whether there was tumor or not in a region, as well as distinguishing squamous cell carcinoma, adenocarcinoma, and neuroendocrine tumor. FINDINGS In the within-cohort, our model achieved a top1-accuracy of 91.35% and a top3-accuracy of 97.75%. In the external cohort, our model displayed a good generalizability with a top3-accuracy of 97.44%. The top1 consistency between the results of the model and the immunohistochemistry markers was 83.90% and the top3 consistency was 94.33%. The model obtained an accuracy of 98.98% to identify whether there was tumor or not and an accuracy of 93.85% to differentiate three types of cancers. INTERPRETATION Our model demonstrated good performance to predict the OBMC from routine histology and had great potential for assisting pathologists with determining the OBMC accurately. FUNDING National Science Foundation of China (61875102 and 61975089), Natural Science Foundation of Guangdong province (2021A15-15012379 and 2022A1515 012550), Science and Technology Research Program of Shenzhen City (JCYJ20200109110606054 and WDZC20200821141349001), and Tsinghua University Spring Breeze Fund (2020Z99CFZ023).
Collapse
Affiliation(s)
- Lianghui Zhu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huiting Wei
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengjiang Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Shanshan Shi
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Fenfen Zhang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Renao Yan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Yiqing Liu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Tingting He
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Liyuan Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junru Cheng
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Hufei Duan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Hong Du
- Department of Pathology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Fengjiao Meng
- Department of Pathology, Zhongshan People's Hospital, Zhongshan, China
| | - Wenli Zhao
- Department of Pathology, The First People's Hospital of Huizhou, Huizhou, China
| | - Xia Gu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingpeng Ni
- Department of Pathology, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-Sen University), Jieyang, China
| | - Yonghong He
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China,Corresponding author.
| | - Tian Guan
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China,Corresponding author.
| | - Anjia Han
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China,Corresponding author.
| |
Collapse
|
15
|
Dong Q, Wu X, Gan W, Mok TN, Shen J, Zha Z, Chen J. Construction and validation of web-based nomograms for detecting and prognosticating in prostate adenocarcinoma with bone metastasis. Sci Rep 2022; 12:18623. [PMID: 36329203 PMCID: PMC9633700 DOI: 10.1038/s41598-022-23275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Bone metastasis (BM) is one of the most common sites of metastasis in prostate adenocarcinoma (PA). PA with BM can significantly diminish patients' quality of life and result in a poor prognosis. The objective of this study was to establish two web-based nomograms to estimate the risk and prognosis of BM in PA patients. From the Surveillance, Epidemiology, and End Results (SEER) database, data on 308,332 patients diagnosed with PA were retrieved retrospectively. Logistic and Cox regression, respectively, were used to determine independent risk and prognostic factors. Then, We constructed two web-based nomograms and the results were validated by receiver operating characteristic (ROC) curves, calibration curves, decision curve analysis (DCA) , and the Kaplan-Meier analyses. The independent risk factors for BM in PA patients included race, PSA, ISUP, T stage, N stage, brain, liver, lung metastasis, surgery, radiation and chemotherapy. The independent prognostic predictors for overall survival (OS) were age, marital status, PSA, ISUP and liver metastasis. Both nomograms could effectively predict risk and prognosis of BM in PA patients according to the results of ROC curves, calibration, and DCA in the training and validation sets. And the Kaplan-Meier analysis illustrated that the prognostic nomogram could significantly distinguish the population with different survival risks. We successfully constructed the two web-based nomograms for predicting the incidence of BM and the prognosis of PA patients with BM, which may assist clinicians in optimizing the establishment of individualized treatment programs and enhancing patients' prognoses.
Collapse
Affiliation(s)
- Qiu Dong
- Center for Bone, Joint and Sports Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China
| | - Xiaoting Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wenyi Gan
- Center for Bone, Joint and Sports Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China
| | - Tsz Ngai Mok
- Center for Bone, Joint and Sports Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhengang Zha
- Center for Bone, Joint and Sports Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China
| | - Junyuan Chen
- Center for Bone, Joint and Sports Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
16
|
Dai R, Liu M, Xiang X, Xi Z, Xu H. Osteoblasts and osteoclasts: an important switch of tumour cell dormancy during bone metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:316. [PMID: 36307871 PMCID: PMC9615353 DOI: 10.1186/s13046-022-02520-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
Abstract
Bone metastasis occurs when tumour cells dissociate from primary tumours, enter the circulation (circulating tumour cells, CTCs), and colonize sites in bone (disseminated tumour cells, DTCs). The bone marrow seems to be a particularly dormancy-inducing environment for DTCs, yet the mechanisms of dormancy initiation, reactivation, and interaction within the bone marrow have to be elucidated. Intriguingly, some evidence has suggested that dormancy is a reversible state that is switched 'on' or 'off' depending on the presence of various bone marrow resident cells, particularly osteoclasts and osteoblasts. It has become clear that these two cells contribute to regulating dormant tumour cells in bone both directly (interaction) and indirectly (secreted factors). The involved mechanisms include TGFβ signalling, the Wnt signalling axis, the Notch2 pathway, etc. There is no detailed review that specifically focuses on ascertaining the dynamic interactions between tumour cell dormancy and bone remodelling. In addition, we highlighted the roles of inflammatory cytokines during this 'cell-to-cell' communication. We also discussed the potential clinical relevance of remodelling the bone marrow niche in controlling dormant tumour cells. Understanding the unique role of osteoclasts and osteoblasts in regulating tumour dormancy in bone marrow will provide new insight into preventing and treating tumour bone metastasis.
Collapse
Affiliation(s)
- Rongchen Dai
- grid.412540.60000 0001 2372 7462School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Mengfan Liu
- grid.412540.60000 0001 2372 7462School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Xincheng Xiang
- grid.47840.3f0000 0001 2181 7878Rausser College of Natural Resources, University of California Berkeley, Berkeley, CA 94720 USA
| | - Zhichao Xi
- grid.412540.60000 0001 2372 7462School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203 China
| | - Hongxi Xu
- grid.412585.f0000 0004 0604 8558Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
17
|
HIF1α lactylation enhances KIAA1199 transcription to promote angiogenesis and vasculogenic mimicry in prostate cancer. Int J Biol Macromol 2022; 222:2225-2243. [DOI: 10.1016/j.ijbiomac.2022.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
|
18
|
Wu YH, Gugala Z, Barry MM, Shen Y, Dasgupta S, Wang H. Optimization and Characterization of a Bone Culture Model to Study Prostate Cancer Bone Metastasis. Mol Cancer Ther 2022; 21:1360-1368. [PMID: 35666809 PMCID: PMC9357208 DOI: 10.1158/1535-7163.mct-21-0684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/29/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023]
Abstract
Nearly 90% of patients with advanced prostate cancer manifest bone metastases. Distinct from the osteolytic metastasis mostly observed in other cancer types, prostate cancer bone metastasis is typically more osteoblastic, which is relatively understudied due to the lack of reliable and efficient models to resemble the indolent cellular growth and complexity of metastatic progression. In our previous studies, we developed bone-in-culture array (BICA) to primarily model the osteoblast-involved, pre-osteolytic stage of breast cancer bone metastasis. Given that the progression of prostate cancer bone metastasis is largely osteoblastic, it is reasonable to speculate that the original BICA model can be adjusted to investigate prostate cancer bone metastases. In this study, we refined BICA by reducing the surgical labor and improving its reproducibility and capacity. The optimized BICA can successfully recapitulate important features of prostate cancer bone metastasis such as the osteoblastic phenotype, indolent growth, cancer-niche interactions, and response to hormones. Our efforts address the long-standing need for reliable and efficient models to study prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Zbigniew Gugala
- Department of Orthopaedic Surgery & Rehabilitation, University of Texas Medical Branch, Galveston, TX 77555
| | - Megan M. Barry
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030
| | - Subhamoy Dasgupta
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263
| | - Hai Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263
| |
Collapse
|
19
|
Yamaguchi M, Murata T, Ramos JW. Extracellular Regucalcin Suppresses the Growth, Migration, Invasion and Adhesion of Metastatic Human Prostate Cancer Cells. Oncology 2022; 100:399-412. [PMID: 35340010 DOI: 10.1159/000524303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/11/2022] [Indexed: 11/19/2022]
Abstract
Regucalcin plays a multifunctional role in the regulation of cellular function including metabolism, signaling process and transcriptional activity in maintaining cell homeostasis. Downregulated expression or activity of regucalcin contributes to the development of malignancies in various types of human cancer. Survival of cancer patients, including metastatic prostate cancer, is prolonged with high expression of regucalcin in the tumor tissues. Furthermore, we elucidate whether extracellular regucalcin conquers the growth, migration, invasion and adhesion of metastatic human prostate cancer PC-3 and DU-145 cells. Extracellular regucalcin (0.1, 1, and 10 nM) of physiologic levels inhibited colony formation and growth of PC-3 and DU-145 cells, while it did not have an effect on cell death. Repressive effects of extracellular regucalcin on the proliferation were not exhibited by the presence of inhibitors of cell cycle, intracellular signaling process and transcriptional activity, suggesting that the signals of extracellular regucalcin are transmitted to block cell growth. Furthermore, extracellular regucalcin (0.1, 1, or 10 nM) inhibited migration, invasion and adhesion of PC-3 and DU-145 cells. Mechanistically, extracellular regucalcin (10 nM) decreased the levels of various signaling proteins including Ras, hosphatidylinositol-3 kinase, mitogen-activated protein kinase, mTOR, RSK-2, caveolin-1 and integrin β1 in PC-3 cells. Thus, extracellular regucalcin may play a suppressive role in growth, migration, invasion and adhesion, which are involved in metastatic activity of human prostate cancer cells, via affecting diverse signaling processes. This study may provide a new strategy in preventing metastatic prostate cancer with exogenous regucalcin.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Tomiyasu Murata
- Laboratory of Molecular Biology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
20
|
Saganich C, Zgaljardic M. Radium-223 Dichloride in Peritoneal Dialysate Following Treatment of Metastatic Castration-resistant Prostate Cancer. HEALTH PHYSICS 2022; 122:433-439. [PMID: 34999662 DOI: 10.1097/hp.0000000000001518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
ABSTRACT Radium-223 dichloride was administrated to a patient with castration-resistant metastatic prostate cancer (mCRPC) undergoing in-home peritoneal dialysis. The peritoneal dialysate and tubing sets were collected following four treatment cycles and analyzed for removable contamination, dose rate, and radioactivity. The purpose of the study was to assess radiation safety concerns and compare data with patients having normal renal function. Sixty-two liters of dialysate in 22 samples were collected over 4 mo. The mean surface dose rate of the dialysate bags was 0.16 μSv h-1 (range 0.11-0.23 μSv h-1). The highest measured removable surface activity was 150 dpm per 100 cm2. The mean cumulative percent of administered activity was 1.7% (range 1.4-1.9%). The mean concentration of 223Ra in dialysate was 4.0 kBq L-1 (range 0.56-14.1 kBq L-1). There was no association between the measured 223Ra activity in dialysate with peritoneal dwell time or the number of exchanges following administration. The measurement of the 211Bi and 223Ra ratio in the dialysate compared with a standard showed an increase of 23% by 40 h post administration. The data presented suggest that 223Ra dialysate can be safely managed in the home without risk of radiation dose or contamination if minimal precautions are taken. Patients with normal renal function have been shown to excrete up to six times more 223Ra in urine compared to those undergoing hemodialysis or peritoneal dialysis. A potential consequence may be an increase in 223Ra activity transiting GI tract leading to intestinal effects.
Collapse
Affiliation(s)
- Christopher Saganich
- Medical Health Physics, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065
| | | |
Collapse
|
21
|
Hong H, Liang D, Liu Q, Wu G, Sun R, Liu J, Wang F, Wang F. Value of transrectal contrast-enhanced ultrasound with clinical indicators in the prediction of bone metastasis in prostate cancer. Quant Imaging Med Surg 2022; 12:1750-1761. [PMID: 35284288 PMCID: PMC8899971 DOI: 10.21037/qims-21-365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/29/2021] [Indexed: 11/09/2024]
Abstract
BACKGROUND Transrectal contrast-enhanced ultrasound is an examination that can be used to diagnose and characterize prostate cancer by displaying tissue blood perfusion. To explore the value of transrectal contrast-enhanced ultrasound combined with clinical factors in predicting prostate cancer bone metastasis. METHODS We retrospectively analyzed transrectal contrast-enhanced ultrasound examination data, imaging examination data [single-photon emission computed tomography (SPECT)/computed tomography (CT), CT, magnetic resonance imaging (MRI), and/or bone scan], clinical laboratory data, and pathological Gleason score of 163 patients with prostate cancer. They were randomly divided into the modeling and validation data sets. A model for predicting prostate cancer bone metastasis was established by logistic regression in the modeling data set. The differentiation, consistency, and benefits of the model were verified using the validation data set. A nomogram of the prediction model for bone metastasis of prostate cancer was drawn. RESULTS Among 163 patients with prostate cancer, 65 had bone metastasis. Total prostate-specific antigen, alkaline phosphatase, and the transrectal contrast-enhanced ultrasound parameter area under the curve were independently associated with prostate cancer bone metastasis, with OR values of 2.845, 2.839, and 1.004, respectively. The area under the receiver operating characteristic curve of the prostate cancer bone metastasis prediction model was 0.804. In the training set, using a cutoff of 0.659, sensitivity was 52.8%, and specificity was 95.7%. In the validation set, using a cutoff of 0.659, sensitivity was 58.6%, and specificity was 98.1%. The area under the curve of the validation set was 0.799. The Hosmer-Lemeshow goodness-of-fit test showed that the calibration ability of the validation set was not statistically different from the training set (P=0.136). The decision curve analysis showed that the model had high benefits. CONCLUSIONS The nomogram that includes the transrectal contrast-enhanced ultrasound parameter area under the curve and the clinical parameters total prostate-specific antigen, and alkaline phosphatase can be used to personalize the risk of prostate cancer bone metastases.
Collapse
Affiliation(s)
- Hua Hong
- Department of Ultrasonography, Inner Mongolia People’s Hospital, Hohhot, China
| | - Danyan Liang
- Department of Ultrasonography, Inner Mongolia People’s Hospital, Hohhot, China
| | - Qian Liu
- Department of Ultrasonography, Inner Mongolia People’s Hospital, Hohhot, China
| | - Guozhu Wu
- Department of Ultrasonography, Inner Mongolia People’s Hospital, Hohhot, China
| | - Ran Sun
- Department of Ultrasonography, Inner Mongolia People’s Hospital, Hohhot, China
| | - Juzhen Liu
- Department of Nuclear Medicine, Inner Mongolia People’s Hospital, Hohhot, China
| | - Feng Wang
- Department of Pathology, Inner Mongolia People’s Hospital, Hohhot, China
| | - Fang Wang
- Department of Ultrasonography, Inner Mongolia People’s Hospital, Hohhot, China
| |
Collapse
|
22
|
Satcher RL, Zhang XHF. Evolving cancer-niche interactions and therapeutic targets during bone metastasis. Nat Rev Cancer 2022; 22:85-101. [PMID: 34611349 DOI: 10.1038/s41568-021-00406-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
Many cancer types metastasize to bone. This propensity may be a product of genetic traits of the primary tumour in some cancers. Upon arrival, cancer cells establish interactions with various bone-resident cells during the process of colonization. These interactions, to a large degree, dictate cancer cell fates at multiple steps of the metastatic cascade, from single cells to overt metastases. The bone microenvironment may even influence cancer cells to subsequently spread to multiple other organs. Therefore, it is imperative to spatiotemporally delineate the evolving cancer-bone crosstalk during bone colonization. In this Review, we provide a summary of the bone microenvironment and its impact on bone metastasis. On the basis of the microscopic anatomy, we tentatively define a roadmap of the journey of cancer cells through bone relative to various microenvironment components, including the potential of bone to function as a launch pad for secondary metastasis. Finally, we examine common and distinct features of bone metastasis from various cancer types. Our goal is to stimulate future studies leading to the development of a broader scope of potent therapies.
Collapse
Affiliation(s)
- Robert L Satcher
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
23
|
Tian G, Hu K, Qiu S, Xie Y, Cao Y, Ni S, Zhang L. Exosomes derived from PC-3 cells suppress osteoclast differentiation by downregulating miR-148a and blocking the PI3K/AKT/mTOR pathway. Exp Ther Med 2021; 22:1304. [PMID: 34630659 PMCID: PMC8461599 DOI: 10.3892/etm.2021.10739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 05/04/2021] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer is a leading malignancy in men that can also disrupt the bone tissue balance. Among all urological cancers, prostate cancer is associated with the highest rate of bone metastases, which can greatly reduce a patient's quality of life. In recent years, cell-derived exosomes, which can contain a wide range of biologically active molecules, have been reported as a novel method of communication among individual cells. However, the specific role that exosomes serve in this disease has not been fully elucidated. The prostate cancer cell line PC-3 were applied in the present study, where its exosomes were isolated to explore their potential effects on osteoclast differentiation. Exosomes are extracellular vesicles secreted by cells. The size of exosomes is 30-150 nm. They have double membrane structure and saucer-like morphology. They contain rich contents (including nucleic acid, protein and lipid) and participate in molecular transmission between cells. The combined results of tartrate-resistant acid phosphatase staining (to identify osteoclasts obtained from human peripheral blood mononuclear cells), reverse transcription-quantitative PCR and western blotting showed that PC-3-derived exosomes attenuated osteoclast differentiation by downregulating marker genes associated with osteoclastic maturation, including V-maf musculoaponeurotic fibrosarcoma oncogene homolog B, matrix metalloproteinase 9 and integrin β3. microRNA (miR)-148a expression was also found to be downregulated in osteoclasts by PC-3-derived exosomes. In addition, the mTOR and AKT signaling pathways were blocked after exposure to these PC-3 cell-derived exosomes. Therefore, results from the present study suggest that miR-148a mimics may be a new therapeutic approach for the prevention of prostate cancer bone metastases.
Collapse
Affiliation(s)
- Gaoqiang Tian
- Department of Spine Surgery, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong 512025, P.R. China
| | - Konghe Hu
- Department of Spine Surgery, The Affiliated Yuebei People's Hospital of Shantou University Medical College, Shaoguan, Guangdong 512025, P.R. China
| | - Sujun Qiu
- Department of Orthopedics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yingming Xie
- Department of Orthopedics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Yanlin Cao
- Department of Orthopedics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Songjia Ni
- Department of Orthopedics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
- Correspondence to: Dr Songjia Ni, Department of Orthopedics, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong 510280, P.R. China
| | - Lifang Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Southern Medical University, Guangzhou, Guangdong 510250, P.R. China
| |
Collapse
|
24
|
Zhang G, Liu Y, Yang J, Wang H, Xing Z. Inhibition of circ_0081234 reduces prostate cancer tumor growth and metastasis via miR-1/MAP3K1 axis. J Gene Med 2021; 24:e3376. [PMID: 34191363 DOI: 10.1002/jgm.3376] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Circular RNAs (circRNAs) are crucial regulators in tumor occurrence and progression, and circRNAs are enriched and stable in exosomes. This study aimed to explore the role and potential mechanism of cancer-derived exosomal circ_0081234 in prostate cancer (PCa). METHODS Exosomes were extracted using the ExoQuick Precipitation Kit. The levels of circ_0081234, miR-1 and mitogen-activated protein kinase kinase kinase 1 (MAP 3K1) were examined using qRT-PCR or western blot. Cell migration and invasion were evaluated via transwell assay. The protein levels of N-cadherin, Vimentin and E-cadherin were detected by western blot. The interaction between miR-1 and circ_0081234 or MAP 3K1 was verified via dual-luciferase reporter assay and RNA pull-down assay. RESULTS Circ_0081234 level was increased in PC a tissues with spinal metastasis (SM) in comparison to primary PCa tissues without SM. Exosomal circ_0081234 promoted the migration, invasion and epithelial-mesenchymal transition (EMT) of PCa cells. Knockdown of circ_0081234 blocked PCa cell progression via regulating miR-1. In addition, miR-1 overexpression suppressed PCa cell progression by repressing MAP 3K1. Moreover, circ_0081234 increased MAP 3K1 level via sponging miR-1. Depletion of circ_0081234 inhibited tumor growth in vivo. CONCLUSION Exosomal circ_0081234 promoted migration, invasion and EMT of PCa cells by regulating the miR-1/MAP 3K1 axis.
Collapse
Affiliation(s)
- Guangyao Zhang
- Department of General Surgery, Huiji Hospital of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yibin Liu
- Department of Emergency Surgery, Huiji Hospital of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianhui Yang
- Department of General Surgery, Huiji Hospital of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiming Wang
- Department of General Surgery, Huiji Hospital of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhengwei Xing
- Medical office, Huiji Hospital of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Polavaram NS, Dutta S, Islam R, Bag AK, Roy S, Poitz D, Karnes J, Hofbauer LC, Kohli M, Costello BA, Jimenez R, Batra SK, Teply BA, Muders MH, Datta K. Tumor- and osteoclast-derived NRP2 in prostate cancer bone metastases. Bone Res 2021; 9:24. [PMID: 33990538 PMCID: PMC8121836 DOI: 10.1038/s41413-021-00136-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/22/2020] [Accepted: 11/26/2020] [Indexed: 01/13/2023] Open
Abstract
Understanding the role of neuropilin 2 (NRP2) in prostate cancer cells as well as in the bone microenvironment is pivotal in the development of an effective targeted therapy for the treatment of prostate cancer bone metastasis. We observed a significant upregulation of NRP2 in prostate cancer cells metastasized to bone. Here, we report that targeting NRP2 in cancer cells can enhance taxane-based chemotherapy with a better therapeutic outcome in bone metastasis, implicating NRP2 as a promising therapeutic target. Since, osteoclasts present in the tumor microenvironment express NRP2, we have investigated the potential effect of targeting NRP2 in osteoclasts. Our results revealed NRP2 negatively regulates osteoclast differentiation and function in the presence of prostate cancer cells that promotes mixed bone lesions. Our study further delineated the molecular mechanisms by which NRP2 regulates osteoclast function. Interestingly, depletion of NRP2 in osteoclasts in vivo showed a decrease in the overall prostate tumor burden in the bone. These results therefore indicate that targeting NRP2 in prostate cancer cells as well as in the osteoclastic compartment can be beneficial in the treatment of prostate cancer bone metastasis.
Collapse
Affiliation(s)
- Navatha Shree Polavaram
- Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ridwan Islam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Arup K Bag
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sohini Roy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Poitz
- Institute for Clinical Chemistry, University Hospital Dresden, Dresden, Germany
| | | | - Lorenz C Hofbauer
- Center for Healthy Aging and Bone Lab Dresden, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Manish Kohli
- School of Medicine, Division of Oncology, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | | | - Raffael Jimenez
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benjamin A Teply
- Internal Medicine, Division of Oncology & Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael H Muders
- Rudolf- Becker Laboratory for Prostate Cancer Research, Institute of Pathology, University of Bonn Medical Center, Bonn, Germany.
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
26
|
Huang YS, Mao JX, Zhang L, Guo HW, Yan C, Chen M. Antiprostate Cancer Activity of Ineupatolide Isolated from Carpesium cernuum L. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5515961. [PMID: 33996996 PMCID: PMC8105106 DOI: 10.1155/2021/5515961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/27/2021] [Accepted: 04/09/2021] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The aim of the study was to investigate the antiprostate cancer effects and mechanism of ineupatolide (T-21), a natural product isolated from the Compositae plant Carpesium cernuum L., on PC-3 human prostate cancer cells. METHODS The effect of T-21 on the proliferation of PC-3 cells was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, cell migration, and invasion experiments; the morphology of cell apoptosis was observed by Hoechst-propidium iodide staining; the effects of T-21 on PC-3 cell apoptosis and the cell cycle were evaluated by flow cytometry; and the effect of T-21 on the expression levels of phosphorylated protein kinase B (p-AKT), AKT, X-linked inhibitor of apoptosis protein (xlAP), procaspase-3, and poly (ADP-ribose) polymerase (PARP) in PC-3 cells was measured by western blotting. RESULTS T-21 significantly inhibited the proliferation of cells, and its half-maximal inhibitory concentrations at 12, 24, and 48 h were 38.46 ± 1.01, 24.63 ± 0.70, and 7.36 ± 0.58 μM, respectively. T-21 may promote cell apoptosis in a concentration-dependent manner and block the cell cycle in the G2 and S phases. In addition, T-21 significantly reduced the protein expression levels of p-AKT, AKT, xlAP, procaspase-3, and PARP. CONCLUSION T-21 exhibits antiproliferation effects on PC-3 cells by promoting apoptosis and arresting the cell cycle in the G2 and S phases. The possible mechanism underlying its potential therapeutic effects against prostate cancer is related to the AKT/xlAP pathway.
Collapse
Affiliation(s)
- Yuan-she Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
- Anshun College, Anshun Guizhou 561000, China
| | - Jing-xin Mao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lai Zhang
- Anshun College, Anshun Guizhou 561000, China
| | - Hong-wei Guo
- An Shun City People's Hospital, Anshun 561000, China
| | - Chen Yan
- An Shun City People's Hospital, Anshun 561000, China
| | - Min Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
27
|
Chen S, Wang Q, Eltit F, Guo Y, Cox M, Wang R. An Ammonia-Induced Calcium Phosphate Nanostructure: A Potential Assay for Studying Osteoporosis and Bone Metastasis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17207-17219. [PMID: 33845570 DOI: 10.1021/acsami.1c00495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Osteoclastic resorption of bones plays a central role in both osteoporosis and bone metastasis. A reliable in vitro assay that simulates osteoclastic resorption in vivo would significantly speed up the process of developing effective therapeutic solutions for those diseases. Here, we reported the development of a novel and robust nanostructured calcium phosphate coating with unique functions on the track-etched porous membrane by using an ammonia-induced mineralization (AiM) technique. The calcium phosphate coating uniformly covers one side of the PET membrane, enabling testing for osteoclastic resorption. The track-etched pores in the PET membrane allow calcium phosphate mineral pins to grow inside, which, on the one hand, enhances coating integration with a membrane substrate and, on the other hand, provides diffusion channels for delivering drugs from the lower chamber of a double-chamber cell culture system. The applications of the processed calcium phosphate coating were first demonstrated as a drug screening device by using alendronate, a widely used drug for osteoporosis. It was confirmed that the delivery of alendronate significantly decreased both the number of monocyte-differentiated osteoclasts and coating resorption. To demonstrate the application in studying bone metastasis, we delivered a PC3 prostate cancer-conditioned medium and confirmed that both the differentiation of monocytes into osteoclasts and the osteoclastic resorption of the calcium phosphate coating were significantly enhanced. This novel assay thus provides a new platform for studying osteoclastic activities and assessing drug efficacy in vitro.
Collapse
Affiliation(s)
- Sijia Chen
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Qiong Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Felipe Eltit
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Yubin Guo
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Michael Cox
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Rizhi Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
28
|
Fu C, Zhang Q, Wang A, Yang S, Jiang Y, Bai L, Wei Q. EWI-2 controls nucleocytoplasmic shuttling of EGFR signaling molecules and miRNA sorting in exosomes to inhibit prostate cancer cell metastasis. Mol Oncol 2021; 15:1543-1565. [PMID: 33605506 PMCID: PMC8096798 DOI: 10.1002/1878-0261.12930] [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: 10/30/2020] [Revised: 01/18/2021] [Accepted: 02/17/2021] [Indexed: 02/05/2023] Open
Abstract
Early and accurate diagnosis of prostate cancer (PCa) is extremely important, as metastatic PCa remains hard to treat. EWI-2, a member of the Ig protein subfamily, is known to inhibit PCa cell migration. In this study, we found that EWI-2 localized on both the cell membrane and exosomes regulates the distribution of miR-3934-5p between cells and exosomes. Interestingly, we observed that EWI-2 is localized not only on the plasma membrane but also on the nuclear envelope (nuclear membrane), where it regulates the nuclear translocation of signaling molecules and miRNA. Collectively, these functions of EWI-2 found in lipid bilayers appear to regulate PCa cell metastasis through the epidermal growth factor receptor-mitogen-activated protein kinase-extracellular-signal-regulated kinase (EGFR-MAPK-ERK) pathway. Our research provides new insights into the molecular function of EWI-2 on PCa metastasis, and highlights EWI-2 as a potential PCa biomarker.
Collapse
Affiliation(s)
- Chenying Fu
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Ani Wang
- Cadiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Songpeng Yang
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangfu Jiang
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Bai
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, China
| | - Quan Wei
- State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Rehabilitation Medicine Center, Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Muscarella AM, Aguirre S, Hao X, Waldvogel SM, Zhang XHF. Exploiting bone niches: progression of disseminated tumor cells to metastasis. J Clin Invest 2021; 131:143764. [PMID: 33720051 PMCID: PMC7954594 DOI: 10.1172/jci143764] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Many solid cancers metastasize to the bone and bone marrow (BM). This process may occur even before the diagnosis of primary tumors, as evidenced by the discovery of disseminated tumor cells (DTCs) in patients without occult malignancies. The cellular fates and metastatic progression of DTCs are determined by complicated interactions between cancer cells and BM niches. Not surprisingly, these niches also play important roles in normal biology, including homeostasis and turnover of skeletal and hematopoiesis systems. In this Review, we summarize recent findings on functions of BM niches in bone metastasis (BoMet), particularly during the early stage of colonization. In light of the rich knowledge of hematopoiesis and osteogenesis, we highlight how DTCs may progress into overt BoMet by taking advantage of niche cells and their activities in tissue turnover, especially those related to immunomodulation and bone repair.
Collapse
Affiliation(s)
- Aaron M. Muscarella
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah M. Waldvogel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Yamaguchi M, Murata T, Ramos JW. The botanical component p-hydroxycinnamic acid suppresses the growth and bone metastatic activity of human prostate cancer PC-3 cells in vitro. J Cancer Res Clin Oncol 2021; 147:339-350. [PMID: 33001270 DOI: 10.1007/s00432-020-03405-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/21/2020] [Indexed: 11/29/2022]
Abstract
Bone metastatic prostate cancer is one of the most common malignancies in developed countries and the second leading cause of cancer-related death in men. There remains no effective treatment for metastatic prostate cancer. We investigate here the anticancer effects of botanical component p-hydroxycinnamic acid (HCA) on the PC-3 cells in vitro model of bone metastatic human prostate cancer. Culturing with HCA (10-1000 nM) suppressed colony formation and growth of PC-3 cells. Mechanistically, culturing with HCA decreased protein levels of Ras, PI3K, Akt, MAPK, NF-κB p65 and β-catenin related to processes of cell signaling and transcription, and it increased levels of p21, p53, retinoblastoma and regucalcin, which are suppressors in carcinogenesis. These alterations can lead to suppression of cell growth. Furthermore, culturing with HCA increased cell death and caspase-3 levels. The effects of HCA on the growth and death of PC-3 cells were blocked by culturing with CH223191, an antagonist of aryl hydrocarbon receptor (AHR), suggesting that HCA effects are partly involved in AHR signaling. Interestingly, HCA suppressed the stimulatory effects of Bay K 8644, an agonist of L-type calcium channel, on the growth of PC-3 cells. Coculturing of PC-3 cells and preosteoblastic MC-3T3 E1 cells increased osteoblastic mineralization. This increase was not attenuated by treatment of HCA that stimulated mineralization. Notably, osteoclastogenesis from preosteoclastic RAW264.7 cells was enhanced by coculturing with PC-3 cells, and this enhancement was suppressed by treatment with HCA (10-1000 nM). Thus, HCA has anticancer effects on bone metastatic human prostate cancer, potentially providing a novel therapeutic tool.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Hawaii, HI, 96813, USA.
| | - Tomiyasu Murata
- Laboratory of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Yagotoyama 150, Tempaku, Nagoya, 468-8503, Japan
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Hawaii, HI, 96813, USA
| |
Collapse
|
31
|
Zhao Q, Hai B, Kelly J, Wu S, Liu F. Extracellular vesicle mimics made from iPS cell-derived mesenchymal stem cells improve the treatment of metastatic prostate cancer. Stem Cell Res Ther 2021; 12:29. [PMID: 33413659 PMCID: PMC7792192 DOI: 10.1186/s13287-020-02097-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/10/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) and their mimics from mesenchymal stem cells (MSCs) are promising drug carriers to improve cancer treatment, but their application is hindered by donor variations and expansion limitations of conventional tissue-derived MSCs. To circumvent these issues, we made EV-mimicking nanovesicles from standardized MSCs derived from human induced pluripotent stem cells (iPSCs) with a theoretically limitless expandability, and examined the targeting capacity of these nanovesicles to prostate cancer. METHODS Nanovesicles are made from intact iPSC-MSCs through serial extrusion. The selective uptake of fluorescently labeled nanovesicles by prostate cancer cells vs. non-tumor cells was examined with flow cytometry. For in vivo tracing, nanovesicles were labeled with fluorescent dye DiR or renilla luciferase. In mice carrying subcutaneous or bone metastatic PC3 prostate cancer, the biodistribution of systemically infused nanovesicles was examined with in vivo and ex vivo imaging of DiR and luminescent signals. A chemotherapeutic drug, docetaxel, was loaded into nanovesicles during extrusion. The cytotoxicities of nanovesicle-encapsulated docetaxel on docetaxel-sensitive and -resistant prostate cancer cells and non-tumor cells were examined in comparison with free docetaxel. Therapeutic effects of nanovesicle-encapsulated docetaxel were examined in mice carrying subcutaneous or bone metastatic prostate cancer by monitoring tumor growth in comparison with free docetaxel. RESULTS iPSC-MSC nanovesicles are more selectively taken up by prostate cancer cells vs. non-tumor cells in vitro compared with EVs, membrane-only EV-mimetic nanoghosts and liposomes, which is not affected by storage for up to 6 weeks. In mouse models of subcutaneous and bone metastatic PC3 prostate cancer, systemically infused nanovesicles accumulate in tumor regions with significantly higher selectivity than liposomes. The loading of docetaxel into nanovesicles was efficient and did not affect the selective uptake of nanovesicles by prostate cancer cells. The cytotoxicities of nanovesicle-encapsulated docetaxel are significantly stronger on docetaxel-resistant prostate cancer cells and weaker on non-tumor cells than free docetaxel. In mouse models of subcutaneous and bone metastatic prostate cancer, nanovesicle-encapsulated docetaxel significantly decreased the tumor growth and toxicity to white blood cells compared with free docetaxel. CONCLUSIONS Our data indicate that EV-mimicking iPSC-MSC nanovesicles are promising to improve the treatment of metastatic prostate cancer.
Collapse
Affiliation(s)
- Qingguo Zhao
- Institute for Regenerative Medicine, Molecular and Cellular Medicine Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA
| | - Bo Hai
- Institute for Regenerative Medicine, Molecular and Cellular Medicine Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA
| | - Jack Kelly
- Institute for Regenerative Medicine, Molecular and Cellular Medicine Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA
| | - Samuel Wu
- Institute for Regenerative Medicine, Molecular and Cellular Medicine Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA
| | - Fei Liu
- Institute for Regenerative Medicine, Molecular and Cellular Medicine Department, College of Medicine, Texas A&M University Health Science Center, College Station, TX, 77843, USA.
| |
Collapse
|
32
|
Yamaguchi M, Osuka S, Murata T, Ramos JW. Progression-free survival of prostate cancer patients is prolonged with a higher regucalcin expression in the tumor tissues: Overexpressed regucalcin suppresses the growth and bone activity in human prostate cancer cells. Transl Oncol 2021; 14:100955. [PMID: 33232921 PMCID: PMC7691610 DOI: 10.1016/j.tranon.2020.100955] [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: 08/20/2020] [Revised: 10/30/2020] [Accepted: 11/11/2020] [Indexed: 01/13/2023] Open
Abstract
Prostate cancer, which is a bone metastatic cancer, is the second leading cause of cancer-related death in men. There is no effective treatment for metastatic prostate cancer. Regucalcin has been shown to contribute as a suppressor in various types of human cancers. In the present study, furthermore, we investigate an involvement of regucalcin in suppression of prostate cancer. Regucalcin expression was compared in 131 primary tumor tissues and 19 metastatic tumor tissues in prostate cancer patients. Regucalcin expression in the metastatic tumor was found to be reduced as compared with that in primary tumor. The progression-free survival rate was prolonged in patients with a higher regucalcin expression. Translationally, overexpression of regucalcin in bone metastatic human prostate cancer PC-3 and DU-145 cells suppressed colony formation and cell growth in vitro. Mechanistically, overexpressed regucalcin enhanced the levels of p53, Rb, and p21, and decreased the levels of Ras, PI3 kinase, Akt, and mitogen-activated protein kinase, leading to suppression of cell growth. Furthermore, higher regucalcin expression suppressed the levels of nuclear factor-κB p65, β-catenin, and signal transducer and activator of transcription 3, which regulate a transcription activity. Cell growth was promoted by culturing with the calcium agonist Bay K 8644. This effect was blocked by overexpression of regucalcin. Notably, overexpressed regucalcin suppressed bone metastatic activity of PC-3 and DU-145 cells when cocultured with preosteoblastic or preosteoclastic cells. Regucalcin may suppress the development of human prostate cancer, suggesting that gene delivery systems in which its expression is forced may be a novel therapeutic strategy.
Collapse
Affiliation(s)
- Masayoshi Yamaguchi
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Hawaii, HI 96813, USA.
| | - Satoru Osuka
- Department of Neurosurgery, Wallace Tumor Institute, The University of Alabama at Birmingham, WTI 520A, 1720 2nd Avenue South, Birmingham, AL 35294, USA
| | - Tomiyasu Murata
- Laboratory of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Yagotoyama 150, Tempaku, Nagoya 468-8503, Japan
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo Street, Hawaii, HI 96813, USA
| |
Collapse
|
33
|
Ordikhani F, Zandi N, Mazaheri M, Luther GA, Ghovvati M, Akbarzadeh A, Annabi N. Targeted nanomedicines for the treatment of bone disease and regeneration. Med Res Rev 2020; 41:1221-1254. [PMID: 33347711 DOI: 10.1002/med.21759] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 10/14/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022]
Abstract
Targeted delivery by either passive or active targeting of therapeutics to the bone is an attractive treatment for various bone related diseases such as osteoporosis, osteosarcoma, multiple myeloma, and metastatic bone tumors. Engineering novel drug delivery carriers can increase therapeutic efficacy and minimize the risk of side effects. Developmnet of nanocarrier delivery systems is an interesting field of ongoing studies with opportunities to provide more effective therapies. In addition, preclinical nanomedicine research can open new opportunities for preclinical bone-targeted drug delivery; nevertheless, further research is needed to progress these therapies towards clinical applications. In the present review, the latest advancements in targeting moieties and nanocarrier drug delivery systems for the treatment of bone diseases are summarized. We also review the regeneration capability and effective delivery of nanomedicines for orthopedic applications.
Collapse
Affiliation(s)
- Farideh Ordikhani
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nooshin Zandi
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran.,Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Mozhdeh Mazaheri
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| | - Gaurav A Luther
- Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California- Los Angeles, California, Los Angeles, USA
| | - Abolfazl Akbarzadeh
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.,Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California- Los Angeles, California, Los Angeles, USA
| |
Collapse
|
34
|
Zhu Z, Tang G, Yan J. MicroRNA-122 regulates docetaxel resistance of prostate cancer cells by regulating PKM2. Exp Ther Med 2020; 20:247. [PMID: 33178345 PMCID: PMC7651870 DOI: 10.3892/etm.2020.9377] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa), an epithelial malignancy that occurs in the prostate, is the second leading cause of cancer death worldwide. MicroRNAs (miRs/miRNAs) are reported to have important applications in the field of cancer diagnosis and treatment. The present study aimed to investigate the function of miRNA-122 in the chemoresistance of PCa cells and the underlying mechanism. Significantly decreased miR-122 and increased pyruvate kinase (PKM2) levels were observed in docetaxel-resistant PCa cells, and PKM2 was negatively correlated with miR-122. MiR-122 mimic transfection in docetaxel-resistant LNCaP cells significantly inhibited cell proliferation, promoted apoptosis and decreased glucose uptake and lactate production, which was counteracted by PKM2 overexpression. Inhibition of miR-122 in LNCaP cells had an opposite effect to miR-122 mimic transfection. In addition, miR-122 mimic transfection significantly increased the sensitivity of docetaxel-resistant LNCaP cells to docetaxel, while inhibition of miR-122 significantly decreased the sensitivity of LNCaP cells to docetaxel. Luciferase reporter assays showed that miR-122 regulated PKM2 expression by binding to the 3'-untranslated region of PKM2. The results suggest that upregulation of miR-122 could enhance docetaxel sensitivity, inhibit cell proliferation and promote apoptosis in PCa cells,possibly through the downregulation of its target protein PKM2.
Collapse
Affiliation(s)
- Zhirong Zhu
- Department of Urology, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, P.R. China
| | - Guiliang Tang
- Department of Urology, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, P.R. China
| | - Jiajun Yan
- Department of Urology, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
35
|
Wang H, Zhang W, Bado I, Zhang XHF. Bone Tropism in Cancer Metastases. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036848. [PMID: 31615871 DOI: 10.1101/cshperspect.a036848] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone is a frequent site of metastases in many cancers. Both bone properties and the tumor-intrinsic traits are associated with the metastatic propensity to bone (i.e., the bone tropism). Whereas an increasing body of mechanistic studies expanded our understanding on bone tropism, they also revealed complexity across the bone lesions originated from different cancer types. In this review, we will discuss the physical, chemical, and biological properties of bone microenvironment, identify potential players in every stage of bone metastases, and introduce some of the known mechanisms regulating the bone colonization. Our objectives are to integrate the knowledge established in different biological contexts and highlight the determinants of bone tropism.
Collapse
Affiliation(s)
- Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA.,McNair Medical Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
36
|
Wang C, Xu K, Wang Y, Mao Y, Huang Y, Liang Y, Liu Y, Hao J, Gu X, Ma Z, Sun Y. Spinal cannabinoid receptor 2 activation reduces hypersensitivity associated with bone cancer pain and improves the integrity of the blood-spinal cord barrier. Reg Anesth Pain Med 2020; 45:783-791. [PMID: 32796132 PMCID: PMC7513263 DOI: 10.1136/rapm-2019-101262] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 02/05/2023]
Abstract
Background Disruption of the blood–spinal cord barrier (BSCB) can facilitate inflammation that results in pain hypersensitivity. Proinflammatory cytokines produced by activated microglia and astrocytes damage the BSCB. This study aims to explore whether the BSCB is damaged in the bone cancer pain (BCP) model and to investigate a potential role and mechanism of JWH015 ((2-methyl-1-propyl-1H-indol-3-yl)−1-naphthalenylmethanone), a selective cannabinoid receptor 2 (CB2R) agonist, in preserving the BSCB integrity in the BCP model. Methods We used a male mouse model of BCP. Pain hypersensitivity was measured over time. Evans blue dye extravasation, transmission electron microscopy and Western blotting were performed to investigate the permeability and structural integrity of the BSCB. Immunofluorescence staining and western blotting were used to investigate the effect of JWH015 on the activation of glial cells and the levels of proinflammatory cytokines. Results A single intrathecal injection of JWH015 ameliorated pain hypersensitivity, the BSCB disruption and microglia and astrocyte activation. Decreases in the expression of ZO-1 and claudin-5 were partially restored by JWH015. The levels of the proinflammatory cytokines interleukin-1β and tumor necrosis factor-α and the enzyme MMP9 were reduced by JWH015. However, all effects were prevented by pretreatment with a CB2R-selective antagonist, AM630 ((6-iodo-2-methyl-1-(2-morpholinoethyl)−1H-indol-3-yl)(4-methoxyphenyl)methanone). Conclusions JWH015 alleviates neuroinflammation and maintains the BSCB integrity and permeability in a mouse model of BCP, which is probably mediated by inhibiting glial cells activation. This study reveals the new analgesic mechanism of JWH015 on BCP and provides a perspective to explore novel drugs that target the BSCB to control BCP.
Collapse
Affiliation(s)
- Chenchen Wang
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Ke Xu
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yu Wang
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yanting Mao
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Ying Liang
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yue Liu
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Jing Hao
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Yu'e Sun
- Department of Anesthesiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
37
|
Chen J, Liu X, Ke K, Zou J, Gao Z, Habuchi T, Yang X. LINC00992 contributes to the oncogenic phenotypes in prostate cancer via targeting miR-3935 and augmenting GOLM1 expression. BMC Cancer 2020; 20:749. [PMID: 32781986 PMCID: PMC7418399 DOI: 10.1186/s12885-020-07141-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Accumulating evidence has revealed the critical role of long non-coding RNAs (lncRNAs) in cellular processes during tumor progression. As documented in cancer-related literatures, LINC00992 expression is associated with cancer progression, whereas its function in tumors including prostate cancer has not been characterized yet. METHODS Data from GEPIA database suggested LINC00992 expression in prostate cancer tissues. The expression levels of RNAs were monitored via qRT-PCR. Western blot evaluated the levels of proteins. The proliferation, apoptosis and migration of prostate cancer cells were assessed by CCK-8, EdU, TUNEL, Transwell and wound healing assays. Luciferase reporter, RNA pull down and RIP assays were applied to detect the interplays among LINC00992, miR-3935 and GOLM1. RESULTS Elevated levels of LINC00992 and GOLM1 were detected in prostate cancer tissues and cells. LINC00992 exerted facilitating functions in prostate cancer cell proliferation and migration. Mechanically, LINC00992 interacted with and negatively regulated miR-3935 to elevate GOLM1 expression in prostate cancer cells. In addition, the in vitro suppressive effect of silenced LINC00992 on prostate cancer cell proliferation and migration was reversed by GOLM1 upregulation. Likewise, LINC00992 depletion restrained tumor growth in vivo was offset by enhanced GOLM1 expression. CONCLUSIONS LINC00992 competitively bound with miR-3935 to elevate GOLM1 expression and therefore facilitate the oncogenic phenotypes of prostate cancer cells, implying a potential LINC00992-targeted therapy for prostate cancer.
Collapse
Affiliation(s)
- Jianheng Chen
- Department of Urology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xiaodong Liu
- Department of Urology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Kunbin Ke
- Department of Urology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jianan Zou
- Department of Urology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China
| | - Zhan Gao
- Department of Urology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Tomonori Habuchi
- Department of Urology, Akita University School of Medicine, Akita, 010-8543, Japan
| | - Xuezhen Yang
- Department of Urology, the Second Affiliated Hospital of Bengbu Medical College, 220 Hongye Road, Bengbu, 233000, Anhui, China.
| |
Collapse
|
38
|
Chen J, Liu X, Ke K, Zou J, Gao Z, Habuchi T, Yang X. LINC00992 contributes to the oncogenic phenotypes in prostate cancer via targeting miR-3935 and augmenting GOLM1 expression. BMC Cancer 2020. [PMID: 32781986 DOI: 10.1186/s12885-020-07141-4;(corresponding] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accumulating evidence has revealed the critical role of long non-coding RNAs (lncRNAs) in cellular processes during tumor progression. As documented in cancer-related literatures, LINC00992 expression is associated with cancer progression, whereas its function in tumors including prostate cancer has not been characterized yet. METHODS Data from GEPIA database suggested LINC00992 expression in prostate cancer tissues. The expression levels of RNAs were monitored via qRT-PCR. Western blot evaluated the levels of proteins. The proliferation, apoptosis and migration of prostate cancer cells were assessed by CCK-8, EdU, TUNEL, Transwell and wound healing assays. Luciferase reporter, RNA pull down and RIP assays were applied to detect the interplays among LINC00992, miR-3935 and GOLM1. RESULTS Elevated levels of LINC00992 and GOLM1 were detected in prostate cancer tissues and cells. LINC00992 exerted facilitating functions in prostate cancer cell proliferation and migration. Mechanically, LINC00992 interacted with and negatively regulated miR-3935 to elevate GOLM1 expression in prostate cancer cells. In addition, the in vitro suppressive effect of silenced LINC00992 on prostate cancer cell proliferation and migration was reversed by GOLM1 upregulation. Likewise, LINC00992 depletion restrained tumor growth in vivo was offset by enhanced GOLM1 expression. CONCLUSIONS LINC00992 competitively bound with miR-3935 to elevate GOLM1 expression and therefore facilitate the oncogenic phenotypes of prostate cancer cells, implying a potential LINC00992-targeted therapy for prostate cancer.
Collapse
Affiliation(s)
- Jianheng Chen
- Department of Urology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xiaodong Liu
- Department of Urology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Kunbin Ke
- Department of Urology, the First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jianan Zou
- Department of Urology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China
| | - Zhan Gao
- Department of Urology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Tomonori Habuchi
- Department of Urology, Akita University School of Medicine, Akita, 010-8543, Japan
| | - Xuezhen Yang
- Department of Urology, the Second Affiliated Hospital of Bengbu Medical College, 220 Hongye Road, Bengbu, 233000, Anhui, China.
| |
Collapse
|
39
|
Simsek DH, Sanli Y, Civan C, Engin MN, Isik EG, Ozkan ZG, Kuyumcu S. Does bone scintigraphy still have a role in the era of 68 Ga-PSMA PET/CT in prostate cancer? Ann Nucl Med 2020; 34:476-485. [PMID: 32394269 DOI: 10.1007/s12149-020-01474-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/26/2020] [Indexed: 12/21/2022]
Abstract
OBJECTION We aimed to evaluate the role of bone scintigraphy (BS) which has long been the imaging modality of choice in prostate cancer (PCa) and performed a head-to-head comparison between BS, BS + SPECT/CT and 68 Ga-PSMA-PET/CT, for the detection of bone metastasis of PCa. METHODS We evaluated 138 PCa patients who underwent BS and 68 Ga-PSMA PET/CT and SPECT/CT of 102 of 138 patients. Images were interpreted retrospectively and areas of abnormally increased tracer uptake related to PCa were documented as benign, metastatic or equivocal. Equivocal uptakes were finally diagnosed based on a consensus review of correlative imaging. Patient- and lesion-based analysis was performed. Patients with superscan images were excluded from lesion-based analysis. RESULTS At least one metastatic or equivocal uptake in skeleton was defined in 76 of 138 (55%) BS, in 33 of 102 (32.3%) SPECT/CT, and in 49 of 138 (35.5%) 68 Ga-PSMA PET/CT. 23 (16.7%) patients had also superscan findings on BS and 68 Ga-PSMA PET/CT. For patient-based analysis, sensitivity, specificity, accuracy, PPV, and NPV were calculated as 91.1%, 64.5%, 73.1%, 55.4% and 93.7% for BS; 95.5%, 82.7%, 86.9%, 72.8% and 96.2% for BS + SPECT/CT; 97.7%, 95.7%, 95.6%, 91.6% and 98.8% for 68 Ga-PSMA PET/CT. For lesion-based analysis, sensitivity, specificity, accuracy, PPV, and NPV were 53%, 63.9%, 60.1%, 42.8% and 71.8% for BS; 59.2%, 87.6%, 77.7%, 62.6% and 80% for BS + SPECT/CT; 96.4%, 98.1%, 97.5%, 96.4% and 98.1% for 68 Ga-PSMA PET/CT. CONCLUSION This study has shown that 68 Ga-PSMA PET/CT overcomes the limitations of BS and proves superiority in detecting bone metastases, even in patients with SPECT/CT. Our findings present important implications that 68 Ga-PSMA PET/CT can replace BS in future practice.
Collapse
Affiliation(s)
- Duygu Has Simsek
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey.
| | - Yasemin Sanli
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey
| | - Caner Civan
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey
| | - Muge Nur Engin
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey
| | - Emine Goknur Isik
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey
| | - Zeynep Gozde Ozkan
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey
| | - Serkan Kuyumcu
- Department of Nuclear Medicine, Istanbul University, Istanbul Faculty of Medicine, 34093, Fatih, İstanbul, Turkey
| |
Collapse
|
40
|
Liu W, Ju L, Cheng S, Wang G, Qian K, Liu X, Xiao Y, Wang X. Conditional reprogramming: Modeling urological cancer and translation to clinics. Clin Transl Med 2020; 10:e95. [PMID: 32508060 PMCID: PMC7403683 DOI: 10.1002/ctm2.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Patient-derived models, including cell models (organoids and conditionally reprogrammed cells [CRCs]) and patient-derived xenografts, are urgently needed for both basic and translational cancer research. Conditional reprogramming (CR) technique refers to a co-culture system of primary human normal or tumor cells with irradiated murine fibroblasts in the presence of a Rho-associated kinase inhibitor to allow the primary cells to acquire stem cell properties and the ability to proliferate indefinitely in vitro without any exogenous gene or viral transfection. Considering its robust features, the CR technique may facilitate cancer research in many aspects. Under in vitro culturing, malignant CRCs can share certain genetic aberrations and tumor phenotypes with their parental specimens. Thus, tumor CRCs can promisingly be utilized for the study of cancer biology, the discovery of novel therapies, and the promotion of precision medicine. For normal CRCs, the characteristics of normal karyotype maintenance and lineage commitment suggest their potential in toxicity testing and regenerative medicine. In this review, we discuss the applications, limitations, and future potential of CRCs in modeling urological cancer and translation to clinics.
Collapse
Affiliation(s)
- Wei Liu
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Lingao Ju
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Songtao Cheng
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Gang Wang
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Kaiyu Qian
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Yu Xiao
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Xinghuan Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Medical Research InstituteWuhan UniversityWuhanChina
| |
Collapse
|
41
|
Ge J, Mao L, Xu W, Fang W, Wang N, Ye D, Dong Z, Guan H, Guan C. miR-103a-3p Suppresses Cell Proliferation and Invasion by Targeting Tumor Protein D52 in Prostate Cancer. J INVEST SURG 2020; 34:984-992. [PMID: 32166986 DOI: 10.1080/08941939.2020.1738602] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Growing evidence points at an association between microRNAs and tumor development. Although dysregulation of microRNA-103a-3p (miR-103a-3p) in multiple human cancers has been reported, its expression in prostate cancer (PCa) remains unknown and there is currently no research on the relationship between miR-103a-3p and tumor protein D52 (TPD52) in PCa. Our aim in this study was to explore the effect and potential mechanism of miR-103a-3p in PCa. qRT-PCR was performed to detected the level of miR-103a-3p in PCa tissues and cells, and in normal tissues. Colony, wound-healing, invasion, proliferation, and apoptosis assays were performed in search miR-103a-3p effect in PCa. TargetScan was used to predict potential targets of miR-103a-3p. Additionally, dual-luciferase reporter, western blot, and immunofluorescence assays were performed to detected the target gene of miR-103a-3p. Finally, we explore the differences in tumor xenograft experiments between nude mice injected with stably miR-103a-3p expressing cells and those expressing a miR-negative control. Low level of miR-103a-3p was detected in PCa tissues and cells, when compared with normal tissues. Enhancement of miR-103a-3p significantly inhibited migration and invasion of PCa cells, and negatively regulated expression of the oncogenic tumor protein D52 (TPD52) through direct binding to its 3'-UTR. Interestingly, overexpression of TPD52 significantly attenuated the effect of mir-103a-3p on PCa. Our study provides the first evidence that miR-103a-3p directly targets TPD52 and inhibits the proliferation and invasion of PCa. This finding helps clarify the role of mir-103a-3p-TPD52 axis in PCa and may provide new therapeutic targets for the disease.
Collapse
Affiliation(s)
- Jiyue Ge
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China
| | - Likai Mao
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Weiqiang Xu
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Wenge Fang
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ningning Wang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Dawen Ye
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhuang Dong
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Han Guan
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Chao Guan
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
42
|
Abstract
A variety of diet and lifestyle factors have been studied with respect to prostate cancer risk in large, prospective cohort studies. In spite of this work, and in contrast to other common cancers, few modifiable risk factors have been firmly established as playing a role in prostate cancer. There are several possible explanations for the lack of well-established risk factors. First, prostate cancer has among the highest heritability of all common cancers; second, early life exposures may play an important role in risk, rather than mid- and later-life exposures assessed in most epidemiological studies. Finally, prostate-specific antigen (PSA) screening plays a critical role in prostate cancer detection and incidence rates, which has important implications for epidemiological studies.Among modifiable risk factors, smoking and obesity are consistently associated with higher risk specifically of advanced prostate cancer. There is also considerable evidence for a positive association between dairy intake and overall prostate cancer risk, and an inverse association between cooked tomato/lycopene intake and risk of advanced disease. Several other dietary factors consistently associated with risk in observational studies, including selenium and vitamin E, have been cast into doubt by results from clinical trials. Results for other well-studied dietary factors, including fat intake, red meat, fish, vitamin D, soy and phytoestrogens are mixed.In practical terms, men concerned with prostate cancer risk should be encouraged to stop smoking, be as physically active as possible, and achieve or maintain a healthy weight. These recommendations also have the advantage of having a positive impact on risk of type 2 diabetes, cardiovascular disease, and other chronic diseases. Reducing dairy intake while increasing consumption of fish and tomato products is also reasonable advice.
Collapse
|
43
|
Mao Y, Wang C, Tian X, Huang Y, Zhang Y, Wu H, Yang S, Xu K, Liu Y, Zhang W, Gu X, Ma Z. Endoplasmic Reticulum Stress Contributes to Nociception via Neuroinflammation in a Murine Bone Cancer Pain Model. Anesthesiology 2020; 132:357-372. [PMID: 31939851 DOI: 10.1097/aln.0000000000003078] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Prolonged endoplasmic reticulum stress has been identified in various diseases. Inflammatory mediators, which have been shown to induce endoplasmic reticulum stress in several studies, have been suggested to serve as the important modulators in pain development. In this study, the authors hypothesized that the endoplasmic reticulum stress triggered by inflammatory mediators contributed to pain development. METHODS The authors used a male mouse model of bone cancer pain. The control mice were intrathecally injected with tumor necrosis factor-α (TNF-α) and lipopolysaccharide, the bone cancer pain mice were intrathecally injected with the endoplasmic reticulum stress inhibitors 4-PBA and GSK2606414. The nociceptive behaviors, endoplasmic reticulum stress markers, and inflammatory mediators were assessed. RESULTS Increased expression of the p-RNA-dependent protein kinase-like endoplasmic reticulum kinase and p-eukaryotic initiation factor 2α were found in the spinal neurons during bone cancer pain, along with upregulation of inflammatory mediators (TNF-α, interleukin 1β, and interleukin 6). Intrathecal administration of TNF-α or lipopolysaccharide increased the expression of endoplasmic reticulum stress markers in control mice. Inhibition of endoplasmic reticulum stress by intrathecal administration of 4-PBA (baseline vs. 3 h: 0.34 ± 0.16 g vs. 1.65 ± 0.40 g in paw withdrawal mechanical threshold, 8.00 ± 1.20 times per 2 min vs. 0.88 ± 0.64 times per 2 min in number of spontaneous flinches, P < 0.001, n = 8) or GSK2606414 (baseline vs. 3 h: 0.37 ± 0.08 g vs. 1.38 ± 0.11 g in paw withdrawal mechanical threshold, 8.00 ± 0.93 times per 2 min vs. 3.25 ± 1.04 times per 2 min in number of spontaneous flinches, P < 0.001, n = 8) showed time- and dose-dependent antinociception. Meanwhile, decreased expression of inflammatory mediators (TNF-α, interleukin 1β, and interleukin 6), as well as decreased activation of astrocytes in the spinal cord, were found after 4-PBA or GSK2606414 treatment. CONCLUSIONS Inhibition of inflammatory mediator-triggered endoplasmic reticulum stress in spinal neurons attenuates bone cancer pain via modulation of neuroinflammation, which suggests new approaches to pain relief.
Collapse
Affiliation(s)
- Yanting Mao
- From the Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mathematical modelling of the role of Endo180 network in the development of metastatic bone disease in prostate cancer. Comput Biol Med 2020; 117:103619. [PMID: 32072971 DOI: 10.1016/j.compbiomed.2020.103619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/11/2020] [Accepted: 01/11/2020] [Indexed: 12/29/2022]
Abstract
Metastatic bone disease (MBD) is a common complication of advanced cancer and recent research suggests that Endo180 expression is dysregulated through the TGFβ-TGFβR-SMAD2/3 signalling pathway during the invasion of tumour cells in the development of MBD. We here provide a model for the dysregulation of the Endo180 network to demonstrate its vital contribution to bone destruction as well as tumour cell growth. The model consisted of a set of ordinary differential equations and reconstructed variations in the bone cells, resultant bone volume, and biochemical factors involved in the TGFβ-TGFβR-SMAD2/3 signalling pathway over time. The model also investigated the underlying mechanism in which the change of TGFβ affects the TGFβ-TGFβR-SMAD2/3 signalling pathway and the resultant Endo180 expression in osteoblastic and tumour cells. The model links the appearance of tumour cells with the inhibition of TGFβ binding to its receptors on osteoblastic cells, to affect TGFβ-TGFβR-SMAD2/3 signalling and Endo180 expression. Temporal variation in bone cells, bone volume, and the biochemical factors involved in the TGFβ-TGFβR-SMAD2/3 pathway as demonstrated in the model simulations agree with published experimental data. The model can be refined based on further discoveries but allows the influence of Endo180 network dysregulation on bone remodelling in MBD to be established. This model could aid in the development of Endo180 targeted therapies for MBD in the future.
Collapse
|
45
|
Cursano MC, Iuliani M, Casadei C, Stellato M, Tonini G, Paganelli G, Santini D, De Giorgi U. Combination radium-223 therapies in patients with bone metastases from castration-resistant prostate cancer: A review. Crit Rev Oncol Hematol 2020; 146:102864. [PMID: 31986318 DOI: 10.1016/j.critrevonc.2020.102864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 01/26/2023] Open
Abstract
Chemotherapeutic agents (docetaxel, cabazitaxel), hormonal therapies (abiraterone, enzalutamide) and radium-223 improve survival in patients with bone metastatic castration-resistant prostate cancer (mCRPC). Combinations of radium-223 with these agents or novel drugs have been investigated in order to improve survival and decrease bone-related morbidity. In mCRPC, clinical and preclinical data indicate that radium-223, abiraterone and enzalutamide have a direct effect on prostate cancer cells and bone microenvironment when administered as single agents. Initial results from studies of radium-223 and abiraterone, enzalutamide or docetaxel demonstrated efficacy without any safety concern in pre-treated mCRPC; however, this safety profile changed when radium-based combination therapies were administered in un-pretreated mCRPC. This review underline the biological rationale for combining radium strategies, investigating their effects on bone in terms of control of skeletal-related events and bone disease progression. The aim is to understand the possible reasons why different radium-based combination treatments can led to different clinical outcomes.
Collapse
Affiliation(s)
- M C Cursano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy.
| | - M Iuliani
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - C Casadei
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| | - M Stellato
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - G Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - G Paganelli
- Department of Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| | - D Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - U De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| |
Collapse
|
46
|
Wu L, Xiang S, Hu X, Mo M, Zhao C, Cai Y, Tong S, Jiang H, Chen L, Wang Z, Xiong W, Ou Z. Prostate-specific antigen modulates the osteogenic differentiation of MSCs via the cadherin 11-Akt axis. Clin Transl Med 2020; 10:363-373. [PMID: 32508049 PMCID: PMC7240859 DOI: 10.1002/ctm2.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A high prevalence of osteoblastic bone metastases is characteristic of prostate cancer. Prostate-specific antigen (PSA) is a serine protease uniquely produced by prostate cancer cells and is an important serological marker for prostate cancer. However, whether PSA modulates the osteogenic process remains largely unknown. In this study, we explored the effect of PSA on modulating the osteoblastic differentiation of mesenchymal stem cells (MSCs). In this study, we used flow cytometry, CCK-8 assay, Alizarin red S (ARS) staining and quantification, alkaline phosphatase (ALP) activity and staining, Western blotting, and quantitative real-time PCR (qRT-PCR) to explore the effect of PSA on osteogenic differentiation of MSCs. RESULTS We first demonstrated that although PSA did not affect the proliferation, morphology, or phenotype of MSCs, it significantly promoted the osteogenic differentiation of MSCs in a concentration-dependent manner. Furthermore, we demonstrated that PSA promoted the osteogenic differentiation of MSCs by elevating the expression of Cadherin 11 in MSCs and, thus, activating the Akt signaling pathway. CONCLUSIONS In conclusion, we demonstrated that PSA could promote the osteogenesis of MSCs through Akt signaling pathway activation by elevating the expression of cadherin-11 in MSCs. These findings imply a possible role of PSA in osteoblastic bone metastases in prostate cancer.
Collapse
Affiliation(s)
- Longxiang Wu
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Shiqi Xiang
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaP.R. China
| | - Xiheng Hu
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Miao Mo
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Cheng Zhao
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Yi Cai
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Shiyu Tong
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Huichuan Jiang
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Linxiao Chen
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Zhi Wang
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Wei Xiong
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Zhenyu Ou
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| |
Collapse
|
47
|
Ashraf MAB, Zahid A, Ashraf S, Waquar S, Iqbal S, Malik A. Implication of Prophetic Variables and their Impulsive Interplay in CA Prostate Patients Experiencing Osteo-Metastasis. Anticancer Agents Med Chem 2020; 20:2106-2113. [PMID: 32718298 DOI: 10.2174/1871520620666200727094430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/06/2020] [Accepted: 06/11/2020] [Indexed: 12/30/2022]
Abstract
AIMS To identify variables having a critical role in prostate cancer patients experiencing osteometastasis. BACKGROUND Prostatic carcinoma is a multifactorial complex disorder that exhibits an increased propensity to develop bone metastasis. An interplay of inflammatory and bone remodeling parameters promotes the formation of pre-metastatic niches in bones of patients, which could render them more vulnerable to skeletal disabilities. OBJECTIVE To evaluate the multi-dynamic inter-relationship of circulating variables in prostate cancer patients experiencing osteo-metastasis. MATERIALS AND METHODS Fifty-seven (n=57) men with clinically confirmed prostate cancer, fifty-nine (n=59) with skeletal metastases, and one hundred (n=100) healthy subjects i.e., men aging from 53-84 years with no clinical evidence of prostate were recruited from the Jinnah Hospital Lahore, Pakistan. Informed consent was obtained, and a venous blood sample was drawn and stored at -70oC until assayed. Levels of variables were evaluated using appropriate methods. Levels of Matrix Metalloproteinases (MMPs), Osteopontin (OPN), TGH- β, and sRANKL were estimated by the ELISA method. Each sample was suspended and the given protocol was employed. ELISA readings were obtained for the estimation of all variables. RESULTS Highly significant (P˂0.05) differential expression of oxidative stress, inflammatory cytokines, and bone remodeling variables were observed in localized and osteo-metastatic CA prostate patients. A strong positive correlation was revealed among OPN, sRANKL, MMP-7, MMP-9, PSA, and TGF-β (OPN vs. MMP-7, r=0.698* and OPN vs. MMP-9, r=0.765**, OPN vs. RANKL, =0.856*, sRANKL vs. MMP-9, r=0.825**, TGF- β vs. RANKL, r=0.868* and PSA vs. TGF- β, r=0.752*); lower levels of OPG were estimated in metastasized patients, showing that both osteolytic and osteoblastic phases of bone remodeling occur simultaneously. CONCLUSION The altered oxidative and inflammatory responses endorse Matrix Metalloproteinases (MMPs) increased activity, RANKL/OPG imbalance, and enhanced bone matrix proteins turnover, which can foster the process of osteo-metastasis. The perturbed RANKL/OPG drift and enhanced PSA levels are associated with increased TGF-β activity to aggravate Epithelial Mesenchymal transition (EM) and osteo-tropism of prostate cancer. Thus, designing novel targets of these major variables can minimize the incidence of prostate cancer patients.
Collapse
Affiliation(s)
- Muhammad A B Ashraf
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Ayesha Zahid
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Shazia Ashraf
- Department of Pathology, Independent Medical College, Faisalabad, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Saima Iqbal
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| |
Collapse
|
48
|
Cheng J, Moore S, Gomez-Galeno J, Lee DH, Okolotowicz KJ, Cashman JR. A Novel Small Molecule Inhibits Tumor Growth and Synergizes Effects of Enzalutamide on Prostate Cancer. J Pharmacol Exp Ther 2019; 371:703-712. [PMID: 31582422 PMCID: PMC7042720 DOI: 10.1124/jpet.119.261040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/27/2019] [Indexed: 01/20/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death for men in the United States. Approximately 35% of PCa recurs and is often transformed to castration-resistant prostate cancer (CRPCa), the most deadly and aggressive form of PCa. However, the CRPCa standard-of-care treatment (enzalutamide with abiraterone) usually has limited efficacy. Herein, we report a novel molecule (PAWI-2) that inhibits cellular proliferation of androgen-sensitive and androgen-insensitive cells (LNCaP and PC-3, respectively). In vivo studies in a PC-3 xenograft model showed that PAWI-2 (20 mg/kg per day i.p., 21 days) inhibited tumor growth by 49% compared with vehicle-treated mice. PAWI-2 synergized currently clinically used enzalutamide in in vitro inhibition of PCa cell viability and resensitized inhibition of in vivo PC-3 tumor growth. Compared with vehicle-treated mice, PC-3 xenograft studies also showed that PAWI-2 (20 mg/kg per day i.p., 21 days) and enzalutamide (5 mg/kg per day i.p., 21 days) inhibited tumor growth by 63%. Synergism was mainly controlled by the imbalance of prosurvival factors (e.g., Bcl-2, Bcl-xL, Mcl-1) and antisurvival factors (e.g., Bax, Bak) induced by affecting mitochondrial membrane potential/mitochondria dynamics. Thus, PAWI-2 utilizes a distinct mechanism of action to inhibit PCa growth independently of androgen receptor signaling and overcomes enzalutamide-resistant CRPCa. SIGNIFICANCE STATEMENT: Castration-resistant prostate cancer (CRPCa) is the most aggressive human prostate cancer (PCa) but standard chemotherapies for CRPCa are largely ineffective. PAWI-2 potently inhibits PCa proliferation in vitro and in vivo regardless of androgen receptor status and uses a distinct mechanism of action. PAWI-2 has greater utility in treating CRPCa than standard-of-care therapy. PAWI-2 possesses promising therapeutic potency in low-dose combination therapy with a clinically used drug (e.g., enzalutamide). This study describes a new approach to address the overarching challenge in clinical treatment of CRPCa.
Collapse
Affiliation(s)
- Jiongjia Cheng
- Human BioMolecular Research Institute and ChemRegen Inc., San Diego, California
| | - Stephanie Moore
- Human BioMolecular Research Institute and ChemRegen Inc., San Diego, California
| | - Jorge Gomez-Galeno
- Human BioMolecular Research Institute and ChemRegen Inc., San Diego, California
| | - Dong-Hoon Lee
- Human BioMolecular Research Institute and ChemRegen Inc., San Diego, California
| | - Karl J Okolotowicz
- Human BioMolecular Research Institute and ChemRegen Inc., San Diego, California
| | - John R Cashman
- Human BioMolecular Research Institute and ChemRegen Inc., San Diego, California
| |
Collapse
|
49
|
Guo S, Zhu W, Yin Z, Xiao D, Zhang Q, Liu T, Ni J, Ouyang Z, Xie H. Proanthocyanidins attenuate breast cancer-induced bone metastasis by inhibiting Irf-3/c-jun activation. Anticancer Drugs 2019; 30:998-1005. [PMID: 31625993 DOI: 10.1097/cad.0000000000000852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
We have previously demonstrated the pivotal role of Jnk-mediated Irf-3/c-Jun in regulating nuclear factor kappa-Β ligand (RANKL)-induced osteoclastogenesis. Here, we demonstrated that proanthocyanidins (PACs) target Irf-3 to alleviate breast cancer-induced activation of osteoclasts. We also found that PACs induced apoptosis of osteoclast precursors by upregulating the ratio of bax/bcl-2 and activating caspase-3 activity. Such bone protective effect also could be observed in a bone metastasis model of breast cancer. These findings provided a novel therapeutic intervention targeting abnormal bone metabolism to alleviate bone metastasis of breast cancer.
Collapse
Affiliation(s)
- Shuangfei Guo
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mao Y, Huang Y, Zhang Y, Wang C, Wu H, Tian X, Liu Y, Hou B, Liang Y, Rong H, Gu X, Ma Z. Cannabinoid receptor 2‑selective agonist JWH015 attenuates bone cancer pain through the amelioration of impaired autophagy flux induced by inflammatory mediators in the spinal cord. Mol Med Rep 2019; 20:5100-5110. [PMID: 31661120 PMCID: PMC6854597 DOI: 10.3892/mmr.2019.10772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 08/16/2019] [Indexed: 12/28/2022] Open
Abstract
Bone cancer pain (BCP) is a severe complication of advanced bone cancer. Although cannabinoid receptor 2 (CB2) agonists may have an analgesic effect, the underlying mechanism remains unclear. CB2 serves a protective role in various pathological states through the activation of autophagy. Therefore, the present study aimed to determine whether the analgesic effects of the selective CB2 agonist JWH015 was mediated by the activation of autophagy in BCP. BCP was induced by the intra‑femur implantation of NCTC2472 fibrosarcoma cells in C3H/HeN mice. The pain behaviors were assessed on the following postoperative days. The selective CB2 agonist JWH015 (1 and 2 µg) was intrathecally administered on day 14 following implantation. AM630 (1 µg), a CB2 antagonist, was injected 30 min before JWH015 administration. Lipopolysaccharide (LPS; 100 nM)‑stimulated primary neurons were treated with JWH015 (1 µM) and AM630 (1 µM) to further verify the mechanism by which CB2 affects autophagy. The results demonstrated that autophagy flux was impaired in spinal neurons during BCP, as indicated by the increased ratio of microtubule‑associated protein 1 light chain 3β (LC3B)‑II/LC3B‑I and increased expression of p62. Intrathecal administration of JWH015 attenuated BCP, which was accompanied by the amelioration of impaired autophagy flux (decreased LC3B‑II/LC3B‑I ratio and decreased p62expression). In addition, the activation of glia cells and upregulation of the glia‑derived inflammatory mediators, interleukin (IL)‑1β and IL‑6 were suppressed by JWH015. In LPS‑stimulated primary neurons, IL‑1β and IL‑6 were increased, and autophagy flux was impaired; whereas treatment with JWH015 decreased the expression of IL‑1β and IL‑6, LC3B‑II/LC3B‑I ratio and expression of p62. These effects were by pretreatment with the CB2‑selective antagonist AM630. The results of the present study suggested that the impairment of autophagy flux was induced by glia‑derived inflammatory mediators in spinal neurons. Intrathecal administration of the selective CB2 agonist JWH015 ameliorated autophagy flux through the downregulation of IL‑1β and IL‑6 and attenuated BCP.
Collapse
Affiliation(s)
- Yanting Mao
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Yulin Huang
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Ying Zhang
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Chenchen Wang
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Hao Wu
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xinyu Tian
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Yue Liu
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Bailing Hou
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Ying Liang
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Hui Rong
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoping Gu
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Zhengliang Ma
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|