1
|
Modi D, Hussain MS, Ainampudi S, Prajapati BG. Long acting injectables for the treatment of prostate cancer. J Drug Deliv Sci Technol 2024; 100:105996. [DOI: 10.1016/j.jddst.2024.105996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
|
2
|
Zhang C, Aida M, Saggu S, Yu H, Zhou L, Rehman H, Jiao K, Liu R, Wang L, Wang Q. Androgen deprivation therapy exacerbates Alzheimer's-associated cognitive decline via increased brain immune cell infiltration. SCIENCE ADVANCES 2024; 10:eadn8709. [PMID: 38905345 PMCID: PMC11192088 DOI: 10.1126/sciadv.adn8709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Androgen deprivation therapy (ADT) for prostate cancer is associated with an increased risk of dementia, including Alzheimer's disease (AD). The mechanistic connection between ADT and AD-related cognitive impairment in patients with prostate cancer remains elusive. We established a clinically relevant prostate cancer-bearing AD mouse model to explore this. Both tumor-bearing and ADT induce complex changes in immune and inflammatory responses in peripheral blood and in the brain. ADT disrupts the integrity of the blood-brain barrier (BBB) and promotes immune cell infiltration into the brain, enhancing neuroinflammation and gliosis without affecting the amyloid plaque load. Moreover, treatment with natalizumab, an FDA-approved drug targeting peripheral immune cell infiltration, reduces neuroinflammation and improves cognitive function in this model. Our study uncovers an inflammatory mechanism, extending beyond amyloid pathology, that underlies ADT-exacerbated cognitive deficits, and suggests natalizumab as a potentially effective treatment in alleviating the detrimental effects of ADT on cognition.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mae Aida
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Haiyan Yu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lianna Zhou
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hasibur Rehman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
3
|
Zhao J, Sun Y, Ren L, Huang S, Zhang J. Antagonism of androgen receptor signaling by aloe-emodin. Food Chem Toxicol 2023; 181:114092. [PMID: 37806336 DOI: 10.1016/j.fct.2023.114092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Over the past decades, androgen receptor (AR) signaling has been a key driver of both primary and recurrent prostate cancer. In this work, aloe-emodin was identified as a novel AR antagonist, effectively inhibiting AR signaling. Firstly, aloe-emodin can inhibit LNCaP cell growth by promoting apoptosis. Then, the results of Western blot and quantitative real-time PCR further confirmed that aloe-emodin modulated AR protein levels by promoting AR proteasomal degradation, and also inhibited the transcription of the AR downstream target genes, including PSA, KLK2, and TMPRSS2. Furthermore, the result of immunofluorescence showed that aloe-emodin prevented the nuclear translocation of AR. Molecular docking and molecular dynamics simulation suggested that aloe-emodin combined with AR to form stable complexes, which might explain that aloe-emodin prevented the translocation of AR from the cytoplasm to the nucleus by affecting the ligand binding of AR. Therefore, aloe-emodin as a novel AR antagonist may play a crucial role in promoting cancer prevention or complementing pharmacological therapies in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jingqi Zhao
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Yantong Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Shuqing Huang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| |
Collapse
|
4
|
Ertay A, Ewing RM, Wang Y. Synthetic lethal approaches to target cancers with loss of PTEN function. Genes Dis 2023; 10:2511-2527. [PMID: 37533462 PMCID: PMC7614861 DOI: 10.1016/j.gendis.2022.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 02/05/2023] Open
Abstract
Phosphatase and tensin homolog (PTEN) is a tumour suppressor gene and has a role in inhibiting the oncogenic AKT signalling pathway by dephosphorylating phosphatidylinositol 3,4,5-triphosphate (PIP3) into phosphatidylinositol 4,5-bisphosphate (PIP2). The function of PTEN is regulated by different mechanisms and inactive PTEN results in aggressive tumour phenotype and tumorigenesis. Identifying targeted therapies for inactive tumour suppressor genes such as PTEN has been challenging as it is difficult to restore the tumour suppressor functions. Therefore, focusing on the downstream signalling pathways to discover a targeted therapy for inactive tumour suppressor genes has highlighted the importance of synthetic lethality studies. This review focuses on the potential synthetic lethality genes discovered in PTEN-inactive cancer types. These discovered genes could be potential targeted therapies for PTEN-inactive cancer types and may improve the treatment response rates for aggressive types of cancer.
Collapse
Affiliation(s)
- Ayse Ertay
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Rob M. Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
5
|
Dai D, Yu J, Huang T, Li Y, Wang Z, Yang S, Li S, Li Y, Gou W, Li D, Hou W, Fan S, Li Y, Zhao Y. PET imaging of new target CDK19 in prostate cancer. Eur J Nucl Med Mol Imaging 2023; 50:3452-3464. [PMID: 37278941 DOI: 10.1007/s00259-023-06277-2] [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: 12/14/2022] [Accepted: 05/19/2023] [Indexed: 06/07/2023]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA)-positron emission tomography (PET) is a superior method to predict patients' risk of cancer progression and response to specific therapies. However, its performance is limited for neuroendocrine prostate cancer (NEPC) and PSMA-low prostate cancer cells, resulting in diagnostic blind spots. Hence, identifying novel specific targets is our aim for diagnosing those prostate cancers with low PSMA expression. METHODS The Cancer Genome Atlas (TCGA) database and our cohorts from men with biopsy-proven high-risk metastatic prostate cancer were used to identify CDK19 and PSMA expression. PDX lines neP-09 and P-16 primary cells were used for cellular uptake and imaging mass cytometry in vitro. To evaluate in vivo CDK19-specific uptake of gallium(Ga)-68-IRM-015-DOTA, xenograft mice models and blocking assays were used. PET/CT imaging data were obtained to estimate the absorbed dose in organs. RESULTS Our study group had reported the overexpression of a novel tissue-specific gene CDK19 in high-risk metastatic prostate cancer and CDK19 expression correlated with metastatic status and tumor staging, independently with PSMA and PSA levels. Following up on this new candidate for use in diagnostics, small molecules targeting CDK19 labeled with Ga-68 (68Ga-IRM-015-DOTA) were used for PET in this study. We found that the 68Ga-IRM-015-DOTA was specificity for prostate cancer cells, but the other cancer cells also took up little 68Ga-IRM-015-DOTA. Importantly, mouse imaging data showed that the NEPC and CRPC xenografts exhibited similar signal strength with 68Ga-IRM-015-DOTA, but 68Ga-PSMA-11 only stained the CRPC xenografts. Furthermore, target specificity was elucidated by a blocking experiment on a CDK19-bearing tumor xenograft. These data concluded that 68Ga-CDK19 PET/CT was an effective technology to detect lesions with or without PSMA in vitro, in vivo, and in the PDX model. CONCLUSION Thus, we have generated a novel PET small molecule with predictive value for prostate cancer. The findings indicate that 68Ga-CDK19 may merit further evaluation as a predictive biomarker for PET scans in prospective cohorts and may facilitate the identification of molecular types of prostate cancer independent of PSMA.
Collapse
Affiliation(s)
- Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300000, Tianjin, China
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, 300308, Tianjin, China
| | - Jiang Yu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Ting Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Yansheng Li
- Department of PET-CT Diagnostic, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, 300020, Tianjin, China
| | - Ziyang Wang
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, 300308, Tianjin, China
| | - Shuangmeng Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Shuai Li
- Department of PET-CT Diagnostic, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, 300020, Tianjin, China
| | - Yanli Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Wenfeng Gou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Wenbin Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China.
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China.
| | - Yu Zhao
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300000, Tianjin, China.
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 300192, Tianjin, China.
| |
Collapse
|
6
|
Roy A, Chauhan S, Bhattacharya S, Jakhmola V, Tyagi K, Sachdeva A, Wasai A, Mandal S. Runt-related transcription factors in human carcinogenesis: a friend or foe? J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04769-0. [PMID: 37081242 DOI: 10.1007/s00432-023-04769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 04/08/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE Cancer is one of the deadliest pathologies with more than 19 million new cases and 10 million cancer-related deaths across the globe. Despite development of advanced therapeutic interventions, cancer remains as a fatal pathology due to lack of early prognostic biomarkers, therapy resistance and requires identification of novel drug targets. METHODS Runt-related transcription factors (Runx) family controls several cellular and physiological functions including osteogenesis. Recent literatures from PubMed was mined and the review was written in comprehensive manner RESULTS: Recent literature suggests that aberrant expression of Runx contributes to tumorigenesis of many organs. Conversely, cell- and tissue-specific tumor suppressor roles of Runx are also reported. In this review, we have provided the structural/functional properties of Runx isoforms and its regulation in context of human cancer. Moreover, in an urgent need to discover novel therapeutic interventions against cancer, we comprehensively discussed the reported oncogenic and tumor suppressive roles of Runx isoforms in several tumor types and discussed the discrepancies that may have risen on Runx as a driver of malignant transformation. CONCLUSION Runx may be a novel therapeutic target against a battery of deadly human cancers.
Collapse
Affiliation(s)
- Adhiraj Roy
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India.
| | - Shivi Chauhan
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Vibhuti Jakhmola
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Komal Tyagi
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Abha Sachdeva
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Abdul Wasai
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal, 741235, India
| |
Collapse
|
7
|
Patierno BM, Foo WC, Allen T, Somarelli JA, Ware KE, Gupta S, Wise S, Wise JP, Qin X, Zhang D, Xu L, Li Y, Chen X, Inman BA, McCall SJ, Huang J, Kittles RA, Owzar K, Gregory S, Armstrong AJ, George DJ, Patierno SR, Hsu DS, Freedman JA. Characterization of a castrate-resistant prostate cancer xenograft derived from a patient of West African ancestry. Prostate Cancer Prostatic Dis 2022; 25:513-523. [PMID: 34645983 PMCID: PMC9005588 DOI: 10.1038/s41391-021-00460-y] [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: 04/15/2021] [Revised: 08/31/2021] [Accepted: 09/15/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Prostate cancer is a clinically and molecularly heterogeneous disease, with highest incidence and mortality among men of African ancestry. To date, prostate cancer patient-derived xenograft (PCPDX) models to study this disease have been difficult to establish because of limited specimen availability and poor uptake rates in immunodeficient mice. Ancestrally diverse PCPDXs are even more rare, and only six PCPDXs from self-identified African American patients from one institution were recently made available. METHODS In the present study, we established a PCPDX from prostate cancer tissue from a patient of estimated 90% West African ancestry with metastatic castration resistant disease, and characterized this model's pathology, karyotype, hotspot mutations, copy number, gene fusions, gene expression, growth rate in normal and castrated mice, therapeutic response, and experimental metastasis. RESULTS This PCPDX has a mutation in TP53 and loss of PTEN and RB1. We have documented a 100% take rate in mice after thawing the PCPDX tumor from frozen stock. The PCPDX is castrate- and docetaxel-resistant and cisplatin-sensitive, and has gene expression patterns associated with such drug responses. After tail vein injection, the PCPDX tumor cells can colonize the lungs of mice. CONCLUSION This PCPDX, along with others that are established and characterized, will be useful pre-clinically for studying the heterogeneity of prostate cancer biology and testing new therapeutics in models expected to be reflective of the clinical setting.
Collapse
Affiliation(s)
- Brendon M Patierno
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Wen-Chi Foo
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Tyler Allen
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jason A Somarelli
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kathryn E Ware
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Santosh Gupta
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sandra Wise
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - John P Wise
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Dadong Zhang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Lingfan Xu
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Yanjing Li
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Xufeng Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brant A Inman
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Shannon J McCall
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jiaoti Huang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Rick A Kittles
- Division of Health Equities, Department of Population Sciences, City of Hope, Duarte, 91010, CA, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Simon Gregory
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Center for Genomics and Computational Biology, Duke University, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Daniel J George
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Steven R Patierno
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - David S Hsu
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Center for Genomics and Computational Biology, Duke University, Durham, NC, 27710, USA
| | - Jennifer A Freedman
- Department of Medicine, Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
8
|
Dual contribution of the mTOR pathway and of the metabolism of amino acids in prostate cancer. Cell Oncol (Dordr) 2022; 45:831-859. [PMID: 36036882 DOI: 10.1007/s13402-022-00706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Prostate cancer is the leading cause of cancer in men, and its incidence increases with age. Among other risk factors, pre-existing metabolic diseases have been recently linked with prostate cancer, and our current knowledge recognizes prostate cancer as a condition with important metabolic anomalies as well. In malignancies, metabolic disorders are commonly associated with aberrations in mTOR, which is the master regulator of protein synthesis and energetic homeostasis. Although there are reports demonstrating the high dependency of prostate cancer cells for lipid derivatives and even for carbohydrates, the understanding regarding amino acids, and the relationship with the mTOR pathway ultimately resulting in metabolic aberrations, is still scarce. CONCLUSIONS AND PERSPECTIVES In this review, we briefly provide evidence supporting prostate cancer as a metabolic disease, and discuss what is known about mTOR signaling and prostate cancer. Next, we emphasized on the amino acids glutamine, leucine, serine, glycine, sarcosine, proline and arginine, commonly related to prostate cancer, to explore the alterations in their regulatory pathways and to link them with the associated metabolic reprogramming events seen in prostate cancer. Finally, we display potential therapeutic strategies for targeting mTOR and the referred amino acids, as experimental approaches to selectively attack prostate cancer cells.
Collapse
|
9
|
Yang W, Deng J, Gao J, Yang H, Chen Q, Niya Z, Ling X, Zhang G, Zou P, Sun L, Huang L, Liu J, Cao J, Ao L. Associations between isoflavone exposure and reproductive damage in adult males: evidence from human and model system studies. Biol Reprod 2022; 107:1360-1373. [PMID: 35948002 DOI: 10.1093/biolre/ioac157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/21/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
It's controversial whether exposure to isoflavones, constituents of certain plants such as soy bean, exerts male reproductive toxicity. This study was designed to investigate whether isoflavone exposure during adulthood could have deleterious impacts on male reproductive health by the cross-sectional study, animal experiments, and in vitro tests. In the cross-sectional study, we observed that urinary isoflavones were not significantly associated with semen quality including sperm concentrations, sperm count, progressive motility, and total motility, respectively (All P-value for trend>0.05). However, negative associations were found between plasma testosterone and urinary Σisoflavones, genistein, glycitein, and dihydrodaidzein (all P-value for trend <0.05). In the animal experiments, serum and intratesticular testosterone levels were decreased in mice exposed to several dosages of genistein. Genistein administration caused up-regulation of estrogen receptor alpha (ERα) and down-regulation of cytochrome P45017A1 (CYP17A1) protein levels in testes of mice. However, genistein treatment during adulthood did not induce appreciable structural damages of reproductive system in mice. In vitro tests, we observed that genistein of different dosages (0.01, 2.5, 10 μM) caused a concentration dependent inhibition of testosterone production by TM3 Leydig cells (half-maximal inhibitory concentration = 3.796 nM, P < 0.05). Elevated protein expression of ERα and decreased mRNA/protein level of CYP17A1 were also observed in genistein-treated cells. Protein level of CYP17A1 and testosterone concentration were significantly restored in the ERα siRNA-transfected cells, compared to cells that treated with genistein alone (P < 0.05). The results demonstrate that exposure to isoflavones during adulthood may be associated with alterations of reproductive hormones. Particularly for genistein, which inhibits testosterone biosynthesis through up-regulation of ERα in Leydig cells of mice, might induce the disruption of testosterone production in human. The present study provides novel perspective into potential targets for male reproductive compromise induced by isoflavone exposure.
Collapse
Affiliation(s)
- Wang Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Jiuyang Deng
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030001, China
| | - Jianfang Gao
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Huan Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Zhou Niya
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Xi Ling
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Guowei Zhang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Peng Zou
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Lei Sun
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Linping Huang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Jinyi Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| | - Lin Ao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Gaotanyan St 30, Chongqing 400038, China
| |
Collapse
|
10
|
Zhao H, Chen Y, Shen P, Gong L. Prognostic value and immune characteristics of RUNX gene family in human cancers: a pan-cancer analysis. Aging (Albany NY) 2022; 14:4014-4035. [PMID: 35522574 PMCID: PMC9134966 DOI: 10.18632/aging.204065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/23/2022] [Indexed: 11/25/2022]
Abstract
Background: Runt-related transcription factors (RUNX) are involved in numerous fundamental biological processes and play crucial parts in tumorigenesis and metastasis both directly and indirectly. However, the pan-cancer evidence of the RUNX gene family is not available. Methods: In this study, we analyzed the potential association between RUNX gene family expression and patient’s prognosis, immune cell infiltration, drug response, and genetic mutation data across different types of tumors using based on The Cancer Genome Atlas, Gene Expression Omnibus, and Oncomine database. Results: The results showed that the expression of the RUNX gene family varied among different cancer types, revealing its heterogeneity in cancers and that expression of RUNX2 was lower than that of RUNX1 and RUNX3 across all cancer types. RUNX gene family gene expression was related to prognosis in several cancers. Furthermore, our study revealed a clear association between RUNX gene family expression and ESTIMATE score, RNA stemness, and DNA stemness scores. Compared with RUNX1 and RUNX2, RUNX3 showed relatively low levels of genetic alterations. RUNX gene family genes had clear associations with immune infiltrate subtypes, and their expression was positively related to immune checkpoint genes and drug sensitivity in most cases. Two immunotherapy cohorts confirm that the expression of RUNX was correlated with the clinical response of immunotherapy. Conclusions: These findings will help to elucidate the potential oncogenic roles of RUNX gene family genes in different types of cancer and it can function as a prognostic marker in various malignant tumors.
Collapse
Affiliation(s)
- Han Zhao
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai 200000, Shanghai, China.,Laboratory of Myopia, NHC Key Laboratory of Myopia, Fudan University, Chinese Academy of Medical Sciences, Shanghai 200000, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200000, Shanghai, China
| | - Yun Chen
- Department of Stomatology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Peijun Shen
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Lan Gong
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai 200000, Shanghai, China.,Laboratory of Myopia, NHC Key Laboratory of Myopia, Fudan University, Chinese Academy of Medical Sciences, Shanghai 200000, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200000, Shanghai, China
| |
Collapse
|
11
|
Du Y, Sui Y, Cao J, Jiang X, Wang Y, Yu J, Wang B, Wang X, Xue B. Dynamic Changes in Myofibroblasts Affect the Carcinogenesis and Prognosis of Bladder Cancer Associated With Tumor Microenvironment Remodeling. Front Cell Dev Biol 2022; 10:833578. [PMID: 35309916 PMCID: PMC8924465 DOI: 10.3389/fcell.2022.833578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/15/2022] [Indexed: 01/22/2023] Open
Abstract
Bladder cancer (BLCA) is a tumor that possesses significant heterogeneity, and the tumor microenvironment (TME) plays an important role in the development of BLCA. The TME chiefly consists of tumor cells and tumor-infiltrating immune cells admixed with stromal components. Recent studies have revealed that stromal components, especially cancer-associated fibroblasts (CAFs), affect immune cell infiltration and modulate the extracellular matrix in the TME of BLCA, ultimately impacting the prognosis and therapeutic efficacy of BLCA. Among the subgroups of CAFs, myofibroblasts (myCAFs) were the most abundant and were demonstrated to play an essential role in affecting the prognosis of various tumors, including BLCA. However, the dynamic changes in myCAFs during carcinogenesis and tumor progression have been less discussed previously. With the help of bioinformatics algorithms, we discussed the roles of myCAFs in the carcinogenesis and prognosis of BLCA in this manuscript. Our study highlighted the pathogenesis of BLCA was accompanied by a decrease in the abundance of myCAFs, revealing potential protective properties of myCAFs in the carcinogenesis of BLCA. Meanwhile, the reduced expressions of myCAFs marker genes were highly accurate in predicting tumorigenesis. In contrast, we also demonstrated that myCAFs regulated extracellular matrix remodeling, tumor metabolism, cancer stemness, and oncological mutations, ultimately impacting the treatment responsiveness and prognosis of BLCA patients. Thus, our research revealed the bimodal roles of myCAFs in the development of BLCA, which may be associated with the temporal change of the TME. The in-depth study of myofibroblasts and the TME may provide potential diagnostic biomarkers and therapeutic targets for BLCA.
Collapse
Affiliation(s)
- YiHeng Du
- Department of Urology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - YiQun Sui
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Cao
- Department of Pathology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Xiang Jiang
- Department of Pathology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Yi Wang
- Department of Urology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Jiang Yu
- Department of Urology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Bo Wang
- Department of Urology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - XiZhi Wang
- Department of Urology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
- *Correspondence: XiZhi Wang, ; BoXin Xue,
| | - BoXin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: XiZhi Wang, ; BoXin Xue,
| |
Collapse
|
12
|
Icariin promotes mouse Leydig cell testosterone synthesis via the Esr1/Src/Akt/Creb/Sf-1 pathway. Toxicol Appl Pharmacol 2022; 441:115969. [DOI: 10.1016/j.taap.2022.115969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 11/21/2022]
|
13
|
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: 52] [Impact Index Per Article: 26.0] [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
|
14
|
Corti M, Lorenzetti S, Ubaldi A, Zilli R, Marcoccia D. Endocrine Disruptors and Prostate Cancer. Int J Mol Sci 2022; 23:1216. [PMID: 35163140 PMCID: PMC8835300 DOI: 10.3390/ijms23031216] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 01/22/2023] Open
Abstract
The role of endocrine disruptors (EDs) in the human prostate gland is an overlooked issue even though the prostate is essential for male fertility. From experimental models, it is known that EDs can influence several molecular mechanisms involved in prostate homeostasis and diseases, including prostate cancer (PCa), one of the most common cancers in the male, whose onset and progression is characterized by the deregulation of several cellular pathways including androgen receptor (AR) signaling. The prostate gland essentiality relies on its function to produce and secrete the prostatic fluid, a component of the seminal fluid, needed to keep alive and functional sperms upon ejaculation. In physiological condition, in the prostate epithelium the more-active androgen, the 5α-dihydrotestosterone (DHT), formed from testosterone (T) by the 5α-reductase enzyme (SRD5A), binds to AR and, upon homodimerization and nuclear translocation, recognizes the promoter of target genes modulating them. In pathological conditions, AR mutations and/or less specific AR binding by ligands modulate differently targeted genes leading to an altered regulation of cell proliferation and triggering PCa onset and development. EDs acting on the AR-dependent signaling within the prostate gland can contribute to the PCa onset and to exacerbating its development.
Collapse
Affiliation(s)
- Margherita Corti
- Istituto Zooprofilattico Sperimentale del Lazio e della Toscana, Via Appia Nuova 1411, 00178 Rome, Italy; (M.C.); (A.U.); (R.Z.)
| | - Stefano Lorenzetti
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy;
| | - Alessandro Ubaldi
- Istituto Zooprofilattico Sperimentale del Lazio e della Toscana, Via Appia Nuova 1411, 00178 Rome, Italy; (M.C.); (A.U.); (R.Z.)
| | - Romano Zilli
- Istituto Zooprofilattico Sperimentale del Lazio e della Toscana, Via Appia Nuova 1411, 00178 Rome, Italy; (M.C.); (A.U.); (R.Z.)
| | - Daniele Marcoccia
- Istituto Zooprofilattico Sperimentale del Lazio e della Toscana, Via Appia Nuova 1411, 00178 Rome, Italy; (M.C.); (A.U.); (R.Z.)
| |
Collapse
|
15
|
Song L, Huang Y, Long J, Li Y, Pan Z, Fang F, Long Y, Huang C, Qi N, Guo Y, Xia H, Jiang Y. The Role of Osteocalcin in Placental Function in Gestational Diabetes Mellitus. Reprod Biol 2021; 21:100566. [PMID: 34626941 DOI: 10.1016/j.repbio.2021.100566] [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: 06/29/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 10/20/2022]
Abstract
Evidence for the role of osteocalcin in glucose metabolism is increasing. The aim of this study was to examine the associations between osteocalcin and gestational diabetes mellitus. Thirteen discovery study subjects and 76 reduplication study subjects were recruited from the Maternal and Child Health Hospital Guangxi Zhuang Autonomous Region from May 2018 to August 2018. Total osteocalcin and biochemical indices of maternal serum and umbilical vein serum were analyzed. Placental tissue samples were used for transcriptome sequencing. For the discovery study subjects, the total osteocalcin concentration in umbilical vein serum was significantly higher than that in maternal serum and umbilical artery serum (55.32 ng/mL ± 17.37 vs. 12.06 ng/mL ± 5.42 [P < 0.001] vs. 38.31 ng/mL ± 11.52 [P < 0.01]), suggesting that trophoblasts may synthesize osteocalcin. In a reduplication subject study, the gestational diabetes mellitus group had lower umbilical vein serum total osteocalcin (51.46 ng/mL ± 24.29 vs. 67.00 ng/mL ± 25.33, P = 0.008), lower adiponectin (1099.72 μg/L ± 102.65 vs. 1235.85 μg/L ± 94.63, P < 0.001). Spearman's correlation analysis showed that umbilical vein serum total osteocalcin levels were closely correlated with leptin (r = -0.456, P = 0.007). A coexpression model of the placental RNA sequence was constructed. Two modules were correlated with osteocalcin, and the Gene ontology pathways of these modules were rich in glucose and lipid metabolism. In conclusion, the placenta may synthesize osteocalcin by itself, and a lower osteocalcin level in umbilical vein serum is associated with gestational diabetes mellitus.
Collapse
Affiliation(s)
- Liang Song
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yiming Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Junqing Long
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yuanfan Li
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Zongqin Pan
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Fang Fang
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yu Long
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Cheng Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Nana Qi
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yajie Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Hongwei Xia
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China.
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, Guangxi, 530021, China; Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, 530021, China.
| |
Collapse
|
16
|
Huang J, Hagberg Thulin M, Damber JE, Welén K. The roles of RUNX2 and osteoclasts in regulating expression of steroidogenic enzymes in castration-resistant prostate cancer cells. Mol Cell Endocrinol 2021; 535:111380. [PMID: 34216642 DOI: 10.1016/j.mce.2021.111380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 06/08/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022]
Abstract
Intratumoral steroidogenesis is involved in development of castration-resistant prostate cancer (CRPC) as bone metastases. The osteoblast transcription factor RUNX2 influences steroidogenesis and is induced in CRPC cells by osteoblasts. This study investigates osteoclastic influence on RUNX2 in intratumoral steroidogenesis. Steroidogenic enzymes and steroid receptors were detected with immunohistochemistry in xenograft intratibial tumors from CRPC cells. In vitro, expression of RUNX2 was increased by osteoclasts in osteoblastic LNCaP-19 cells, but not in osteolytic PC-3. Silencing of RUNX2 downregulates expression of CYP11A1, CYP17A1 and HSD3B1 in LNCaP-19 cells co-cultured with osteoclasts, leading to inhibition of KLK3 expression. Osteoclasts promoted CYP11A1 and RUNX2 promoted AKR1C3, HSD17B3 and CYP19A1, but suppressed ESR2 in PC-3 cells. This study shows that osteoclasts promote RUNX2 regulated induction of key steroidogenic enzymes, influencing activation of androgen receptor in CRPC cells. The potential of RUNX2 as a target to inhibit progression of skeletal metastases of CRPC needs further investigation.
Collapse
Affiliation(s)
- Junchi Huang
- Department of Urology, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Malin Hagberg Thulin
- Department of Urology, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden; Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jan-Erik Damber
- Department of Urology, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Urology, Gothenburg, Sweden
| | - Karin Welén
- Department of Urology, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
17
|
El-Kenawi A, Dominguez-Viqueira W, Liu M, Awasthi S, Abraham-Miranda J, Keske A, Steiner KK, Noel L, Serna AN, Dhillon J, Gillies RJ, Yu X, Koomen JM, Yamoah K, Gatenby RA, Ruffell B. Macrophage-derived cholesterol contributes to therapeutic resistance in prostate cancer. Cancer Res 2021; 81:5477-5490. [PMID: 34301759 DOI: 10.1158/0008-5472.can-20-4028] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/16/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Castration-resistant prostate cancer (CRPC) is a lethal stage of disease in which androgen receptor (AR) signaling is persistent despite androgen deprivation therapy (ADT). Most studies have focused on investigating cell-autonomous alterations in CRPC, while the contributions of the tumor microenvironment are less well understood. Here we sought to determine the role of tumor-associated macrophages in CRPC, based upon their role in cancer progression and therapeutic resistance. In a syngeneic model that reflected the mutational landscape of CRPC, macrophage depletion resulted in a reduced transcriptional signature for steroid and bile acid synthesis, indicating potential perturbation of cholesterol metabolism. As cholesterol is the precursor of the five major types of steroid hormones, we hypothesized that macrophages were regulating androgen biosynthesis within the prostate tumor microenvironment. Macrophage depletion reduced androgen levels within prostate tumors and restricted androgen receptor (AR) nuclear localization in vitro and in vivo. Macrophages were also cholesterol-rich and were able to transfer cholesterol to tumor cells in vitro. AR nuclear translocation was inhibited by activation of Liver X Receptor (LXR)-β, the master regulator of cholesterol homeostasis. Consistent with these data, macrophage depletion extended survival during ADT and the presence of macrophages correlated with therapeutic resistance in patient-derived explants. Taken together, these findings support the therapeutic targeting of macrophages in CRPC.
Collapse
Affiliation(s)
- Asmaa El-Kenawi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Min Liu
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Shivanshu Awasthi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julieta Abraham-Miranda
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aysenur Keske
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - KayLee K Steiner
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Leenil Noel
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amparo N Serna
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jasreman Dhillon
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert J Gillies
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kosj Yamoah
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Robert A Gatenby
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
18
|
Pang J, Dai L, Zhang C, Zhang Q. MiR-373 Inhibits the Epithelial-Mesenchymal Transition of Prostatic Cancer via Targeting Runt-Related Transcription Factor 2. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6974225. [PMID: 34257854 PMCID: PMC8260310 DOI: 10.1155/2021/6974225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/20/2021] [Accepted: 06/20/2021] [Indexed: 12/23/2022]
Abstract
Prostatic cancer (PCa) is a prevalent form of malignancy based on its high associated levels of mortality and morbidity across the world. MicroRNAs (miRNAs) are significant in the advancement of prostatic cancer. The current study is aimed at exploring the potential roles of miR-373 in PCa. In turn, the study conducted a qRT-PCR test to determine the levels of mRNA. A western blot test was also executed in determining the protein level. The processes of transwell assay and wound healing were integrated in the detection of the potential for PCa cells to invade and migrate. The integration of dual luciferase reporter assay is critical in determining the levels of luciferase activity among prostatic cancer cells. Then, the results showed a net decrease of miR-373 within prostatic cancer cells and tissues. Upregulated miR-373 reduced the invasion and migration potential of PCa cells. Moreover, overexpressed miR-373 increased the levels of E-cadherin and FSP1 as epithelial cell markers. Similarly, the overregulation of miR-373 brought about the upregulation of mesenchymal markers (N-cadherin, Snail, and vimentin). The study predicted runt-related transcription factor 2 (RUNX2) to be a target of miR-373. The luciferase activity of PCa cells was decreased after the cotransfection with miR-373 mimics and RUNX2 3' untranslated region (3'UTR) wild type (WT). Moreover, RUNX2 became upregulated in PCa cells and tissues. The upregulation of miR-373 decreased the mRNA and protein level of RUNX2. However, overexpressed RUNX2 abated the roles of miR-373 in the intrusion and migration of PCa cells and in regulating the expression of epithelial cell markers and mesenchymal markers. In short, miR-373 may regulate the EMT of PCa cells via targeting RUNX2. The miR-373/RUNX2 axis provides a therapeutic target for PCa.
Collapse
Affiliation(s)
- Jianyi Pang
- Department of Urology Surgery, The Affiliated Weihai Second Municipal Hospital of Qingdao University, Weihai, Shandong 264200, China
| | - Limei Dai
- Department of Dermatology and STD, The Affiliated Weihai Second Municipal Hospital of Qingdao University, Weihai, Shandong 264200, China
| | - Chen Zhang
- Department of Urology Surgery, The Affiliated Tengzhou Central People's Hospital of Jining Medical University, Tengzhou, Shandong 277500, China
| | - Qinglei Zhang
- Department of Urology Surgery, The Affiliated Tengzhou Central People's Hospital of Jining Medical University, Tengzhou, Shandong 277500, China
| |
Collapse
|
19
|
Integrated bioinformatics analysis revealed the regulation of angiogenesis by tumor cells in hepatocellular carcinoma. Biosci Rep 2021; 41:229066. [PMID: 34151937 PMCID: PMC8252189 DOI: 10.1042/bsr20210126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/09/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer mortality, metastasis accounts for most of the cases. Angiogenesis plays an important role in cancer metastasis, but how tumor cells affect the function of endothelial cells by dictating their microRNA (miRNA) expression remains largely unknown. Differentially expressed miRNAs (DEMs) were identified through dataset downloaded from the Gene Expression Omnibus (GEO) database and analyzed by GEO2R. We then used online tools to obtain potential targets of candidate miRNAs and functional enrichment analysis, as well as the protein-protein interaction (PPI). Finally, the function of miR-302c-3p was validated through in vitro assay. In the current study, we found that HCC cells altered miRNA expression profiles of human umbilical vein endothelial cells (HUVECs) and miR-302c-3p was the most down-regulated miRNA in HUVECs when they were co-cultured with HCC-LM3 cells. Functional enrichment analysis of the candidate targets revealed that these genes were involved in epigenetic regulation of gene expression, in particular, cytosine methylation. In addition, PPI network demonstrated distinct roles of genes targeted by miR-302c-3p. Importantly, inhibition of angiogenesis, migration and permeability by the most down-regulated miR-302c-3p in HUVECs was confirmed in vitro. These findings brought us novel insight into the regulation of angiogenesis by HCC cells and provided potential targets for the development of therapeutic strategies.
Collapse
|
20
|
Kumar S, Singh SK, Rana B, Rana A. Tumor-infiltrating CD8 + T cell antitumor efficacy and exhaustion: molecular insights. Drug Discov Today 2021; 26:951-967. [PMID: 33450394 PMCID: PMC8131230 DOI: 10.1016/j.drudis.2021.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/20/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023]
Abstract
Host immunity has an essential role in the clinical management of cancers. Therefore, it is advantageous to choose therapies that can promote tumor cell death and concurrently boost host immunity. The dynamic tumor microenvironment (TME) determines whether an antineoplastic drug will elicit favorable or disparaging immune responses from tumor-infiltrating lymphocytes (TILs). CD8+ T cells are one of the primary tumor-infiltrating immune cells that deliver antitumor responses. Here, we review the influence of various factors in the TME on CD8+ T cell exhaustion and survival, and possible strategies for restoring CD8+ T cell effector function through immunotherapy.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA.
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
21
|
Yu H, Liu J, Zhang Z, Zhu Y, Bi J, Kong C. SNHG12 promotes carcinogenesis of human renal cell cancer via functioning as a competing endogenous RNA and sponging miR-30a-3p. J Cell Mol Med 2021; 25:4696-4708. [PMID: 33787057 PMCID: PMC8107103 DOI: 10.1111/jcmm.16417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022] Open
Abstract
Small nucleolar RNA host gene 12 (SNHG12) has been indicated in the tumorigenesis of various human cancers, including clear cell renal cell carcinoma (ccRCC). However, the underlying mechanisms of SNHG12 driving progression of ccRCC remain incompletely understood. In the present study, we discovered that SNHG12 is up-regulated in ccRCC and that overexpression of SNHG12 predicted poor clinical outcome of ccRCC patients. SNHG12 knockdown notably inhibited proliferation and migration of RCC cells. Furthermore, we discovered that miR-30a-3p, a putative ccRCC inhibitor, was competitively sponged by SNHG12. Via the crosstalk network, SNHG12 was capable of up-regulating multiple target genes of miR-30a-3p, namely, RUNX2, WNT2 and IGF-1R, which have been identified to facilitate tumorigenesis of ccRCC. Taken together, our present study suggested a novel ceRNA network, in which SNHG12 could promote the malignancy of ccRCC although competitively binding with miR-30a-3p and consequently release the expression of its downstream cancer-related genes.
Collapse
Affiliation(s)
- Hongyuan Yu
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| | - Junlong Liu
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| | - Zhe Zhang
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| | - Yuyan Zhu
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| | - Jianbin Bi
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| | - Chuize Kong
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Shi L, Yan Y, He Y, Yan B, Pan Y, Orme JJ, Zhang J, Xu W, Pang J, Huang H. Mutated SPOP E3 Ligase Promotes 17βHSD4 Protein Degradation to Drive Androgenesis and Prostate Cancer Progression. Cancer Res 2021; 81:3593-3606. [PMID: 33762355 DOI: 10.1158/0008-5472.can-20-3258] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/01/2021] [Accepted: 03/22/2021] [Indexed: 11/16/2022]
Abstract
Molecular mechanisms underlying intratumoral androgenesis and aberrant androgen receptor (AR) activation in prostate cancer remain poorly understood. Here we demonstrate that ectopic expression of the E3 ubiquitin ligase adaptor speckle-type poxvirus and zinc finger domain protein (SPOP) stabilizes 17βHSD4. SPOP bound a functional substrate-binding consensus (SBC) motif 315RATST319 in 17βHSD4 and promoted nondegradable K27- and K29-linked polyubiquitination of 17βHSD4. The effect of SPOP was antagonized by serum- and glucocorticoid kinase-3 (SGK3)-mediated phosphorylation of serine 318 (S318) in the SBC and S318 phosphorylation-dependent binding of SKP2 E3 ligase and subsequent K48-linked polyubiquitination and proteasomal degradation of 17βHSD4. Prostate cancer-associated SPOP mutations impaired the SPOP-17βHSD4 interaction, caused 17βHSD4 protein destruction in prostate cancer cells in culture and patient specimens, and increased testosterone production and prostate cancer cell growth in vitro and in mouse models. Thus, we have identified SPOP and SKP2 as two essential E3 ubiquitin ligases that exert opposite effects on 17βHSD4 protein degradation and intratumoral androgenesis in prostate cancer cells. We further demonstrate that SPOP mutations or SKP2 overexpression contribute to prostate cancer progression by decreasing 17βHSD4 expression and increasing intratumoral androgen synthesis. SIGNIFICANCE: This study reveals a novel mechanism of aberrant AR activation in SPOP-mutated prostate cancer and uncovers putative biomarkers for effective treatment by AR-targeted therapies.
Collapse
Affiliation(s)
- Lei Shi
- Department of Radiation Oncology, the Fourth Hospital of Harbin Medical University, Harbin, China.,Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yundong He
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Binyuan Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota.,Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jacob J Orme
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Scottsdale, Arizona
| | - Wanhai Xu
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota. .,Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota.,Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
23
|
Zhao W, Yang H, Chai J, Xing L. RUNX2 as a promising therapeutic target for malignant tumors. Cancer Manag Res 2021; 13:2539-2548. [PMID: 33758548 PMCID: PMC7981165 DOI: 10.2147/cmar.s302173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/27/2021] [Indexed: 12/16/2022] Open
Abstract
The transcription factor runt-related protein 2 (RUNX2) has an important impact on the transformation of bone marrow mesenchymal stem cells to osteoblasts. Further studies have shown that RUNX2 plays a key role in the invasion and metastasis of cancers. RUNX2 is a "key" molecule in the regulatory network comprised of multiple signaling pathways upstream and its target downstream molecules. Due to the complex regulatory mechanisms of RUNX2, the specific mechanism underlying the occurrence, development and prognosis of malignant tumors has not been fully understood. Currently, RUNX2 as a promising therapeutic target for cancers has become a research hotspot. Herein, we reviewed the current literature on the modulatory functions and mechanisms of RUNX2 in the development of malignant tumors, aiming to explore its potential clinical application in the diagnosis, prognosis and treatment of tumors.
Collapse
Affiliation(s)
- Weizhu Zhao
- Department of Radiology, Cancer Hospital Affiliated to Shandong First Medical University, Shandong Cancer Hospital and Institute, Jinan, 250117, People’s Republic of China
- Department of Oncology, Binzhou People’s Hospital, Binzhou, 256610, People’s Republic of China
| | - Haiying Yang
- Department of Nursing, Binzhou People’s Hospital, Binzhou, 256610, People’s Republic of China
| | - Jie Chai
- Department of Gastrointestinal Surgery, Cancer Hospital Affiliated to Shandong First Medical University, Shandong Cancer Hospital and Institute, Jinan, 250117, People’s Republic of China
| | - Ligang Xing
- Department of Radiology, Cancer Hospital Affiliated to Shandong First Medical University, Shandong Cancer Hospital and Institute, Jinan, 250117, People’s Republic of China
| |
Collapse
|
24
|
Ashe H, Krakowiak P, Hasterok S, Sleppy R, Roller DG, Gioeli D. Role of the runt-related transcription factor (RUNX) family in prostate cancer. FEBS J 2021; 288:6112-6126. [PMID: 33682350 DOI: 10.1111/febs.15804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/22/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Prostate cancer (PCa) is a very complex disease that is a major cause of death in men worldwide. Currently, PCa dependence on the androgen receptor (AR) has resulted in use of AR antagonists and antiandrogen therapies that reduce endogenous steroid hormone production. However, within two to three years of receiving first-line androgen deprivation therapy, the majority of patients diagnosed with PCa progress to castration-resistant prostate cancer (CRPC). There is an urgent need for therapies that are more durable than antagonism of the AR axis. Studies of runt-related transcription factors (RUNX) and their heterodimerization partner, core-binding factor subunit b (CBFβ), are revealing that the RUNX family are drivers of CRPC. In this review, we describe what is presently understood about RUNX members in PCa, including what regulates and is regulated by RUNX proteins, and the role of RUNX proteins in the tumor microenvironment and AR signaling. We discuss the implications for therapeutically targeting RUNX, the potential for RUNX as PCa biomarkers, and the current pressing questions in the field.
Collapse
Affiliation(s)
- Hannah Ashe
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Patryk Krakowiak
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Sylwia Hasterok
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Rosalie Sleppy
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Devin G Roller
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Daniel Gioeli
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
25
|
Gao Z, Ye X, Bordeaux A, Hettich S, Lin S, Han F, Jia Y. miR-26b regulates cell proliferation and apoptosis of CD117+CD44+ ovarian cancer stem cells by targeting PTEN. Eur J Histochem 2021; 65:3186. [PMID: 33634678 PMCID: PMC7883108 DOI: 10.4081/ejh.2021.3186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OC) is the one of the most common cancer in women globally. However, it still represents the most dangerous gynecologic malignancy even with the advances in detection and therapeutics. Thus, there is an urgent need in finding more effective therapeutic options for OC patients including cancer stem cells (CSC). MicroRNAs (miRNAs) are small, endogenous, and non-coding RNAs that play critical roles in the progression of various types of tumor. Our aim of this study was to find the regulatory function of microRNA-26 (miRNA-26b) on the cell proliferation and apoptosis of ovarian CSCs. Our studies show that miR-26b is under-regulated in human CD117+CD44+ ovarian CSCs. The miR-26b overexpression inhibits the cell proliferation and promotes cell apoptosis. Moreover, phosphatase and tensin homolog (PTEN) is found to be a functional target of miR-26b. Moreover, PTEN overexpression reversed the effects of miR-26b on the cell proliferation and apoptosis. PTEN overexpression remarkably accelerated the cell proliferation, and inhibited cell apoptosis. These results indicate that miR-26b regulates cell proliferation and apoptosis of CD117+CD44+ ovarian CSCs by targeting PTEN.>.
Collapse
Affiliation(s)
- Zubiao Gao
- Department of Obstetrics and Gynecology, Foshan Chancheng Central Hospital, Foshan, Guangdong.
| | - Xiaofeng Ye
- Department of Obstetrics and Gynecology, Foshan Chancheng Central Hospital, Foshan, Guangdong.
| | - Anne Bordeaux
- Department of Pathology, Medical Center, University of Freiburg, Baden-Wuerttemberg.
| | - Stanka Hettich
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Baden-Wuerttemberg.
| | - Siyao Lin
- Department of Obstetrics and Gynecology, Foshan Chancheng Central Hospital, Foshan, Guangdong.
| | - Fang Han
- Department of Obstetrics and Gynecology, Foshan Chancheng Central Hospital, Foshan, Guangdong.
| | - Yan Jia
- Department of Reproductive Immunology, Chengdu Xi' nan Gynecology Hospital, Chengdu, Sichuan.
| |
Collapse
|
26
|
Cancer-associated fibroblasts and the related Runt-related transcription factor 2 (RUNX2) promote bladder cancer progression. Gene 2021; 775:145451. [PMID: 33482279 DOI: 10.1016/j.gene.2021.145451] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/07/2021] [Accepted: 01/15/2021] [Indexed: 11/20/2022]
Abstract
Bladder urothelial cancer (BLCA) has a high incidence worldwide. Cancer-associated fibroblasts (CAFs) in the tumor microenvironment are gradually recognized to play an important role in the occurrence and progression of cancer. However, the research on BLCA CAFs is still in its infancy, and the CAFs related genes are still unclear. We used the identified BLCA-specific CAFs gene signature in our previous work to calculate the CAFs infiltration score of the sample. Furthermore, we used data from multiple public databases to prove that CAFs high infiltration is associated with tumor progression and poor prognosis. In order to select the powerful genes in BLCA that are related to CAFs infiltration and affect prognosis, we chose transcription factors as the research object, and finally defined RUNX2 as the candidate gene for functional verification. In the immunohistochemical images, tissues with higher RUNX2 expression also had deeper staining of CAFs markers. We used public databases and collected specimens to prove that RUNX2 is overexpressed at the mRNA and protein levels in BLCA tissues. Through functional enrichment analysis, RUNX2 is mainly related to epithelialmesenchymal transition and extracellular matrix. Finally, we knocked down RUNX2 in vitro and observed a significant decrease in the metastasis and proliferation ability. In conclusion, high infiltration of CAFs is associated with tumor progression and poor prognosis in BLCA. RUNX2 is a transcription factor related to CAFs, which is overexpressed in bladder cancer and affects the prognosis. RUNX2 is a potential marker relating CAFs and therapy target in BLCA.
Collapse
|
27
|
Turnham DJ, Bullock N, Dass MS, Staffurth JN, Pearson HB. The PTEN Conundrum: How to Target PTEN-Deficient Prostate Cancer. Cells 2020; 9:E2342. [PMID: 33105713 PMCID: PMC7690430 DOI: 10.3390/cells9112342] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Loss of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which negatively regulates the PI3K-AKT-mTOR pathway, is strongly linked to advanced prostate cancer progression and poor clinical outcome. Accordingly, several therapeutic approaches are currently being explored to combat PTEN-deficient tumors. These include classical inhibition of the PI3K-AKT-mTOR signaling network, as well as new approaches that restore PTEN function, or target PTEN regulation of chromosome stability, DNA damage repair and the tumor microenvironment. While targeting PTEN-deficient prostate cancer remains a clinical challenge, new advances in the field of precision medicine indicate that PTEN loss provides a valuable biomarker to stratify prostate cancer patients for treatments, which may improve overall outcome. Here, we discuss the clinical implications of PTEN loss in the management of prostate cancer and review recent therapeutic advances in targeting PTEN-deficient prostate cancer. Deepening our understanding of how PTEN loss contributes to prostate cancer growth and therapeutic resistance will inform the design of future clinical studies and precision-medicine strategies that will ultimately improve patient care.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - Nicholas Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Manisha S. Dass
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| |
Collapse
|
28
|
Aquila S, Santoro M, Caputo A, Panno ML, Pezzi V, De Amicis F. The Tumor Suppressor PTEN as Molecular Switch Node Regulating Cell Metabolism and Autophagy: Implications in Immune System and Tumor Microenvironment. Cells 2020; 9:cells9071725. [PMID: 32708484 PMCID: PMC7408239 DOI: 10.3390/cells9071725] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Recent studies conducted over the past 10 years evidence the intriguing role of the tumor suppressor gene Phosphatase and Tensin Homolog deleted on Chromosome 10 PTEN in the regulation of cellular energy expenditure, together with its capability to modulate proliferation and survival, thus expanding our knowledge of its physiological functions. Transgenic PTEN mice models are resistant to oncogenic transformation, present decreased adiposity and reduced cellular glucose and glutamine uptake, together with increased mitochondrial oxidative phosphorylation. These acquisitions led to a novel understanding regarding the role of PTEN to counteract cancer cell metabolic reprogramming. Particularly, PTEN drives an “anti-Warburg state” in which less glucose is taken up, but it is more efficiently directed to the mitochondrial Krebs cycle. The maintenance of cellular homeostasis together with reduction of metabolic stress are controlled by specific pathways among which autophagy, a catabolic process strictly governed by mTOR and PTEN. Besides, a role of PTEN in metabolic reprogramming and tumor/stroma interactions in cancer models, has recently been established. The genetic inactivation of PTEN in stromal fibroblasts of mouse mammary glands, accelerates breast cancer initiation and progression. This review will discuss our novel understanding in the molecular connection between cell metabolism and autophagy by PTEN, highlighting novel implications regarding tumor/stroma/immune system interplay. The newly discovered action of PTEN opens innovative avenues for investigations relevant to counteract cancer development and progression.
Collapse
Affiliation(s)
- Saveria Aquila
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Marta Santoro
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Annalisa Caputo
- Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
- Health Center, University of Calabria, 87036 Rende, Italy
- Correspondence:
| |
Collapse
|
29
|
Zhang L, Liu L, Xu X, He X, Wang G, Fan C, Zheng Q, Li F. miR-205/RunX2 axis negatively regulates CD44 +/CD24 - breast cancer stem cell activity. Am J Cancer Res 2020; 10:1871-1887. [PMID: 32642297 PMCID: PMC7339278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/31/2020] [Indexed: 06/11/2023] Open
Abstract
Breast Cancer stem cells (BCSCs) have been extensively studied and have been used directly as a therapeutic target, but how the BCSCs themselves are regulated remain unclear. Here we reported identification of miR-205 that may act as a tumor suppressor and negatively-regulate BCSCs stemness and tumor malignance. By qRT-PCR analysis, we have shown that miR-205 was decreased in CD44+/CD24-/low BCSCs compared with non-BCSCs. We have also shown that miR-205 expression level was very low in MB-231 cells with high BCSC percentage, while relatively high in MCF-7 cells with low BCSC percentage. We then overexpressed miR-205 in MB-231 and SUM-149 cells and knocked it down in MCF-7 and BT-474 cells respectively. Our results showed that overexpression of miR-205 could reduce CD44+/CD24-/low population percentage in MB-231 cells. The mechanism might associate with mesenchymal-epithelial transition (MET). Finally, we found an important transcriptional factor and oncogene, RunX2, was a target gene of miR-205. miR-205 overexpression could inhibit breast cancer malignancy by regulating RunX2 both in vitro and in vivo. A rescue experiment by cotransfection of RunX2 and miR-205 into the MCF-7 cell line attenuate cell proliferation, invasion, migration, CD44+/CD24-/low population, mammosphere formation abilities and xengraft tumor formation. Together, our results support that miR-205 is a tumor suppressor during breast cancer development.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Pathophysiology, Basic Medical School, Anhui Medical UniversityHefei 230032, Anhui, China
| | - Lei Liu
- Department of Emergency Surgery, Fuyang Hospital of Anhui Medical UniversityFuyang 236000, China
| | - Xiaodan Xu
- Department of Pathophysiology, Basic Medical School, Anhui Medical UniversityHefei 230032, Anhui, China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical UniversityHefei 230032, China
| | - Xiaogang He
- Department of Pathophysiology, Basic Medical School, Anhui Medical UniversityHefei 230032, Anhui, China
| | - Gang Wang
- Department of Pathophysiology, Basic Medical School, Anhui Medical UniversityHefei 230032, Anhui, China
| | - Chulin Fan
- Department of Pathophysiology, Basic Medical School, Anhui Medical UniversityHefei 230032, Anhui, China
| | - Qiping Zheng
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University ZhenjiangZhenjiang 212013, Jiangsu, China
- Shenzhen Academy of Peptide Targeting Technology at Pingshan, Shenzhen Tyercan Bio-pharm Co., Ltd.Shenzhen 518118, China
| | - Feifei Li
- Department of Pathophysiology, Basic Medical School, Anhui Medical UniversityHefei 230032, Anhui, China
| |
Collapse
|
30
|
Alwanian WM, Tyner AL. Protein tyrosine kinase 6 signaling in prostate cancer. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2020; 8:1-8. [PMID: 32211448 PMCID: PMC7076292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 06/10/2023]
Abstract
More than 25 years have passed since the discovery of protein tyrosine kinase 6 (PTK6), a non-receptor tyrosine kinase distantly related to SRC family kinases. Since then, a variety of data suggest that PTK6 promotes oncogenic signaling and tumorigenesis, generally dependent on its kinase activity. Increased PTK6 expression, activation at the plasma membrane and altered intracellular localization have been discovered in prostate cancers. While PTK6 is localized to nuclei of epithelial cells in normal prostate, it is relocalized and activated at the plasma membrane in prostate tumors. Active PTK6 interacts with and directly phosphorylates AKT, FAK and BCAR1 to promote oncogenic signaling. Furthermore, PTK6 can enhance the epithelial mesenchymal transition by inhibiting E-cadherin expression and inducing expression of the mesenchymal markers vimentin, SLUG and ZEB1. Several lines of evidence suggest that PTK6 plays a role in Pten null prostate tumors. PTEN targets activating phosphorylation of PTK6 and loss of PTEN subsequently leads to PTK6 activation. Different studies provide compelling evidence as to why PTK6 is a potential therapeutic target in prostate cancer. Here, we briefly review the advances and significance of PTK6 in prostate cancer.
Collapse
Affiliation(s)
- Wanian M Alwanian
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago Chicago, IL, The United States
| | - Angela L Tyner
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago Chicago, IL, The United States
| |
Collapse
|
31
|
Yan DY, Tang J, Chen L, Wang B, Weng S, Xie Z, Wu ZY, Shen Z, Bai B, Yang L. Imperatorin promotes osteogenesis and suppresses osteoclast by activating AKT/GSK3 β/β-catenin pathways. J Cell Mol Med 2019; 24:2330-2341. [PMID: 31883297 PMCID: PMC7011130 DOI: 10.1111/jcmm.14915] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/26/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Osteoporosis is caused by disturbance in the dynamic balance of bone remodelling, a physiological process, vital for maintenance of healthy bone tissue in adult humans. In this process, a new bone is formed by osteoblasts and the pre‐existing bone matrix is resorbed by osteoclasts. Imperatorin, a widely available and inexpensive plant extract with antioxidative and apoptotic effects, is reported to treat osteoporosis. However, the underlying mechanism and specific effects on bone metabolism have not been elucidated. In this study, we used rat bone marrow‐derived mesenchymal stem cells and found that imperatorin can activate RUNX2, COL1A1 and osteocalcin by promoting the Ser9 phosphorylation of GSK3β and entry of β‐catenin into the nucleus. Imperatorin also enhanced the production of phospho‐AKT (Ser473), an upstream factor that promotes the Ser9 phosphorylation of GSK3β. We used ipatasertib, a pan‐AKT inhibitor, to inhibit the osteogenic effect of imperatorin, and found that imperatorin promotes osteogenesis via AKT/GSK3β/β‐catenin pathway. Next, we used rat bone marrow‐derived monocytes, to check whether imperatorin inhibits osteoclast differentiation via AKT/GSK3β/β‐catenin pathway. Further, we removed the bilateral ovaries of rats to establish an osteoporotic model. Intragastric administration of imperatorin promoted osteogenesis and inhibited osteoclast in vivo. Our experiments showed that imperatorin is a potential drug for osteoporosis treatment.
Collapse
Affiliation(s)
- De-Yi Yan
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiahao Tang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bingzhang Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sheji Weng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongjie Xie
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zong-Yi Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zijian Shen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bingli Bai
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lei Yang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
32
|
Ouyang P, Wu K, Su L, An W, Bie Y, Zhang H, Kang H, Jiang E, Zhu W, Yao Y, Hu X, Chen Z, Wang S. Inhibition of human cervical cancer cell invasion by IL-37 involving runt related transcription factor 2 suppression. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:568. [PMID: 31807549 DOI: 10.21037/atm.2019.09.38] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background IL-37 is a newly anti-inflammatory cytokine whose function is largely unknown in cancer. Our preliminary experiment found IL-37 could inhibit the invasion of human cervical cancer (CC) cells and influence the expression of RUNX family whose function was also unclear in CC. The present study aims to further investigate the effects of IL-37 on cell invasion and runt related transcription factor 2 (RUNX2) expression in CC cell lines. Methods Firstly, plasmid overexpressing IL-37 or RUNX2 was transfected into Siha and C33A cells by Hilymax. Then, the effects of IL-37 on the mRNA expression of RUNX1, RUNX2 and RUNX3 gene were detected by quantitative real-time polymerase chain reaction. Protein expression was measured by Western blot and the grayscale scanning analysis. Finally, the effects of IL-37 or RUNX2 on cell invasion were tested by transwell assay. Results IL-37 inhibited the mRNA expression of RUNX1 and RUNX2, and increased that of RUNX3 in CC cells. Among the three RUNX genes, RUNX2 showed the most significant change in mRNA expression (decreased by78.5% in Siha cells and by 61.5% in C33A cells) and thus was chosen for the following study. Overexpressed IL-37 inhibited cell invasion by 36.23% in Siha cells (P<0.05) and 26.21% in C33A cells (P<0.01). Overexpression of RUNX2 promoted cell invasion. Up-regulation of IL-37 suppressed markedly the mRNA and protein expression of RUNX2. Furthermore, overexpressed RUNX2 partially restored the inhibited cell invasion by IL-37 to 86.62% in Siha cells (P<0.01) and 87.08% in C33A cells (P<0.01). Conclusions IL-37 can significantly inhibit the cell invasion of Siha and C33A cells, which involves the suppression of RUNX2.
Collapse
Affiliation(s)
- Ping Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China
| | - Kun Wu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China.,Department of Histology and Embryology, Basic Medical College, Guangdong Medical University, Dongguan 523808, China
| | - Liudan Su
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China.,Department of Histology and Embryology, Basic Medical College, Guangdong Medical University, Dongguan 523808, China
| | - Weifang An
- Department of Histology and Embryology, Basic Medical College, Guangdong Medical University, Dongguan 523808, China
| | - Yanhong Bie
- Department of Histology and Embryology, Basic Medical College, Guangdong Medical University, Dongguan 523808, China
| | - He Zhang
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Haixian Kang
- Department of Histology and Embryology, Basic Medical College, Guangdong Medical University, Dongguan 523808, China
| | - Enping Jiang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China
| | - Wei Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China
| | - Yunhong Yao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China
| | - Xinrong Hu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China
| | - Zhangquan Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China
| | - Sen Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan 523808, China.,Department of Histology and Embryology, Basic Medical College, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
33
|
Sawada Y, Kikugawa T, Iio H, Sakakibara I, Yoshida S, Ikedo A, Yanagihara Y, Saeki N, Győrffy B, Kishida T, Okubo Y, Nakamura Y, Miyagi Y, Saika T, Imai Y. GPRC5A facilitates cell proliferation through cell cycle regulation and correlates with bone metastasis in prostate cancer. Int J Cancer 2019; 146:1369-1382. [PMID: 31276604 DOI: 10.1002/ijc.32554] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022]
Abstract
The prognosis of patients with progressive prostate cancers that are hormone refractory and/or have bone metastasis is poor. Multiple therapeutic targets to improve prostate cancer patient survival have been investigated, including orphan GPCRs. In our study, we identified G Protein-Coupled Receptor Class C Group 5 Member A (GPRC5A) as a candidate therapeutic molecule using integrative gene expression analyses of registered data sets for prostate cancer cell lines. Kaplan-Meier analysis of TCGA data sets revealed that patients who have high GPRC5A expression had significantly shorter overall survival. PC3 prostate cancer cells with CRISPR/Cas9-mediated GPRC5A knockout exhibited significantly reduced cell proliferation both in vitro and in vivo. RNA-seq revealed that GPRC5A KO PC3 cells had dysregulated expression of cell cycle-related genes, leading to cell cycle arrest at the G2/M phase. Furthermore, the registered gene expression profile data set showed that the expression level of GPRC5A in original lesions of prostate cancer patients with bone metastasis was higher than that without bone metastasis. In fact, GPRC5A KO PC3 cells failed to establish bone metastasis in xenograft mice models. In addition, our clinical study revealed that GPRC5A expression levels in prostate cancer patient samples were significantly correlated with bone metastasis as well as the patient's Gleason score (GS). Combined assessment with the immunoreactivity of GPRC5A and GS displayed higher specificity for predicting the occurrence of bone metastasis. Together, our findings indicate that GPRC5A can be a possible therapeutic target and prognostic marker molecule for progressive prostate cancer.
Collapse
Affiliation(s)
- Yuichiro Sawada
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Japan.,Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Japan.,Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Tadahiko Kikugawa
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Hiroyuki Iio
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Japan.,Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Japan.,Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Iori Sakakibara
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shuhei Yoshida
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Aoi Ikedo
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Yuta Yanagihara
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Japan.,Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Japan.,Division of Laboratory Animal Research, Advanced Research Support Center, Ehime University, Toon, Japan
| | - Noritaka Saeki
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Japan.,Division of Laboratory Animal Research, Advanced Research Support Center, Ehime University, Toon, Japan
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary.,Semmelweis University 2nd Dept. of Pediatrics, Budapest, Hungary
| | - Takeshi Kishida
- Department of Urology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yoichiro Okubo
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Takashi Saika
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yuuki Imai
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Japan.,Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Japan.,Division of Laboratory Animal Research, Advanced Research Support Center, Ehime University, Toon, Japan
| |
Collapse
|
34
|
Honda M, Kimura T, Kamata Y, Tashiro K, Kimura S, Koike Y, Sato S, Yorozu T, Furusato B, Takahashi H, Kiyota H, Egawa S. Differential expression of androgen receptor variants in hormone-sensitive prostate cancer xenografts, castration-resistant sublines, and patient specimens according to the treatment sequence. Prostate 2019; 79:1043-1052. [PMID: 30998834 DOI: 10.1002/pros.23816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/08/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Androgen receptor variants (AR-vs), especially AR-v7 and AR-v 5, 6, and 7 exon-skipped (AR-v567es), are reportedly key players in the development of castration-resistant prostate cancer (CRPC). We previously established a mouse xenograft model (JDCaP) from a metastatic skin lesion from a Japanese patient with CRPC and that was revealed to exhibit androgen sensitivity. In the present study, we established multiple castration-resistant xenograft models from JDCaP mice to investigate the biological features of CRPC. METHODS Tissue from JDCaP mice was transplanted into male and female nude mice, and after serial passaging, castration-resistant sublines (JDCaP-CR2M and JDCaP-CR4M in male mice, JDCaP-CR2F and JDCaP-CR4F in female mice) were established. We investigated anti-androgen and testosterone sensitivity and the messenger RNA expression pattern of full-length AR and AR-vs. In addition, we compared AR protein levels of patient specimens among primary, local-recurrent, and two skin-metastatic tumors. RESULTS All JDCaP-CR sublines showed continuous growth following the administration of bicalutamide, although the effects of testosterone varied among sublines. Parental JDCaP and JDCaP-CR2M, JDCaP-CR4M, and JDCaP-CR4F sublines expressed AR-v7, whereas JDCaP-CR2F exhibited elevated AR-v567es expression resulting from genomic deletion, which was confirmed by DNA sequencing. Moreover, we confirmed AR-v7 expression in the tumor of the original patient after androgen-deprivation therapy. CONCLUSIONS Each JDCaP-CR subline showed different AR-v-expression patterns, with JDCaP-CR2F expressing AR-v567es due to genomic deletion. Our results indicated that AR-vs emerged after androgen-deprivation therapy and appeared essential for acquisition of castration resistance.
Collapse
Affiliation(s)
- Mariko Honda
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kamata
- Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Kojiro Tashiro
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shoji Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Koike
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shun Sato
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yorozu
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Bungo Furusato
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Kiyota
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shin Egawa
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Bai Y, Yang Y, Yan Y, Zhong J, Blee AM, Pan Y, Ma T, Karnes RJ, Jimenez R, Xu W, Huang H. RUNX2 overexpression and PTEN haploinsufficiency cooperate to promote CXCR7 expression and cellular trafficking, AKT hyperactivation and prostate tumorigenesis. Theranostics 2019; 9:3459-3475. [PMID: 31281490 PMCID: PMC6587168 DOI: 10.7150/thno.33292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Rationale: The overall success rate of prostate cancer (PCa) diagnosis and therapy has been improved over the years. However, genomic and phenotypic heterogeneity remains a major challenge for effective detection and treatment of PCa. Efforts to better classify PCa into functional subtypes and elucidate the molecular mechanisms underlying prostate tumorigenesis and therapy resistance are warranted for further improvement of PCa outcomes. Methods: We generated Cre+;Runx2-cTg;Ptenp/+ (Runx2-Pten double mutant) mice by crossbreeding Cre+;Runx2-cTg males with Pten conditional (Ptenp/p) females. By using Hematoxylin and Eosin (H&E) staining, SMA and Masson's Trichrome staining, we investigated the effect of PTEN haploinsufficiency in combination with Runx2 overexpression on prostate tumorigenesis. Moreover, we employed immunohistochemistry (IHC) to stain Ki67 for cell proliferation, cleaved caspase 3 for apoptosis and AKT phosphorylation for signaling pathway in prostate tissues. Chromatin immunoprecipitation coupled quantitative PCR (ChIP-qPCR), reverse transcription coupled quantitative PCR (RT-qPCR), western blot (WB) analyses and immunofluorescence (IF) were conducted to determine the underlying mechanism by which RUNX2 regulates CXCR7 and AKT phosphorylation in PCa cells. Results: We demonstrated that mice with prostate-specific Pten heterozygous deletion and Runx2 overexpression developed high-grade prostatic intraepithelial neoplasia (HGPIN) and cancerous lesions at age younger than one year, with concomitant high level expression of Akt phosphorylation and the chemokine receptor Cxcr7 in malignant glands. RUNX2 overexpression induced CXCR7 transcription and membrane location and AKT phosphorylation in PTEN-deficient human PCa cell lines. Increased expression of RUNX2 also promoted growth of PCa cells and this effect was largely mediated by CXCR7. CXCR7 expression also positively correlated with AKT phosphorylation in PCa patient specimens. Conclusions: Our results reveal a previously unidentified cooperative role of RUNX2 overexpression and PTEN haploinsufficiency in prostate tumorigenesis, suggesting that the defined RUNX2-CXCR7-AKT axis can be a viable target for effective treatment of PCa.
Collapse
Affiliation(s)
- Yang Bai
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yinhui Yang
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Jian Zhong
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Alexandra M. Blee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Tao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - R. Jeffrey Karnes
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Rafael Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Wanhai Xu
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
36
|
Thomas P. Membrane Androgen Receptors Unrelated to Nuclear Steroid Receptors. Endocrinology 2019; 160:772-781. [PMID: 30753403 DOI: 10.1210/en.2018-00987] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/05/2019] [Indexed: 01/08/2023]
Abstract
Rapid (nongenomic) membrane-initiated androgen actions have been described in nuclear androgen receptor-null cells. Four distinct proteins have been proposed as membrane androgen receptors (mARs) or sensors. Transient receptor potential melastatin 8 (TRPM8) is a calcium channel that acts as a pain receptor and mediates androgen- and menthol-induced increases in calcium levels and survival of prostate cancer cells. Testosterone (T) directly interacts with TRPM8, but extensive androgen receptor binding studies to confirm its role as an mAR are lacking. Oxoeicosanoid receptor 1 (OXER1) is highly expressed in prostate cancer tissues, and its major ligand, 5-oxoeicosatretraenoic acid (5-oxo-ETE), is a potent inducer of prostate cancer cell proliferation and survival. T competes for 5-oxo-ETE binding to OXER1 and antagonizes 5-oxo-ETE-mediated inhibition of cAMP production. However, OXER1 does not meet a traditional criterion for its designation as an mAR because T treatment alone does not alter cAMP signaling. GPRC6A is a class C G protein-coupled receptor activated by l-α-amino acids and is modulated by calcium. Although there has been controversy over the proposed role of T as a GPRC6A ligand, androgen induction of GPRC6A signaling has recently been confirmed by several researchers. ZIP9 belongs to the zinc transporter ZIP (SLC39A) family and displays specific T binding characteristic of an mAR. ZIP9 mediates androgen-dependent intracellular signaling and apoptosis of breast and prostate cancer cells through activation of G proteins. Androgen-signaling functions of ZIP9 have been confirmed in other cells, but the overall importance of ZIP9 in androgen physiology remains unclear. Here, the current status of these four proteins as mARs or sensors is critically reviewed.
Collapse
Affiliation(s)
- Peter Thomas
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| |
Collapse
|
37
|
Enabling precision medicine by unravelling disease pathophysiology: quantifying signal transduction pathway activity across cell and tissue types. Sci Rep 2019; 9:1603. [PMID: 30733525 PMCID: PMC6367506 DOI: 10.1038/s41598-018-38179-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
Signal transduction pathways are important in physiology and pathophysiology. Targeted drugs aim at modifying pathogenic pathway activity, e.g., in cancer. Optimal treatment choice requires assays to measure pathway activity in individual patient tissue or cell samples. We developed a method enabling quantitative measurement of functional pathway activity based on Bayesian computational model inference of pathway activity from measurements of mRNA levels of target genes of the pathway-associated transcription factor. Oestrogen receptor, Wnt, and PI3K-FOXO pathway assays have been described previously. Here, we report model development for androgen receptor, Hedgehog, TGFβ, and NFκB pathway assays, biological validation on multiple cell types, and analysis of data from published clinical studies (multiple sclerosis, amyotrophic lateral sclerosis, contact dermatitis, Ewing sarcoma, lymphoma, medulloblastoma, ependymoma, skin and prostate cancer). Multiple pathway analysis of clinical prostate cancer (PCa) studies showed increased AR activity in hyperplasia and primary PCa but variable AR activity in castrate resistant (CR) PCa, loss of TGFβ activity in PCa, increased Wnt activity in TMPRSS2:ERG fusion protein-positive PCa, active PI3K pathway in advanced PCa, and active PI3K and NFκB as potential hormonal resistance pathways. Potential value for future clinical practice includes disease subtyping and prediction and targeted therapy response prediction and monitoring.
Collapse
|
38
|
Yan Y, Huang H. Interplay Among PI3K/AKT, PTEN/FOXO and AR Signaling in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:319-331. [DOI: 10.1007/978-3-030-32656-2_14] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
39
|
Storbeck KH, Mostaghel EA. Canonical and Noncanonical Androgen Metabolism and Activity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:239-277. [PMID: 31900912 DOI: 10.1007/978-3-030-32656-2_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Androgens are critical drivers of prostate cancer. In this chapter we first discuss the canonical pathways of androgen metabolism and their alterations in prostate cancer progression, including the classical, backdoor and 5α-dione pathways, the role of pre-receptor DHT metabolism, and recent findings on oncogenic splicing of steroidogenic enzymes. Next, we discuss the activity and metabolism of non-canonical 11-oxygenated androgens that can activate wild-type AR and are less susceptible to glucuronidation and inactivation than the canonical androgens, thereby serving as an under-recognized reservoir of active ligands. We then discuss an emerging literature on the potential non-canonical role of androgen metabolizing enzymes in driving prostate cancer. We conclude by discussing the potential implications of these findings for prostate cancer progression, particularly in context of new agents such as abiraterone and enzalutamide, which target the AR-axis for prostate cancer therapy, including mechanisms of response and resistance and implications of these findings for future therapy.
Collapse
Affiliation(s)
- Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Geriatric Research, Education and Clinical Center S-182, VA Puget Sound Health Care System, Seattle, WA, USA.
| |
Collapse
|
40
|
Liu S, Tian Y, Zhu C, Yang X, Sun Q. High miR-718 Suppresses Phosphatase and Tensin Homolog (PTEN) Expression and Correlates to Unfavorable Prognosis in Gastric Cancer. Med Sci Monit 2018; 24:5840-5850. [PMID: 30131483 PMCID: PMC6116637 DOI: 10.12659/msm.909527] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Phosphatase and tensin homolog (PTEN) is a kind of phosphatase which has been demonstrated to suppress progression of gastric cancer. Many micro-RNAs (miRNAs), such as miR-106b, miR-93, and miR-200c, could inhibit expression of PTEN in cell lines; and many miRNAs including miR-21, miR-22, miR-18a, and miR-222 are related to the progression and prognosis of gastric cancer. However, among these miRNAs, the clinical significance of miR-718 has not yet been elucidated. MATERIAL AND METHODS The expression of PTEN and miR-718 in 141 gastric cancer tissues were detected by immunohistochemistry and quantitative real-time PCR respectively. The correlation between PTEN, miR-718, and the clinicopathological factors was analyzed by χ² test. The prognostic significance of PTEN and miR-718 was evaluated by univariate and multivariate analysis. Luciferase reporter assay was performed to evaluate the regulation of PTEN by miR-718. The effect of miR-718 on gastric cancer proliferation and invasion was investigated by MTT assay and Transwell assay. RESULTS Low expression of PTEN and high expression of miR-718 were both significantly associated with unfavorable prognosis, and both were identified as biomarkers predicting poorer prognosis of patients with gastric cancer. Increased miR-718 expression could decrease PTEN expression, thus enhancing phosphatidylinositide 3-kinases/protein kinase B (PI3K/Akt) signaling. Moreover, the abilities of proliferation and invasion of gastric cells transfected with miR-718 were promoted significantly compared with those transfected with control miRNA. CONCLUSIONS Low expression of PTEN and increased expression of miR-718 in gastric cancer tissues were both independent unfavorable prognostic factors of gastric cancer. Upregulation of miR-718 could increase PI3K/Akt signaling by directly downregulating PTEN, thus promoting the proliferation and invasion of gastric cancer cells.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Clinical Laboratory, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Ying Tian
- Department of Clinical Laboratory, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Chanchan Zhu
- Department of Pathology, Medical School of Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaoqing Yang
- Department of Pathology, Qianfoshan Hospital, Jinan, Shandong, China (mainland)
| | - Qing Sun
- Department of Pathology, Qianfoshan Hospital, Jinan, Shandong, China (mainland)
| |
Collapse
|
41
|
Shen M, Kang Y. Complex interplay between tumor microenvironment and cancer therapy. Front Med 2018; 12:426-439. [PMID: 30097962 DOI: 10.1007/s11684-018-0663-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/05/2018] [Indexed: 12/16/2022]
Abstract
Tumor microenvironment (TME) is comprised of cellular and non-cellular components that exist within and around the tumor mass. The TME is highly dynamic and its importance in different stages of cancer progression has been well recognized. A growing body of evidence suggests that TME also plays pivotal roles in cancer treatment responses. TME is significantly remodeled upon cancer therapies, and such change either enhances the responses or induces drug resistance. Given the importance of TME in tumor progression and therapy resistance, strategies that remodel TME to improve therapeutic responses are under developing. In this review, we provide an overview of the essential components in TME and the remodeling of TME in response to anti-cancer treatments. We also summarize the strategies that aim to enhance therapeutic efficacy by modulating TME.
Collapse
Affiliation(s)
- Minhong Shen
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
42
|
Zhou XG, Huang XL, Liang SY, Tang SM, Wu SK, Huang TT, Mo ZN, Wang QY. Identifying miRNA and gene modules of colon cancer associated with pathological stage by weighted gene co-expression network analysis. Onco Targets Ther 2018; 11:2815-2830. [PMID: 29844680 PMCID: PMC5961473 DOI: 10.2147/ott.s163891] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Introduction Colorectal cancer (CRC) is the fourth most common cause of cancer-related mortality worldwide. The tumor, node, metastasis (TNM) stage remains the standard for CRC prognostication. Identification of meaningful microRNA (miRNA) and gene modules or representative biomarkers related to the pathological stage of colon cancer helps to predict prognosis and reveal the mechanisms behind cancer progression. Materials and methods We applied a systems biology approach by combining differential expression analysis and weighted gene co-expression network analysis (WGCNA) to detect the pathological stage-related miRNA and gene modules and construct a miRNA–gene network. The Cancer Genome Atlas (TCGA) colon adenocarcinoma (CAC) RNA-sequencing data and miRNA-sequencing data were subjected to WGCNA analysis, and the GSE29623, GSE35602 and GSE39396 were utilized to validate and characterize the results of WGCNA. Results Two gene modules (Gmagenta and Ggreen) and one miRNA module were associated with the pathological stage. Six hub genes (COL1A2, THBS2, BGN, COL1A1, TAGLN and DACT3) were related to prognosis and validated to be associated with the pathological stage. Five hub miRNAs were identified to be related to prognosis (hsa-miR-125b-5p, hsa-miR-145-5p, hsa-let-7c-5p, hsa-miR-218-5p and hsa-miR-125b-2-3p). A total of 18 hub genes and seven hub miRNAs were predominantly expressed in tumor stroma. Proteoglycans in cancer, focal adhesion, extracellular matrix (ECM)–receptor interaction and so on were common pathways of the three modules. Hsa-let-7c-5p was located at the core of miRNA–gene network. Conclusion These findings help to advance the understanding of tumor stroma in the progression of CAC and provide prognostic biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Xian-Guo Zhou
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao-Liang Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Si-Yuan Liang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Department of Colorectal Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shao-Mei Tang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Si-Kao Wu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Tong-Tong Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zeng-Nan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Department of Urology and Nephrology, The First Affiliated Hospital of Guangxi, Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qiu-Yan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
43
|
Patel R, Fleming J, Mui E, Loveridge C, Repiscak P, Blomme A, Harle V, Salji M, Ahmad I, Teo K, Hamdy FC, Hedley A, van den Broek N, Mackay G, Edwards J, Sansom OJ, Leung HY. Sprouty2 loss-induced IL6 drives castration-resistant prostate cancer through scavenger receptor B1. EMBO Mol Med 2018; 10:e8347. [PMID: 29540470 PMCID: PMC5887544 DOI: 10.15252/emmm.201708347] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 02/09/2018] [Accepted: 02/20/2018] [Indexed: 12/19/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is a lethal form of treatment-resistant prostate cancer and poses significant therapeutic challenges. Deregulated receptor tyrosine kinase (RTK) signalling mediated by loss of tumour suppressor Sprouty2 (SPRY2) is associated with treatment resistance. Using pre-clinical human and murine mCRPC models, we show that SPRY2 deficiency leads to an androgen self-sufficient form of CRPC Mechanistically, HER2-IL6 signalling axis enhances the expression of androgen biosynthetic enzyme HSD3B1 and increases SRB1-mediated cholesterol uptake in SPRY2-deficient tumours. Systemically, IL6 elevated the levels of circulating cholesterol by inducing host adipose lipolysis and hepatic cholesterol biosynthesis. SPRY2-deficient CRPC is dependent on cholesterol bioavailability and SRB1-mediated tumoral cholesterol uptake for androgen biosynthesis. Importantly, treatment with ITX5061, a clinically safe SRB1 antagonist, decreased treatment resistance. Our results indicate that cholesterol transport blockade may be effective against SPRY2-deficient CRPC.
Collapse
Affiliation(s)
| | | | - Ernest Mui
- Institute of Cancer Sciences, Glasgow, UK
| | | | | | | | | | - Mark Salji
- Institute of Cancer Sciences, Glasgow, UK
| | - Imran Ahmad
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, Glasgow, UK
| | - Katy Teo
- Institute of Cancer Sciences, Glasgow, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Headington, Oxford, UK
| | - Ann Hedley
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | | | | | | | - Hing Y Leung
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, Glasgow, UK
| |
Collapse
|
44
|
Profiling Prostate Cancer Therapeutic Resistance. Int J Mol Sci 2018; 19:ijms19030904. [PMID: 29562686 PMCID: PMC5877765 DOI: 10.3390/ijms19030904] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023] Open
Abstract
The major challenge in the treatment of patients with advanced lethal prostate cancer is therapeutic resistance to androgen-deprivation therapy (ADT) and chemotherapy. Overriding this resistance requires understanding of the driving mechanisms of the tumor microenvironment, not just the androgen receptor (AR)-signaling cascade, that facilitate therapeutic resistance in order to identify new drug targets. The tumor microenvironment enables key signaling pathways promoting cancer cell survival and invasion via resistance to anoikis. In particular, the process of epithelial-mesenchymal-transition (EMT), directed by transforming growth factor-β (TGF-β), confers stem cell properties and acquisition of a migratory and invasive phenotype via resistance to anoikis. Our lead agent DZ-50 may have a potentially high efficacy in advanced metastatic castration resistant prostate cancer (mCRPC) by eliciting an anoikis-driven therapeutic response. The plasticity of differentiated prostate tumor gland epithelium allows cells to de-differentiate into mesenchymal cells via EMT and re-differentiate via reversal to mesenchymal epithelial transition (MET) during tumor progression. A characteristic feature of EMT landscape is loss of E-cadherin, causing adherens junction breakdown, which circumvents anoikis, promoting metastasis and chemoresistance. The targetable interactions between androgens/AR and TGF-β signaling are being pursued towards optimized therapeutic regimens for the treatment of mCRPC. In this review, we discuss the recent evidence on targeting the EMT-MET dynamic interconversions to overcome therapeutic resistance in patients with recurrent therapeutically resistant prostate cancer. Exploitation of the phenotypic landscape and metabolic changes that characterize the prostate tumor microenvironment in advanced prostate cancer and consequential impact in conferring treatment resistance are also considered in the context of biomarker discovery.
Collapse
|