1
|
Orange ST, Leslie J, Ross M, Mann DA, Wackerhage H. The exercise IL-6 enigma in cancer. Trends Endocrinol Metab 2023; 34:749-763. [PMID: 37633799 DOI: 10.1016/j.tem.2023.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/28/2023]
Abstract
Interleukin (IL)-6 elicits both anticancer and procancer effects depending on the context, which we have termed the 'exercise IL-6 enigma'. IL-6 is released from skeletal muscles during exercise to regulate short-term energy availability. Exercise-induced IL-6 provokes biological effects that may protect against cancer by improving insulin sensitivity, stimulating the production of anti-inflammatory cytokines, mobilising immune cells, and reducing DNA damage in early malignant cells. By contrast, IL-6 continuously produced by leukocytes in inflammatory sites drives tumorigenesis by promoting chronic inflammation and activating tumour-promoting signalling pathways. How can a molecule have such opposing effects on cancer? Here, we review the roles of IL-6 in chronic inflammation, tumorigenesis, and exercise-associated cancer prevention and define the factors that underpin the exercise IL-6 enigma.
Collapse
Affiliation(s)
- Samuel T Orange
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Jack Leslie
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mark Ross
- Institute of Life and Earth Sciences, School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, UK
| | - Derek A Mann
- Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK; Newcastle Fibrosis Research Group, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Henning Wackerhage
- Department of Sport & Health Science, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Anguiano B, Álvarez L, Delgado-González E, Ortiz-Martínez Z, Montes de Oca C, Morales G, Aceves C. Protective effects of iodine on rat prostate inflammation induced by sex hormones and on the DU145 prostate cancer cell line treated with TNF. Mol Cell Endocrinol 2023; 572:111957. [PMID: 37192707 DOI: 10.1016/j.mce.2023.111957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
Molecular iodine (I2) prevents oxidative stress and prostate hyperplasia induced by hyperandrogenism and reduces cell viability in prostate cancer cell lines. Here, we aimed to evaluate the protective effect of I2 and testosterone (T) on hyperestrogenism-induced prostate inflammation. Additionally, the effects of I2 and/or tumor necrosis factor (TNF) on cell viability and interleukin 6 (IL6) secretion were evaluated in a prostate cancer cell line (DU145). We also investigated whether the effects of I2 on viability are peroxisome proliferator-activated receptor gamma (PPARG)-dependent. Castrated (Cx) rats received pellets of either 17β estradiol (E2) or E2 and T and were treated with I2 (0.05%) in the drinking water for four weeks. The experimental groups were sham, Cx, Cx + E2, Cx + E2+I2, Cx + E2+T, and Cx + E2+T + I2. As expected, inflammation was triggered in the Cx + E2 group (high inflammation score; increase in TNF and transcriptional activity of RELA [nuclear factor-kappa B p65 subunit]), and this effect was diminished in the Cx + E2+T group (medium inflammation score and decrease in TNF). The lowest inflammation score (decrease of TNF and RELA and increase of PPARG) was obtained in the Cx + E2+T + I2 group. In DU145 cells, I2 (400 μM) and TNF (10 ng/ml) additively reduced cell viability, and I2 reduced the production of TNF-stimulated IL6. The PPARG antagonist (GW9662) did not inhibit the effects of I2 on the loss of cell viability. In summary, our data suggest that I2 and T exert a synergistic anti-inflammatory action on the normal prostate, and the interrelationship between I2 and TNF leads to anti-proliferative effects in DU145 cells. PPARG does not seem to participate in the I2-induced cell viability loss in the prostate.
Collapse
Affiliation(s)
- Brenda Anguiano
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico.
| | - Lourdes Álvarez
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico
| | - Evangelina Delgado-González
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico
| | - Zamira Ortiz-Martínez
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico
| | - Carlos Montes de Oca
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico
| | - Giapsy Morales
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico
| | - Carmen Aceves
- Departamento de Neurobiología Celular y Molecular. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, 76230, Querétaro, Mexico
| |
Collapse
|
3
|
Gano CA, Fatima S, Failes TW, Arndt GM, Sajinovic M, Mahns D, Saedisomeolia A, Coorssen JR, Bucci J, de Souza P, Vafaee F, Scott KF. Anti-cancer potential of synergistic phytochemical combinations is influenced by the genetic profile of prostate cancer cell lines. Front Nutr 2023; 10:1119274. [PMID: 36960209 PMCID: PMC10029761 DOI: 10.3389/fnut.2023.1119274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/09/2023] [Indexed: 03/10/2023] Open
Abstract
Introduction Despite strong epidemiological evidence that dietary factors modulate cancer risk, cancer control through dietary intervention has been a largely intractable goal for over sixty years. The effect of tumour genotype on synergy is largely unexplored. Methods The effect of seven dietary phytochemicals, quercetin (0-100 μM), curcumin (0-80 μM), genistein, indole-3-carbinol (I3C), equol, resveratrol and epigallocatechin gallate (EGCG) (each 0-200 μM), alone and in all paired combinations om cell viability of the androgen-responsive, pTEN-null (LNCaP), androgen-independent, pTEN-null (PC-3) or androgen-independent, pTEN-positive (DU145) prostate cancer (PCa) cell lines was determined using a high throughput alamarBlue® assay. Synergy, additivity and antagonism were modelled using Bliss additivism and highest single agent equations. Patterns of maximum synergy were identified by polygonogram analysis. Network pharmacology approaches were used to identify interactions with known PCa protein targets. Results Synergy was observed with all combinations. In LNCaP and PC-3 cells, I3C mediated maximum synergy with five phytochemicals, while genistein was maximally synergistic with EGCG. In contrast, DU145 cells showed resveratrol-mediated maximum synergy with equol, EGCG and genistein, with I3C mediating maximum synergy with only quercetin and curcumin. Knockdown of pTEN expression in DU145 cells abrogated the synergistic effect of resveratrol without affecting the synergy profile of I3C and quercetin. Discussion Our study identifies patterns of synergy that are dependent on tumour cell genotype and are independent of androgen signaling but are dependent on pTEN. Despite evident cell-type specificity in both maximally-synergistic combinations and the pathways that phytochemicals modulate, these combinations interact with similar prostate cancer protein targets. Here, we identify an approach that, when coupled with advanced data analysis methods, may suggest optimal dietary phytochemical combinations for individual consumption based on tumour molecular profile.Graphical abstract.
Collapse
Affiliation(s)
- Carol A. Gano
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Shadma Fatima
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
- School of Biotechnology and Biological Sciences, UNSW Sydney, Sydney, NSW, Australia
- Shadma Fatima, ;
| | - Timothy W. Failes
- ACRF Drug Discovery Centre, Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Gregory M. Arndt
- ACRF Drug Discovery Centre, Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Mila Sajinovic
- Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
| | - David Mahns
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Ahmad Saedisomeolia
- School of Human Nutrition, McGill University, Sainte Anne-de-Bellevue, QC, Canada
| | - Jens R. Coorssen
- Departments of Health Sciences and Biological Sciences, Faculties of Applied Health Science, and Mathematics and Science, Brock University, St. Catharines, ON, Canada
| | - Joseph Bucci
- St George Hospital Clinical School, UNSW, Kogarah, NSW, Australia
| | - Paul de Souza
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biological Sciences, UNSW Sydney, Sydney, NSW, Australia
- UNSW Data Science Hub (uDASH), UNSW Sydney, Sydney, NSW, Australia
| | - Kieran F. Scott
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
- *Correspondence: Kieran F. Scott,
| |
Collapse
|
4
|
Benlier N, Solakhan M, Sever ÖN, Yıldırım Z, Orhan N, Çiçek H, Yıldırım M. Role of serum cathelicidin in diagnosis of patient with prostatitis and prostate carcinoma. AFRICAN JOURNAL OF UROLOGY 2022. [DOI: 10.1186/s12301-022-00330-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Abstract
Background
This study investigated the diagnostic role of 75 levels measured in serum prostatitis and prostate carcinoma and in the differentiation of these two conditions.
Methods
The study was conducted with 75 patients histopathologically diagnosed with prostate carcinoma or prostatitis and followed up at the Departments of Urology and Medical Oncology and 21 healthy male subjects. Serum cathelicidin levels were investigated using the ELISA method. Statistical analyses were performed using the SPSS for Windows 22.0 package software. Compliance of the variables to normal distribution was examined using visual and analytic methods. In the Kolmogorov–Smirnov test, cases with a p value of greater than 0.05 were accepted as normal distribution.
Results
A total of 75 patients including 45 diagnosed with prostate carcinoma and 30 diagnosed with prostatitis, as well as 21 healthy control subjects were included in the study. Prostate-specific antigen (PSA) was detected as 23 (4–1200) ng/mL in the patients with prostate carcinoma and as 9.85 (3.9–405 ng/mL) in the patients with prostatitis. The cathelicidin levels were diagnostically significant when assessed by ROC analysis in the prostate cancer, prostatitis and control groups (p = 0.005). The cutoff values derived from the ROC curve analysis were 3.5151 ng/mL for distinguishing prostate cancer from prostatitis, 2.2620 ng/mL for prostate cancer versus control group and 1.2340 ng/mL for prostatitis versus control group.
Conclusions
In this study we showed that the serum cathelicidin levels were significantly higher in the patients diagnosed with prostate carcinoma. Measurement of serum cathelicidin levels could be used as a diagnostic marker in prostate carcinoma as well as facilitating differential diagnosis to strengthen the diagnostic suspicion before prostate biopsy and distinguish the diagnosis from prostatitis cases.
Collapse
|
5
|
Seo E, Jang H, Kwon S, Kwon Y, Kim S, Lee S, Jeong AJ, Shin HM, Kim Y, Ma S, Kim H, Lee Y, Suh P, Ye S. Loss of phospholipase Cγ1 suppresses hepatocellular carcinogenesis through blockade of STAT3-mediated cancer development. Hepatol Commun 2022; 6:3234-3246. [PMID: 36153805 PMCID: PMC9592768 DOI: 10.1002/hep4.2077] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/11/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022] Open
Abstract
Phospholipase C gamma 1 (PLCγ1) plays an oncogenic role in several cancers, alongside its usual physiological roles. Despite studies aimed at identifying the effect of PLCγ1 on tumors, the pathogenic role of PLCγ1 in the tumorigenesis and development of hepatocellular carcinoma (HCC) remains unknown. To investigate the function of PLCγ1 in HCC, we generated hepatocyte-specific PLCγ1 conditional knockout (PLCγ1f/f ; Alb-Cre) mice and induced HCC with diethylnitrosamine (DEN). Here, we identified that hepatocyte-specific PLCγ1 deletion effectively prevented DEN-induced HCC in mice. PLCγ1f/f ; Alb-Cre mice showed reduced tumor burden and tumor progression, as well as a decreased incidence of HCC and less marked proliferative and inflammatory responses. We also showed that oncogenic phenotypes such as repressed apoptosis, and promoted proliferation, cell cycle progression and migration, were induced by PLCγ1. In terms of molecular mechanism, PLCγ1 regulated the activation of signal transducer and activator of transcription 3 (STAT3) signaling. Moreover, PLCγ1 expression is elevated in human HCC and correlates with a poor prognosis in patients with HCC. Our results suggest that PLCγ1 promotes the pathogenic progression of HCC, and PLCγ1/STAT3 axis was identified as a potential therapeutic target pathway for HCC.
Collapse
Affiliation(s)
- Eun‐Bi Seo
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
- Biomedical Science Project (BK21PLUS)Seoul National University College of MedicineSeoulRepublic of Korea
| | - Hyun‐Jun Jang
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanRepublic of Korea
| | - Sun‐Ho Kwon
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
| | - Yong‐Jin Kwon
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
- Biomedical Science Project (BK21PLUS)Seoul National University College of MedicineSeoulRepublic of Korea
| | - Seul‐Ki Kim
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
| | - Song‐Hee Lee
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
| | - Ae Jin Jeong
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
| | - Hyun Mu Shin
- Wide River Institute of ImmunologySeoul National UniversityHongcheonRepublic of Korea
| | - Yong‐Nyun Kim
- Division of Translational ScienceNational Cancer CenterGoyangRepublic of Korea
| | - Stephanie Ma
- State Key Laboratory of Liver ResearchLi Ka Shing Faculty of Medicine, The University of Hong KongHong Kong
| | - Haeryoung Kim
- Department of PathologySeoul National University College of MedicineSeoulRepublic of Korea
| | - Yun‐Han Lee
- Department of Molecular MedicineKeimyung University School of MedicineDaeguRepublic of Korea
| | - Pann‐Ghill Suh
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanRepublic of Korea
- Korea Brain Research Institute (KBRI)DaeguRepublic of Korea
| | - Sang‐Kyu Ye
- Department of Pharmacology and Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
- Biomedical Science Project (BK21PLUS)Seoul National University College of MedicineSeoulRepublic of Korea
- Wide River Institute of ImmunologySeoul National UniversityHongcheonRepublic of Korea
- Ischemic/Hypoxic Disease InstituteSeoul National University College of MedicineSeoulRepublic of Korea
- Neuro‐Immune Information Storage Network Research CenterSeoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
6
|
Yang HY, Su RY, Chung CH, Huang KY, Lin HA, Wang JY, Chen CC, Chien WC, Lin HC. Association between trichomoniasis and prostate and bladder diseases: a population-based case–control study. Sci Rep 2022; 12:15358. [PMID: 36100630 PMCID: PMC9468537 DOI: 10.1038/s41598-022-19561-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
Trichomonas vaginalis infection is one of the most widespread sexually transmitted infections in the world. There are approximately 276 million cases worldwide. Most men remain undiagnosed and untreated because they are asymptomatic. The chronic inflammation induced by persistent infection may increase the risk of developing genitourinary cancers. In this study, we aimed to investigate the association between trichomoniasis and benign prostate hyperplasia (BPH), prostate cancer (PCa), and bladder cancer (BC) in Taiwan. We designed a case–control study by using the database of the National Health Insurance program in Taiwan. We used the International Classification of Diseases, 9th Revision classifications to classify all the medical conditions in the case and control groups. All odds ratios (ORs) and 95% confidence intervals (CIs) were analyzed using multivariable logistic regression to adjust for all comorbidities and variables. From 2000 to 2015, we enrolled a total of 62,544 individuals as the case group and 187,632 as the control group. Trichomoniasis exposure had a significant association with BPH and PCa (adjusted OR: BPH = 2.685, 95% CI = 1.233–4.286, P = 0.013; PCa = 5.801, 95% CI = 1.296–26.035, P = 0.016). The relative risk was much higher if patients had both trichomoniasis and depression (adjusted OR = 7.682, 95% CI = 5.730–9.451, P < 0.001). Men with trichomoniasis had a significantly higher risk of developing BPH and PCa than those without. Healthcare professionals should not only pay more attention to disease treatment, but also to public health education.
Collapse
|
7
|
Association of Cytokine Gene Polymorphisms with Prostate Cancer Risk from a Study in Central China. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:1259009. [PMID: 36034203 PMCID: PMC9392598 DOI: 10.1155/2022/1259009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022]
Abstract
Our study intended to investigate five cytokine gene single nucleotide polymorphisms (SNPs) and their associations with prostate cancer risk. Genotypes of five cytokine gene SNPs were detected by MassARRAY for blood samples from a group of patients with prostate cancer (n = 90) and a control group (n = 140) in central China. The differences in tumor clinical stages, Gleason scores, and PSA values in patients with prostate cancer were also investigated. The frequencies of the five cytokine gene SNPs (L-1β rs16944, IL-4 rs2070874, IL-4rs2227284, IL-16 rs7175701, and IL-16 rs11556218) genotypes were not found to be significantly mutated in prostate cancer patients compared with the control group. In addition, for five cytokine gene SNPs genotypic comparisons, patients with different Gleason scores, clinical stages, and PSA values were grouped into two subgroups. There was also no statistically significant association in all these subgroups. Our study suggests that cytokine gene polymorphisms may not be a risk factor for prostate cancer in a central Chinese population. Nevertheless, more large-scale studies on the Chinese population are necessary to examine our conclusions. The discovery of cytokine gene polymorphisms related to prostate cancer could update our understanding of the etiology and improve our knowledge of the early detection, diagnosis, and treatment of prostate cancer.
Collapse
|
8
|
Moran NE, Thomas-Ahner JM, Wan L, Zuniga KE, Erdman JW, Clinton SK. Tomatoes, Lycopene, and Prostate Cancer: What Have We Learned from Experimental Models? J Nutr 2022; 152:1381-1403. [PMID: 35278075 PMCID: PMC9178968 DOI: 10.1093/jn/nxac066] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/04/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Human epidemiology suggests a protective effect of tomatoes or tomato phytochemicals, such as lycopene, on prostate cancer risk. However, human epidemiology alone cannot reveal causal relations. Laboratory animal models of prostate cancer provide opportunities to investigate hypotheses regarding dietary components in precisely controlled, experimental systems, contributing to our understanding of diet and cancer risk relations. We review the published studies evaluating the impact of tomatoes and/or lycopene in preclinical models of prostate carcinogenesis and tumorigenesis. The feeding of tomatoes or tomato components demonstrates anti-prostate cancer activity in both transplantable xenograft models of tumorigenesis and models of chemically- and genetically-driven carcinogenesis. Feeding pure lycopene shows anticancer activity in most studies, although outcomes vary by model system, suggesting that the impact of pure lycopene can depend on dose, duration, and specific carcinogenic processes represented in different models. Nonetheless, studies with the transgenic adenocarcinoma of the mouse prostate (TRAMP) model of carcinogenesis typically demonstrate similar bioactivity to that of tomato feeding. In general, interventions that commence earlier in carcinogenesis and are sustained tend to be more efficacious. Accumulated data suggest that lycopene is one, but perhaps not the only, anticancer bioactive compound in tomatoes. Although it is clear that tomatoes and lycopene have anti-prostate cancer activity in rodent models, major knowledge gaps remain in understanding dose-response relations and molecular mechanisms of action. Published and future findings from rodent studies can provide guidance for translational scientists to design and execute informative human clinical trials of prostate cancer prevention or in support of therapy.
Collapse
Affiliation(s)
- Nancy E Moran
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jennifer M Thomas-Ahner
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Lei Wan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Interdisciplinary Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Krystle E Zuniga
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA.,Livestrong Cancer Institutes, Dell Medical School, University of Texas, Austin, TX, USA
| | - John W Erdman
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| | - Steven K Clinton
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH, USA
| |
Collapse
|
9
|
Terada N, Aizawa R, Nihei K, Shiota M, Kojima T, Kimura T, Inoue T, Kitamura H, Sugimoto M, Nishiyama H, Mizowaki T, Kamoto T. Narrative review of local prostate and metastasis-directed radiotherapy in the treatment of metastatic prostate cancer. Jpn J Clin Oncol 2022; 52:633-641. [PMID: 35325157 DOI: 10.1093/jjco/hyac035] [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: 08/26/2021] [Accepted: 02/22/2022] [Indexed: 11/14/2022] Open
Abstract
The role of local treatment in patients with de novo metastatic prostate cancer is controversial. In population-based retrospective studies, metastatic prostate cancer patients who received local treatment with prostate radiotherapy showed a better prognosis than those who did not. In addition, several prospective randomized studies demonstrated that prostate radiotherapy achieves a survival benefit for patients with oligo-metastasis. Moreover, the efficacy of metastasis-directed radiotherapy was evaluated, revealing a potential benefit for patients with oligo-metastasis. Importantly, these radiotherapies may reduce the occurrence of symptomatic local events. In this review, the rationale, efficacy and future perspectives for local prostate and metastasis-directed radiotherapy in the treatment of metastatic prostate cancer were described and summarized.
Collapse
Affiliation(s)
- Naoki Terada
- Department of Urology, Miyazaki University, Miyazaki, Japan
| | - Rihito Aizawa
- Department of Radiation Oncology & Image-Applied Therapy, Kyoto University, Kyoto, Japan
| | - Keiji Nihei
- Department of Radiation Oncology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Masaki Shiota
- Department of Urology, Kyushu University, Fukuoka, Japan
| | | | | | | | | | - Mikio Sugimoto
- Department of Urology, Kagawa University, Takamatsu, Japan
| | | | - Takashi Mizowaki
- Department of Radiation Oncology & Image-Applied Therapy, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
10
|
The Metastasis Suppressor NDRG1 Directly Regulates Androgen Receptor Signaling in Prostate Cancer. J Biol Chem 2021; 297:101414. [PMID: 34785213 PMCID: PMC8668986 DOI: 10.1016/j.jbc.2021.101414] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
N-myc-downregulated gene 1 (NDRG1) has potent anticancer effects and inhibits cell growth, survival, metastasis, and angiogenesis. Previous studies suggested that NDRG1 is linked to the androgen signaling network, but this mechanistic relationship is unclear. Considering the crucial role of the androgen receptor (AR) in prostate cancer (PCa) progression, here we examined for the first time the effect of NDRG1 on AR expression, activation, and downstream signaling in LNCaP, 22Rv1, and C4-2B PCa cell types. We demonstrate that NDRG1 effectively promotes interaction of AR with the chaperone HSP90, which in turn stabilizes the AR while decreasing its androgen-mediated activation. The expression of NDRG1 suppressed: (1) AR activation, as measured by p-ARSer213 and p-ARSer81; (2) expression of a major AR transcriptional target, prostate-specific antigen (PSA); and (3) AR transcriptional activity, probably via inhibiting the c-Jun-AR interaction by reducing c-Jun phosphorylation (p-c-JunSer63). NDRG1 was also demonstrated to inhibit multiple key molecules involved in androgen-dependent and -independent signaling (namely EGFR, HER2, HER3, PI3K, STAT3, and NF-κB), which promote the development of castration-resistant prostate cancer. We also identified the cysteine-rich secretory protein/antigen 5/pathogenesis related-1 (CAP) domain of NDRG1 as vital for inhibition of AR activity. Examining NDRG1 and p-NDRG1 in PCa patient specimens revealed a significant negative correlation between NDRG1 and PSA levels in prostatectomy patients that went on to develop metastasis. These results highlight a vital role for NDRG1 in androgen signaling and its potential as a key therapeutic target and biomarker in PCa.
Collapse
|
11
|
Wang S, Li Y, Li W, Zhang K, Yuan Z, Cai Y, Xu K, Zhou J, Du Z. Curcuma oil ameliorates benign prostatic hyperplasia through suppression of the nuclear factor-kappa B signaling pathway in rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:113703. [PMID: 33340599 PMCID: PMC9586842 DOI: 10.1016/j.jep.2020.113703] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 05/05/2023]
Abstract
ETHNO PHARMACOLOGICAL RELEVANCE Curcuma longa L is traditionally used as an anti-inflammatory remedy in Chinese traditional medicine. Curcuma oil (CO), a lipophilic fraction from Curcuma longa L. has been reported to have anti-proliferative, anti-inflammatory and anti-oxidant activities. However, CO has never been investigated for its possible therapeutic effects on benign prostatic hyperplasia (BPH). AIMS OF THE STUDY The study is thus to determine the therapeutic effects of curcuma oil on BPH and also the possible mechanism (s) of action. MATERIALS &METHODS A BPH-1 cell line and Sprague Dawley (SD) rats were used to establish BPH models in vitro and in vivo, respectively. Rats were treated by CO (2.4, 7.2 mg/kg/i.g.) and finasteride (5 mg/kg/i.g.), respectively. Histological changes were examined by hematoxylin and eosin (H&E) staining. Protein expression was analyzed for 5α-reductase (5AR), dihydrotestosterone (DHT), interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α by ELISA. Ki-67, Caspase-8,-9 and -3 expressions were evaluated via immunohistochemistry (IHC). RESULTS CO effectively induced apoptosis in BPH-1 cells. BPH was successfully established by administration of testosterone propionate (TP) in rats, which upregulated both 5α-reductase expression and DHT production. Importantly, TP establishment significantly stimulated the phosphorylation of p65, one subunit of NF-κB, thus led to activation of the NF-κB signaling pathway in prostatic tissues of rats. In turn, the activation of NF-κB pathway induced concomitant upregulation of proinflammatory factors IL-1β, IL-6, TNF-α, and COX-2 and significant increase of the Bcl2/Bax expression ratio for enhanced cell survival, contributing to the initiation and progression of BPH in rats. Notably, CO therapy significantly decreased prostate weight and hyperplasia in BPH-induced animals. Also CO was found to suppress the expression of 5α-reductase and thus the production of DHT, which is essential for the amelioration of BPH. More importantly, CO was shown to suppress the activation of NF-κB pathway through decreasing the expression of phosphorylated p65 and consequently reduced the inflammatory responses and cell survival in prostatic tissues, leading to the inhibition of BPH development in rats. CONCLUSION Curcuma oil is very effective for ameliorating BPH in rats. The underlying mechanisms involve in reduced inflammatory responses and cell survival through suppression of the NF-κB signaling pathway by CO in prostatic tissues.
Collapse
Affiliation(s)
- Shanshan Wang
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Yun Li
- R&D Centre, Infinitus (China) Company Ltd, Guangzhou, China
| | - Wenzhi Li
- R&D Centre, Infinitus (China) Company Ltd, Guangzhou, China
| | - Kun Zhang
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Zhengqiang Yuan
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Yina Cai
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Kuncheng Xu
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Jinrong Zhou
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| | - Zhiyun Du
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China; Conney Allan Biotechnology Company Ltd, Guangzhou, 510095, China.
| |
Collapse
|
12
|
Natani S, Dhople VM, Parveen A, Sruthi KK, Khilar P, Bhukya S, Ummanni R. AMPK/SIRT1 signaling through p38MAPK mediates Interleukin-6 induced neuroendocrine differentiation of LNCaP prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119085. [PMID: 34171447 DOI: 10.1016/j.bbamcr.2021.119085] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/29/2022]
Abstract
Neuroendocrine Prostate Cancer (NEPC) is an aggressive form of androgen independent prostate cancer (AIPC), correlated with therapeutic resistance. Interleukin (IL)-6 promotes proliferation and neuroendocrine differentiation (NED) of androgen dependent LNCaP cells. We treated LNCaP cells with IL-6 and observed for in vitro NED of cells and also expression of NE markers βIII tubulin, neuron-specific enolase (NSE) and chromogranin A (ChA). Here we investigated the proteins and/or pathways involved in NED of LNCaP cells induced by IL-6 and characterized their role in NED of PCa cells. We found that the altered proteins modulated AMPK signaling pathway in NE cells. Remarkably, IL-6 induces NED of LNCaP cells through activation of AMPK and SIRT1 and also both of these are co-regulated while playing a predominant role in NED of LNCaP cells. Of the few requirements of AMPK-SIRT1 activation, increased eNOS is essential for NED by elevating Nitric oxide (NO) levels. Pleiotropic effects of NO ultimately regulate p38MAPK in IL-6 induced NED. Hence, IL-6 induced AMPK-SIRT1 activation eventually transfers its activation signals through p38MAPK for advancing NED of LNCaP cells. Moreover, inactivation of p38MAPK with specific inhibitor (SB203580) attenuated IL-6 induced NED of LNCaP cells. Therefore, IL-6 promotes NED of PCa cells via AMPK/SIRT1/p38MAPK signaling. Finally, targeting AMPK-SIRT1 or p38MAPK in androgen independent PC3 cells with neuroendocrine features reversed their neuroendocrine characteristics. Taken together these novel findings reveal that targeting p38MAPK mitigated NED of PCa cells, and thus it can be a favorable target to overcome progression of NEPC.
Collapse
Affiliation(s)
- Sirisha Natani
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vishnu M Dhople
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Asha Parveen
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - K K Sruthi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priyanka Khilar
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Supriya Bhukya
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Ramesh Ummanni
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
13
|
Inhibitory Activities of Dimeric Ellagitannins Isolated from Cornus alba on Benign Prostatic Hypertrophy. Molecules 2021; 26:molecules26113446. [PMID: 34204056 PMCID: PMC8201330 DOI: 10.3390/molecules26113446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/02/2022] Open
Abstract
Benign prostatic hypertrophy (BPH) is an intractable chronic inflammatory disease. We studied the efficacy of two ellagitannins, namely camptothin B (1) and cornusiin A (2) that were isolated from Cornus alba (CA) for the treatment of BPH, which is a common health issue in older men. The ellagitannins (1 and 2) were evaluated on its inhibitory activities of the enzyme 5α-reductase and tumor necrosis factor (TNF)-α, its interleukin (IL)-1β, IL-6, and IL-8 production, and its anti-proliferation and apoptosis induction in prostate cells that show hypertrophy (RWPE-1 cell). In inhibition of 5α-reductase, the ellagitannins (1 and 2) showed potential effects, compared to the positive control, finasteride. In the case of IL-1β, IL-6, IL-8, and TNF-α, 1 and 2 showed good inhibitory effects as compared to the control group treated with LPS. The ellagitannins (1 and 2) were also shown to inhibit proliferation of, and induce apoptosis in, the RWPE-1 cell. These results suggest that the ellagitannins (1 and 2) may be good candidates for the treatment of BPH.
Collapse
|
14
|
Skeletal Muscle-Adipose Tissue-Tumor Axis: Molecular Mechanisms Linking Exercise Training in Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094469. [PMID: 33922898 PMCID: PMC8123194 DOI: 10.3390/ijms22094469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Increased visceral adiposity may influence the development of prostate cancer (PCa) aggressive tumors and cancer mortality. White adipose tissue (WAT), usually referred to as periprostatic adipose tissue (PPAT), surrounds the prostatic gland and has emerged as a potential mediator of the tumor microenvironment. Exercise training (ET) induces several adaptations in both skeletal muscle and WAT. Some of these effects are mediated by ET-induced synthesis and secretion of several proteins, known as myo- and adipokines. Together, myokines and adipokines may act in an endocrine-like manner to favor communication between skeletal muscle and WAT, as they may work together to improve whole-body metabolic health. This crosstalk may constitute a potential mechanism by which ET exerts its beneficial role in the prevention and treatment of PCa-related disorders; however, this has not yet been explored. Therefore, we reviewed the current evidence on the effects of skeletal muscle–WAT–tumor crosstalk in PCa, and the potential mediators of this process to provide a better understanding of underlying ET-related mechanisms in cancer.
Collapse
|
15
|
Pihlstrøm N, Jin Y, Nenseth Z, Kuzu OF, Saatcioglu F. STAMP2 Expression Mediated by Cytokines Attenuates Their Growth-Limiting Effects in Prostate Cancer Cells. Cancers (Basel) 2021; 13:cancers13071579. [PMID: 33808059 PMCID: PMC8036285 DOI: 10.3390/cancers13071579] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Prostate cancer (PCa) is the most common non-skin cancer and one of the leading causes of cancer death in men. Despite significant developments in therapy options with improved survival, no curative treatment is currently available. We have previously identified six transmembrane protein of prostate 2 (STAMP2) as an important factor for PCa growth and survival. We now show that STAMP2 expression is regulated by inflammatory signaling, which has recently been implicated in PCa. Two proinflammatory cytokines, interleukin 6 and interleukin 1 beta, synergize with each other to induce STAMP2 expression. Interestingly, STAMP2 knockdown increased the sensitivity of PCa cells to cytokine treatment. Thus, STAMP2 that acts as a survival factor in PCa, is both independently and synergistically regulated by inflammatory signaling that may affect disease progression. Abstract Inflammatory events and dysregulated cytokine expression are implicated in prostate cancer (PCa), but the underlying molecular mechanisms are poorly understood at present. We have previously identified six transmembrane protein of the prostate 2 (STAMP2, also known as STEAP4) as an androgen-regulated gene, as well as a key regulator of PCa growth and survival. STAMP2 is also regulated by, and participates in, inflammatory signaling in other tissues and pathologies. Here, we show that the proinflammatory cytokines interleukin 6 (IL-6) and Interleukin 1 beta (IL-1β) significantly increase and strongly synergize in promoting STAMP2 expression in PCa cells. The two cytokines increase androgen-induced STAMP2 expression, but not expression of other known androgen target genes, suggesting a unique interplay of androgens and cytokines in regulating STAMP2 expression. Interestingly, STAMP2 knockdown significantly increased the ability of IL-6 and IL-1β to inhibit PCa cell growth in vitro. These results suggest that STAMP2 may represent a unique node through which inflammatory events mediate their effects on PCa growth and survival.
Collapse
Affiliation(s)
- Nicklas Pihlstrøm
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Yang Jin
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Zeynep Nenseth
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
| | - Omer F. Kuzu
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
- Correspondence: (O.F.K.); (F.S.); Tel.: +47-22-854-569 (F.S.); Fax: +47-22-857-207 (F.S.)
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, 0315 Oslo, Norway; (N.P.); (Y.J.); (Z.N.)
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0188 Oslo, Norway
- Correspondence: (O.F.K.); (F.S.); Tel.: +47-22-854-569 (F.S.); Fax: +47-22-857-207 (F.S.)
| |
Collapse
|
16
|
Liu Y, Long W, Zhang Z, Mai L, Huang S, Liu B, Cao W, Wu J, Zhou F, Li Y, He L. Cytoreductive radiotherapy combined with abiraterone in metastatic castration-resistance prostate cancer: a single center experience. Radiat Oncol 2021; 16:5. [PMID: 33407637 PMCID: PMC7789459 DOI: 10.1186/s13014-020-01732-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background To investigate the potential benefit of cytoreductive radiotherapy (cRT) in metastatic castration-resistant prostate cancer (mCRPC) patients receiving abiraterone. Methods From February 2014 to February 2019, 149 mCRPC patients treated with abiraterone were identified. Patients receiving cRT before abiraterone failure (AbiRT group) were matched by one-to-two propensity score to patients without cRT before abiraterone failure (non-AbiRT group). Results The median follow-up was 23.5 months. Thirty patients (20.1%) were in the AbiRT group, whereas 119 patients (79.9%) were in the non-AbiRT group. The 2-year OS of patients managed by AbiRT and non-AbiRT were 89.5% and 73.5%, respectively (P = 0.0003). On multivariate analysis, only AbiRT (HR 0.17; 95% CI 0.05–0.58; P = 0.004) and prognostic index (HR 2.71; 95% CI 1.37–5.35; P = 0.004) were significant factors. After matching, AbiRT continued to be associated with improved OS (median OS not reached vs. 44.0 months, P = 0.009). Subgroup analysis revealed that patients aged ≤ 65 years (HR 0.09; 95% CI 0.01–0.65; P = 0.018), PSA ≤ 20 ng/mL (HR 0.29; 95% CI 0.09–0.99; P = 0.048), chemotherapy-naïve upon abiraterone treatment (HR 0.20; 95% CI 0.06–0.66; P = 0.008) and in intermediate prognosis groups by COU-AA-301 prognostic index (HR 0.13; 95% CI 0.03–0.57; P = 0.007) had improved OS with AbiRT. Conclusions cRT before resistance to abiraterone may improve survival in selected mCRPC patients: age ≤ 65 years old, chemotherapy-naïve, with a relatively low PSA level at the diagnosis of mCRPC and intermediate prognosis.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Wen Long
- Department of Nuclear Medicine, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guanghzou, People's Republic of China
| | - Zitong Zhang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Lixin Mai
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Sijuan Huang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Boji Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Wufei Cao
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Jianhua Wu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Yonghong Li
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| | - Liru He
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
17
|
Interleukin-6 Function and Targeting in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:1-8. [PMID: 33559852 DOI: 10.1007/978-3-030-55617-4_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin-6 (IL-6) is a proinflammatory cytokine, which is involved in pathogenesis of several cancers. Its expression and function in prostate cancer have been extensively studied in cellular models and clinical specimens. High levels of IL-6 were detected in conditioned media from cells which do not respond to androgens. Increased phosphorylation of signal transducer and activator of transcription (STAT)3 factor which is induced in response to IL-6 is one of the typical features of prostate cancer. However, reports in the literature show regulation of neuroendocrine phenotype by IL-6. Effects of IL-6 on stimulation of proliferation, migration, and invasion lead to the establishment of experimental and clinical approaches to target IL-6. In prostate cancer, anti-IL-6 antibodies were demonstrated to inhibit growth in vitro and in vivo. Clinically, application of anti-IL-6 therapies did not improve survival of patients with metastatic prostate cancer. However, clinical trial design in the future may include different treatment schedule and combinations with experimental and clinical therapies. Endogenous inhibitors of IL-6 such as suppressors of cytokine signaling and protein inhibitors of activated STAT have variable effects on prostate cells, depending on presence or absence of the androgen receptor.
Collapse
|
18
|
Chen M, Yuan C, Xu T. An increase in prostate cancer diagnosis during inflammatory bowel disease: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol 2020; 44:302-309. [PMID: 31447293 DOI: 10.1016/j.clinre.2019.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND In recent years, some studies showed that inflammatory bowel disease (IBD) might be associated with an increased risk of prostate cancer (PC), whereas some other studies indicated that IBD was not associated with the increased risk of PC. In the present study, we aimed to conduct a systematic review and meta-analysis to evaluate the association of IBD and PC risk. METHODS Web of Science and PubMed were systematically searched on for link of PC risk and IBD published from January 1976 to February 2019. The pooled multivariate odd ratio (OR) or relative risk (RR) and 95% confidence intervals (CI) were obtained by the use of STATA 12.0 software. RESULTS The meta-analysis indicated that IBD showed a 78% increase in PC risk (95% CI: 1.32-2.41). Sensitivity analyses showed no changes in the direction of effect after excluding any one study (supplementary figure 1). A significant heterogeneity was detected between different studies (Q test, P<0.001). Moreover, Begg's test, Egger's tests and funnel plots indicated no significant publication bias between included studies [Begg's test (P=0.371); Egger's test (p=0.100)]. CONCLUSION The present meta-analysis demonstrated that IBD was associated with an increased diagnosis of PC. In addition, large scale prospective studies are essential to determine whether IBD increase the PC risk.
Collapse
Affiliation(s)
- Mingshi Chen
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, N°321 Zhongshan Road, 210008 Nanjing, Jiangsu, China
| | - Caihong Yuan
- Department of Endocrinology, Kunshan Hospital on Integration of Chinese and Western Medicine, 215332 Jiangsu, China
| | - Tianshu Xu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, N°321 Zhongshan Road, 210008 Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Hacioglu C, Kacar S, Kar F, Kanbak G, Sahinturk V. Concentration-Dependent Effects of Zinc Sulfate on DU-145 Human Prostate Cancer Cell Line: Oxidative, Apoptotic, Inflammatory, and Morphological Analyzes. Biol Trace Elem Res 2020; 195:436-444. [PMID: 31463762 DOI: 10.1007/s12011-019-01879-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/19/2019] [Indexed: 12/31/2022]
Abstract
Zinc takes part in several of cellular signaling pathways, containing defense against free radicals, apoptosis, and inflammation. However, interaction between zinc and prostate cancer progression is poorly understood. Therefore, zinc treatment in DU-145 human prostate cancer cells was investigated. First, zinc sulfate (ZnSO4) concentrations with antiproliferative effect were determined using MTT assay. Then, ZnSO4-induced oxidative damage was evaluated by malondialdehyde (MDA) levels, glutathione (GSH) levels, total oxidant status (TOS) levels, and total antioxidant status (TAS) levels. Apoptotic effects of ZnSO4 were determined by measuring biochemical and immunohistochemical parameters including caspase 3 (CASP3), cytochrome C (CYC), Bcl-2-associated X protein (Bax), and B cell CLL/lymphoma 2 (Bcl-2) levels. Inflammatory effects of ZnSO4 were investigated by measuring interleukin-6 (IL-6) levels and tumor necrosis factor-alpha (TNF-α) levels. Finally, morphological analysis was performed using hematoxylin-eosin staining. We found that ZnSO4 caused a concentration-dependent increase in oxidative stress, apoptosis, and inflammation pathways. Moreover, there were a number of morphological alterations in treated cells depending on the ZnSO4 concentration. Consequently, our data showed that zinc acts as a regulator of increased oxidative damage and apoptosis through the upregulation of TNF-α and IL-6.
Collapse
Affiliation(s)
- Ceyhan Hacioglu
- Department of Medical Biochemistry, Faculty of Medicine, Duzce University, Duzce, Turkey.
| | - Sedat Kacar
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Gungor Kanbak
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Varol Sahinturk
- Department of Histology and Embryology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
20
|
The effect of Metapanax delavayi leaf extract on testosterone‐induced benign prostatic hyperplasia in rats. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
21
|
Cytokines and Janus kinase/signal transducer and activator of transcription signaling in prostate cancer: overview and therapeutic opportunities. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.coemr.2020.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Komarova EY, Marchenko LV, Zhakhov AV, Nikotina AD, Aksenov ND, Suezov RV, Ischenko AM, Margulis BA, Guzhova IV. Extracellular Hsp70 Reduces the Pro-Tumor Capacity of Monocytes/Macrophages Co-Cultivated with Cancer Cells. Int J Mol Sci 2019; 21:ijms21010059. [PMID: 31861801 PMCID: PMC6982218 DOI: 10.3390/ijms21010059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer cells are known to contain high levels of the heat shock protein 70 kDa (Hsp70), which mediates increased cell proliferation, escape from programmed cell death, enhanced invasion, and metastasis. A part of Hsp70 molecules may release from cancer cells and affect the behavior of adjacent stromal cells. To explore the effects of Hsp70 on the status of monocytes/macrophages in the tumor locale, we incubated human carcinoma cells of three distinct lines with normal and reduced content of Hsp70 with THP1 monocytes. Using two methods, we showed that the cells with knock-down of Hsp70 released a lower amount of protein in the extracellular medium. Three cycles of the co-cultivation of cancer and monocytic cells led to the secretion of several cytokines typical of the tumor microenvironment (TME) and to pro-cancer activation of the monocytes/macrophages as established by elevation of F4/80 and arginase-1 markers. Unexpectedly, the efficacy of epithelial–mesenchymal transition and resistance of carcinoma cells to anticancer drugs after incubation with monocytic cells were more pronounced in cells with lower Hsp70, e.g., releasing less Hsp70 into the extracellular milieu. These data suggest that Hsp70 released from tumor cells into the TME is able, together with the development of an anti-cancer immune response, to limit the conversion of a considerable part of monocytic cells to the pro-tumor phenotype.
Collapse
Affiliation(s)
- Elena Y. Komarova
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
| | - Larisa V. Marchenko
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
| | - Alexander V. Zhakhov
- Institute of Highly Pure Biopreparation of Federal Medical and Biological Agency of Russia, Pudozhskaya street, 7, St. Petersburg 197110, Russia; (A.V.Z.); (A.M.I.)
| | - Alina D. Nikotina
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
| | - Nikolay D. Aksenov
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
| | - Roman V. Suezov
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
| | - Alexander M. Ischenko
- Institute of Highly Pure Biopreparation of Federal Medical and Biological Agency of Russia, Pudozhskaya street, 7, St. Petersburg 197110, Russia; (A.V.Z.); (A.M.I.)
| | - Boris A. Margulis
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
| | - Irina V. Guzhova
- Laboratory of Cell Protection Mechanisms, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia; (E.Y.K.); (L.V.M.); (A.D.N.); (N.D.A.); (R.V.S.); (B.A.M.)
- Correspondence: ; Tel.: +7812-2973794
| |
Collapse
|
23
|
Do small cities affect bird assemblages? An evaluation from Patagonia. Urban Ecosyst 2019. [DOI: 10.1007/s11252-019-00915-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
24
|
Ahmad AE, Mohammed A, Bhindi B, Richard PO, Fadaak K, Leão R, Finelli A, Fleshner NE, Kulkarni GS. Serum Adipokines as Predictors for the Outcome of Prostate Biopsies at Early Stage Prostate Cancer Diagnosis. Cancer Manag Res 2019; 11:10043-10050. [PMID: 31819637 PMCID: PMC6890197 DOI: 10.2147/cmar.s226174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/26/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose Elevated adipokines in patients with obesity and metabolic syndrome have been linked to increased risk of prostate cancer (PCa). The association between select serum adipokines and the outcome of prostate biopsies alone and in combination with clinical parameters at different early stages of PCa was investigated. Patients and methods Clinical data and serum adipokines were retrieved from three retrospective cohorts representing men at different points in PCa detection: 1. Subjects with no prior biopsies (n=1061), 2. subjects with a prior negative biopsy (REDUCE trial, control arm) (n=1209), 3. subjects with low-risk PCa on active surveillance (AS) (n=154). Adipokines were chosen based on an unpublished pilot study and included: Resistin, Tumor Necrosis Factor-α, Interleukin-6, Monocyte Chemoattractant Protein-1, Hepatocyte Growth Factor, and Nerve Growth Factor. The primary outcome was the absence of PCa on biopsy and the secondary outcome was diagnosis of low-risk PCa fitting the criteria for continuing AS. Logistic regression analysis was used to assess the association of adipokines and negative and/or low-risk PCa at prostate biopsy. Results In men with no prior prostate biopsy or with prior negative biopsy, adipokines were not predictors of prostate biopsy outcomes on multivariable regression analysis controlling for known clinical variables. In the AS cohort, MCP-1 and Resistin were significant predictors of biopsy outcome on multivariable analysis (OR 0.20, 95% CI: 0.05–0.85, p= 0.03 & OR 0.30, 95% CI: 0.10 −0.86, p= 0.03). Conclusion Our findings do not support a strong role for adipokines for predicting the outcome of prostate biopsies at any early stage in PCa diagnosis.
Collapse
Affiliation(s)
- Ardalan E Ahmad
- Division of Urology, Department of Surgery, University Health Network, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Aza Mohammed
- Department of Urology, Luton and Dunstable University Hospital, Luton, UK
| | - Bimal Bhindi
- Division of Urology, Department of Surgery, University Health Network, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,Southern Alberta Institute of Urology, Calgary, Alberta, Canada
| | - Patrick O Richard
- Division of Urology, Department of Surgery, Centre Hospitalier Universitaire de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Kamel Fadaak
- Department of Urology, King Fahd Hospital of the University, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ricardo Leão
- CUF Department of Urology, Hospital De Braga, Faculty of Medicine, University of Coimbra, Portugal
| | - Antonio Finelli
- Division of Urology, Department of Surgery, University Health Network, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Neil E Fleshner
- Division of Urology, Department of Surgery, University Health Network, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Girish S Kulkarni
- Division of Urology, Department of Surgery, University Health Network, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Connor MJ, Shah TT, Horan G, Bevan CL, Winkler M, Ahmed HU. Cytoreductive treatment strategies for de novo metastatic prostate cancer. Nat Rev Clin Oncol 2019; 17:168-182. [PMID: 31712648 DOI: 10.1038/s41571-019-0284-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
In the past decade, a revolution in the treatment of metastatic prostate cancer has occurred with the advent of novel hormonal agents and life-prolonging chemotherapy regimens in combination with standard androgen-deprivation therapy. Notwithstanding, the use of systemic therapy alone can result in a castrate-resistant state; therefore, increasing focus is being placed on the additional survival benefits that could potentially be achieved with local cytoreductive and/or metastasis-directed therapies. Local treatment of the primary tumour with the established modalities of radiotherapy and radical prostatectomy has been explored in this context, and the use of novel minimally invasive ablative therapies has been proposed. In addition, evidence of the potential clinical benefits of metastasis-directed therapy with ionizing radiation (primarily stereotactic ablative radiotherapy) is accumulating. Herein, we summarize the pathobiological rationale for local cytoreduction and the potentially systemic immunological responses to radiotherapy and ablative therapies in patients with metastatic prostate cancer. We also discuss the current evidence base for a cytoreductive strategy, including metastasis-directed therapy, in the current era of sequential multimodal therapy incorporating novel treatments. Finally, we outline further research questions relating to this complex and evolving treatment landscape.
Collapse
Affiliation(s)
- Martin J Connor
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK. .,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK.
| | - Taimur T Shah
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Gail Horan
- Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Charlotte L Bevan
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Mathias Winkler
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Hashim U Ahmed
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.,Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
26
|
Chen X, Chen F, Ren Y, Weng G, Xu L, Xue X, Keng PC, Lee SO, Chen Y. IL-6 signaling contributes to radioresistance of prostate cancer through key DNA repair-associated molecules ATM, ATR, and BRCA 1/2. J Cancer Res Clin Oncol 2019; 145:1471-1484. [PMID: 31020420 DOI: 10.1007/s00432-019-02917-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE To study an association between IL-6 signaling and resistance to radiotherapy of prostate cancer (PCa) and explore the molecular mechanisms involved. METHODS IL-6 expressing C4-2 and CWR22Rv1 (C4-2IL-6/CWRIL-6) and vector control (C4-2vec/CWRvec) cell lines were developed. Radiation-sensitivities of these cells were compared in clonogenic assay, Comet assay, and γH2AX staining. In xenograft animal studies, radiation-sensitivity of C4-2IL-6 cell-derived tumors vs. C4-2vec cell-derived tumors was investigated. To reveal IL-6 downstream molecules involved in DNA repair after radiation, qPCR and Western blot analyses as well as immunofluorescence staining were performed. Transcriptional control of IL-6 on ATM and ATR molecules was also investigated. RESULTS We found C4-2IL-6 and CWRIL-6 cells survived better than their vector control cells after irradiation, and animal studies confirmed such in vitro results. We discovered that DNA repair-related molecules such as ATM, ATR, BRCA1, and BRCA2 were significantly upregulated in irradiated IL-6 expressing cells compared with vector control cells. We further defined that IL-6 signaling regulated cellular expressions of ATM and ATR at the transcriptional level through the activation of Stat3 signaling pathway. CONCLUSIONS IL-6 leads to PCa resistance to radiation through upregulation of DNA repair associated molecules ATM, ATR, BRCA1, and BRCA2.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Feng Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Yu Ren
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Guobin Weng
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Lijun Xu
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Xiang Xue
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA.
| | - Yuhchyau Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA.
| |
Collapse
|
27
|
McAllister MJ, Underwood MA, Leung HY, Edwards J. A review on the interactions between the tumor microenvironment and androgen receptor signaling in prostate cancer. Transl Res 2019; 206:91-106. [PMID: 30528321 DOI: 10.1016/j.trsl.2018.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/23/2018] [Accepted: 11/19/2018] [Indexed: 12/29/2022]
Abstract
Prostate cancer growth is controlled by androgen receptor signaling via both androgen-dependent and androgen-independent pathways. Furthermore, the prostate is an immune competent organ with inflammatory changes both within the systemic and local environment contributing to the reprogramming of the prostatic epithelium with consistently elevated lymphocyte infiltration and proinflammatory cytokines being found in prostate cancer. The crosstalk between the tumor microenvironment and androgen receptor signaling is complex with both protumorigenic and antitumorigenic roles observed. However, despite an increase in immune checkpoint inhibitors and inflammatory signaling blockades available for a range of cancer types, we are yet to see substantial progress in the treatment of prostate cancer. Therefore, this review aims to summarize the tumor microenvironment and its impact on androgen receptor signaling in prostate cancer.
Collapse
Affiliation(s)
- Milly J McAllister
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| | - Mark A Underwood
- Department of Urology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Hing Y Leung
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Urology, Queen Elizabeth University Hospital, Glasgow, United Kingdom; Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Joanne Edwards
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
28
|
Jiang W, Ji M. Receptor tyrosine kinases in PI3K signaling: The therapeutic targets in cancer. Semin Cancer Biol 2019; 59:3-22. [PMID: 30943434 DOI: 10.1016/j.semcancer.2019.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/09/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway, one of the most commonly activated signaling pathways in human cancers, plays a crucial role in the regulation of cell proliferation, differentiation, and survival. This pathway is usually activated by receptor tyrosine kinases (RTKs), whose constitutive and aberrant activation is via gain-of-function mutations, chromosomal rearrangement, gene amplification and autocrine. Blockage of PI3K pathway by targeted therapy on RTKs with tyrosine kinases inhibitors (TKIs) and monoclonal antibodies (mAbs) has achieved great progress in past decades; however, there still remain big challenges during their clinical application. In this review, we provide an overview about the most frequently encountered alterations in RTKs and focus on current therapeutic agents developed to counteract their aberrant functions, accompanied with discussions of two major challenges to the RTKs-targeted therapy in cancer - resistance and toxicity.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
29
|
Gaudreau PO, Clairefond S, Class CA, Boulay PL, Chrobak P, Allard B, Azzi F, Pommey S, Do KA, Saad F, Trudel D, Young M, Stagg J. WISP1 is associated to advanced disease, EMT and an inflamed tumor microenvironment in multiple solid tumors. Oncoimmunology 2019; 8:e1581545. [PMID: 31069142 PMCID: PMC6492985 DOI: 10.1080/2162402x.2019.1581545] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/21/2019] [Accepted: 02/06/2019] [Indexed: 02/08/2023] Open
Abstract
Background: WNT1-Inducible Signaling Pathway Protein 1 (WISP1) is implicated in prostate cancer growth and metastasis and the regulation of inflammation in diverse benign diseases. The objectives of this study were to assess the prognostic value of WISP1, its association to inflammation and its relevance as a biomarker for immune checkpoint blockade (ICB) response. Methods: Publicly available RNA-seq datasets were used to evaluate the prognostic value of WISP1 gene expression and its association with tumor-infiltrating lymphocytes, inflamed tumor microenvironment, and anti-PD-1 ICB response. A tissue microarray (TMA) including 285 radical prostatectomy specimens was used to confirm these associations in prostate cancer. The effect of recombinant WISP1 (rWISP1) on inflammatory cytokines was assessed in vitro. Results: High levels of WISP1 correlated with BCR-free survival in prostate adenocarcinoma and overall survival in primary melanoma, low-grade glioma, and kidney papillary cell carcinoma. Some effects could be accounted for by higher WISP1 expression in advanced disease. High WISP1 expression in prostate adenocarcinoma was correlated with CD8+ cells density. In vitro, rWISP1 increased inflammatory cytokine production. High WISP1 gene expression in RNA-seq datasets was correlated with gene signatures of multiple immune cell types as well as an inflammatory cytokine, immune checkpoint, and epithelial-mesenchymal transition (EMT) gene expression. WISP1 mRNA expression was associated with primary resistance to ICB in datasets showing EMT. Conclusions: Our results support an association between WISP1 expression and advanced disease, EMT and an inflamed tumor microenvironment in multiple solid tumors. The consequences of WISP1 expression on cancer immunotherapy remains to be addressed.
Collapse
Affiliation(s)
- Pierre-Olivier Gaudreau
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sylvie Clairefond
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Caleb A Class
- T. Boone Pickens Academic Tower, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pierre-Luc Boulay
- Département de pharmacologie et de physiologie, Université de Montréal, Montreal, QC, Canada
| | - Pavel Chrobak
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Bertrand Allard
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Feryel Azzi
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Sandra Pommey
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Kim-Anh Do
- T. Boone Pickens Academic Tower, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fred Saad
- Département d'Urologie du Centre Hospitalier Universitaire de Montréal (CHUM) et Institut du Cancer de Montréal / CRCHUM, Montreal, QC, Canada
| | - Dominique Trudel
- Centre Hospitalier de l'Université de Montréal (Département de pathologie), Département de pathologie et axe cancer, Université de Montréal (Département de pathologie et de biologie cellulaire) et Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marian Young
- NIDCR, National Institutes of Health, Bethesda, MD, USA
| | - John Stagg
- Faculté de Pharmacie, Université de Montréal et Institut du Cancer de Montréal / CRCHUM, Axe Cancer, Montreal, QC, Canada
| |
Collapse
|
30
|
Baci D, Gallazzi M, Cascini C, Tramacere M, De Stefano D, Bruno A, Noonan DM, Albini A. Downregulation of Pro-Inflammatory and Pro-Angiogenic Pathways in Prostate Cancer Cells by a Polyphenol-Rich Extract from Olive Mill Wastewater. Int J Mol Sci 2019; 20:ijms20020307. [PMID: 30646518 PMCID: PMC6359159 DOI: 10.3390/ijms20020307] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/18/2022] Open
Abstract
Dietary phytochemicals are particularly attractive for chemoprevention and are able to modulate several signal transduction pathways linked with cancer. Olive oil, a major component of the Mediterranean diet, is an abundant source of phenolic compounds. Olive oil production is associated with the generation of a waste material, termed 'olive mill wastewater' (OMWW) that have been reported to contain water-soluble polyphenols. Prostate cancer (PCa) is considered as an ideal cancer type for chemopreventive approaches, due to its wide incidence but relatively long latency period and progression time. Here, we investigated activities associated with potential preventive properties of a polyphenol-rich olive mill wastewater extract, OMWW (A009), on three in vitro models of PCa. A009 was able to inhibit PCa cell proliferation, adhesion, migration, and invasion. Molecularly, we found that A009 targeted NF-κB and reduced pro-angiogenic growth factor, VEGF, CXCL8, and CXCL12 production. IL-6/STAT3 axis was also regulated by the extract. A009 shows promising properties, and purified hydroxytyrosol (HyT), the major polyphenol component of A009, was also active but not always as effective as A009. Finally, our results support the idea of repositioning a food waste-derived material for nutraceutical employment, with environmental and industrial cost management benefits.
Collapse
Affiliation(s)
- Denisa Baci
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Matteo Gallazzi
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milano, Italy.
| | - Caterina Cascini
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milano, Italy.
| | - Matilde Tramacere
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milano, Italy.
| | | | - Antonino Bruno
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milano, Italy.
| | - Douglas M Noonan
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milano, Italy.
- Department of Biotechnology and Life Sciences, Laboratory of Immunology and General Pathology, University of Insubria, 21100 Varese, Italy.
| | - Adriana Albini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy.
- Scientific and Technology Pole, IRCCS MultiMedica, 20138 Milano, Italy.
| |
Collapse
|
31
|
Marchenko LV, Nikotina AD, Aksenov ND, Smagina LV, Margulis BA, Guzhova IV. Phenotypic Characteristics of Macrophages and Tumor Cells in Coculture. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s1990519x18050036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
32
|
Dargahi Abbasabad G, Banan Khojasteh SM, Eskandari Naji H, Zamani MR, Hajipour H, Serati-Nouri H. An Interleukin-6 Single Nucleotide Polymorphism and Susceptibility to Prostate Adenocarcinoma and Bone Metastasis
in an Iranian Population. Asian Pac J Cancer Prev 2018; 19:1717-1720. [PMID: 29938471 PMCID: PMC6103583 DOI: 10.22034/apjcp.2018.19.6.1717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: Interleukin-6 (IL-6) is an inflammatory cytokine shown to be a strong factor for growth, proliferation and metastasis with many malignancies. The promoter single nucleotide polymorphism (SNPs) -174G>C (rs1800795) can alter the transcriptional pattern of this gene. The present study was aimed at assessing effects of the IL-6 (rs1800795) SNP on risk of benign prostate hyperplasia (BPH) and prostatic adenocarcinoma (PCa). Methods: The project was performed on 112 men with PCa, 118 with BPH and 250 healthy controls. After DNA extraction, genotyping of IL-6 (rs1800795) was performed using PCR TaqMan Allelic Discrimination (ABI MGB). Results: The G allele frequency for rs1800795 of the IL-6 gene was 74.1%, 68.6% and 67% in PCa patients, BPH patients and healthy men, respectively. PCa and control groups showed significant differences (P =0.030, OR = 1.73, 95% CI: 1.05-2.21). The GG genotype was more frequent in the PCa group, whereas the GC genotype was more common in the BPH in comparison to other groups. Conclusion: The current study identified IL-6 -174G>C (rs1800795) as a significant predictor of susceptibility for prostate cancer and bone metastasis in a northwest Iranian population.
Collapse
|
33
|
Zhou L, Zheng Y, Tian T, Liu K, Wang M, Lin S, Deng Y, Dai C, Xu P, Hao Q, Kang H, Dai Z. Associations of interleukin-6 gene polymorphisms with cancer risk: Evidence based on 49,408 cancer cases and 61,790 controls. Gene 2018; 670:136-147. [PMID: 29842912 DOI: 10.1016/j.gene.2018.05.104] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/03/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022]
Abstract
Many molecular epidemiologic studies have shown that interleukin-6 (IL-6) polymorphisms are significantly associated with susceptibility for various cancers. However, the conclusions of these studies are inconsistent. The purpose of the present study was to explore the association between three common IL-6 loci (rs1800795, rs1800796, and rs1800797) and the risk for various cancers. We systematically searched the PubMed, Web of Science, Wanfang and China national knowledge infrastructure (CNKI) databases for relevant publications and obtained 108 eligible studies, involving 49,408 cancer patients and 61,790 cancer-free controls. Odds ratio (OR), 95% confidence interval (CI), and false positive reporting probability (FPRP) were used to evaluate cancer risk. All statistical analyses were performed using the R software meta package. We observed a non-significant association between rs1800795 and overall cancer risk, while rs1800797 was found to have a false positive association with overall risk of cancer. Subgroup analyses of rs1800797 also suggested non-significant association and rs1800795 played a protective role in liver cancer. Rs1800796 was found to be associated with overall cancer risk, particularly in Asian patients and those with prostate cancer. These findings provide evidence that IL-6 polymorphisms may affect cancer risk.
Collapse
Affiliation(s)
- Linghui Zhou
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yi Zheng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Tian Tian
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Kang Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Meng Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuai Lin
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yujiao Deng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Cong Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Peng Xu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Qian Hao
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Huafeng Kang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Zhijun Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
34
|
Duscharla D, Reddy Kami Reddy K, Dasari C, Bhukya S, Ummanni R. Interleukin-6 induced overexpression of valosin-containing protein (VCP)/p97 is associated with androgen-independent prostate cancer (AIPC) progression. J Cell Physiol 2018; 233:7148-7164. [PMID: 29693262 DOI: 10.1002/jcp.26639] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/30/2018] [Indexed: 12/21/2022]
Abstract
Though Androgen deprivation therapy (ADT) is effective initially, numerous patients become resistant to it and develop castration resistant PCa (CRPC). Cytokines promotes ligand independent activation of AR. Interleukin-6 (IL-6) levels are elevated in CRPC patients and regulate AR activity. However, progression to CRPC is not fully understood. In this study, we analyzed differential protein expression in LNCaP cells treated with IL-6 using proteomics. Results revealed altered expression of 27 proteins and Valosin-containing protein (VCP)/p97 plays a predominant role in co-regulation of altered proteins. Interestingly, IL-6 induced VCP expression through Pim-1 via STAT3 is AR independent there by suggesting a role for VCP in CRPC. Transfection of LNCaP cells for VCP overexpression showed an increased cell proliferation, migration, and invasion where as its inhibition by NMS-873 showed the reverse effect causing cell death. Mechanistic studies demonstrate that cell death occurs due to apoptosis by endoplasmic reticulum (ER) stress, elevated cell cycle inhibitors p21, p27kip1, and active PARP and reduced Bcl-2. VCP promotes cell invasion and migration by altering E-cadherin and Vimentin levels inversely triggering EMT of PCa cells. VCP immunostaining revealed no staining in BPH but strong staining in PCa. This study determines VCP may play an important role in progression to CRPC and it can be a favorable target with to develop new therapies to treat ADT resistant prostate cancer.
Collapse
Affiliation(s)
- Divya Duscharla
- Center for Chemical Biology, Indian Institute of Chemical Technology (IICT), Hyderabad, India.,Center for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Karthik Reddy Kami Reddy
- Center for Chemical Biology, Indian Institute of Chemical Technology (IICT), Hyderabad, India.,Center for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Chandrashekhar Dasari
- Center for Chemical Biology, Indian Institute of Chemical Technology (IICT), Hyderabad, India.,Center for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| | - Supriya Bhukya
- Center for Chemical Biology, Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Ramesh Ummanni
- Center for Chemical Biology, Indian Institute of Chemical Technology (IICT), Hyderabad, India.,Center for Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
| |
Collapse
|
35
|
Culig Z, Puhr M. Interleukin-6 and prostate cancer: Current developments and unsolved questions. Mol Cell Endocrinol 2018; 462:25-30. [PMID: 28315704 DOI: 10.1016/j.mce.2017.03.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/27/2017] [Accepted: 03/13/2017] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-6 is a pro-inflammatory cytokine that is expressed in prostate tumors and in the stromal tumor micro-enviroment. It is known to regulate proliferation, apoptosis, angiogenesis, and differentiation. The signaling pathway of Janus kinase and signal transducer and activator of transcription (STAT)3, which is activated by IL-6, is in the focus of scientific investigations for improved treatment approaches. Different effects of IL-6 and/or STAT3 on tumor cell growth have been observed in human and murine prostate cancer (PCa) models. Experimental therapies have been proposed in order to block the IL-6/STAT3 signaling pathway. In this context, the anti-IL-6 antibody siltuximab (CNTO 328) has been demonstrated to inhibit growth of prostate tumors in vitro and in vivo and delays progression towards castration resistance. However, clinically, the anti-IL-6 antibody was not successful as a monotherapy in phase II studies in patients with metastatic PCa. IL-6 is implicated in regulation of cellular stemness by increasing phosphorylation of STAT3. The cytokine has also a role in development of resistance to the non-steroidal anti-androgen enzalutamide. Endogenous inhibitors of IL-6 are suppressors of cytokine signaling and protein inhibitors of activated STAT. Although they inhibit signal transduction through STAT3, they may also exhibit anti-apoptotic effects. On the basis of complexity of IL-6 action in PCa, an individualized approach is needed to identify patients who will benefit from anti-IL-6 therapy in combination with standard treatments.
Collapse
Affiliation(s)
- Zoran Culig
- Experimental Urology, Department of Urology, Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria.
| | - Martin Puhr
- Experimental Urology, Department of Urology, Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| |
Collapse
|
36
|
Constructing Bayesian networks by integrating gene expression and copy number data identifies NLGN4Y as a novel regulator of prostate cancer progression. Oncotarget 2018; 7:68688-68707. [PMID: 27626693 PMCID: PMC5356583 DOI: 10.18632/oncotarget.11925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/24/2016] [Indexed: 12/27/2022] Open
Abstract
To understand the heterogeneity of prostate cancer (PCa) and identify novel underlying drivers, we constructed integrative molecular Bayesian networks (IMBNs) for PCa by integrating gene expression and copy number alteration data from published datasets. After demonstrating such IMBNs with superior network accuracy, we identified multiple sub-networks within IMBNs related to biochemical recurrence (BCR) of PCa and inferred the corresponding key drivers. The key drivers regulated a set of common effectors including genes preferentially expressed in neuronal cells. NLGN4Y—a protein involved in synaptic adhesion in neurons—was ranked as the top gene closely linked to key drivers of myogenesis subnetworks. Lower expression of NLGN4Y was associated with higher grade PCa and an increased risk of BCR. We show that restoration of the protein expression of NLGN4Y in PC-3 cells leads to decreased cell proliferation, migration and inflammatory cytokine expression. Our results suggest that NLGN4Y is an important negative regulator in prostate cancer progression. More importantly, it highlights the value of IMBNs in generating biologically and clinically relevant hypotheses about prostate cancer that can be validated by independent studies.
Collapse
|
37
|
Ding G, Wang J, Feng C, Jiang H, Xu J, Ding Q. Lipocalin 2 over-expression facilitates progress of castration-resistant prostate cancer via improving androgen receptor transcriptional activity. Oncotarget 2018; 7:64309-64317. [PMID: 27602760 PMCID: PMC5325444 DOI: 10.18632/oncotarget.11790] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/13/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) is the lethal phenotype of prostate cancer. Lipocalin 2 (LCN2) is aberrantly expressed in many cancers including primary prostate cancer (PCa), but its role in CRPC has not been reported. RESULTS LCN2 expression was upregulated in human primary PCa and CRPC tissues. Overexpression of LCN2 promoted C4-2B and 22RV1 cell proliferation while knockdown of LCN2 markedly inhibited C4-2B and 22RV1 cell growth. LCN2 overexpression led to increased AR downstream gene SLC45A3 without upregulating AR expression. In the xenograft model, overexpression of LCN2 significantly promoted tumor growth. METHODS LCN2 expression was detected in primary PCa and CRPC tissues and cell lines C4-2B and 22RV1 using immunohistochemistry and western blotting, respectively. Serum LCN2 level was detected vi ELISA. Lentiviruses-mediated over-expression of LCN2 and LCN2 knockdown were performed in CRPC cell lines. Expressions of androgen receptor (AR) downstream genes was examined in cell lines, in CRPC tissues, and in animal models. CONCLUSION LCN2 could facilitate cell proliferation of CRPC via AR transcriptional activity. LCN2 could be a novel target in CRPC.
Collapse
Affiliation(s)
- Guanxiong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianqing Wang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Department of Urology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Kumar S, Singh R, Malik S, Manne U, Mishra M. Prostate cancer health disparities: An immuno-biological perspective. Cancer Lett 2018; 414:153-165. [PMID: 29154974 PMCID: PMC5743619 DOI: 10.1016/j.canlet.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) is the most commonly diagnosed malignancy in males, and, in the United States, is the second leading cause of cancer-related death for men older than 40 years. There is a higher incidence of PCa for African Americans (AAs) than for European-Americans (EAs). Investigations related to the incidence of PCa-related health disparities for AAs suggest that there are differences in the genetic makeup of these populations. Other differences are environmentally induced (e.g., diet and lifestyle), and the exposures are different. Men who immigrate from Eastern to Western countries have a higher risk of PCa than men in their native countries. However, the number of immigrants developing PCa is still lower than that of men in Western countries, suggesting that genetic factors are involved in the development of PCa. Altered genetic polymorphisms are associated with PCa progression. Androgens and the androgen receptor (AR) are involved in the development and progression of PCa. For populations with diverse racial/ethnic backgrounds, differences in lifestyle, diet, and biology, including genetic mutations/polymorphisms and levels of androgens and AR, are risk factors for PCa. Here, we provide an immuno-biological perspective on PCa in relation to racial/ethnic disparities and identify factors associated with the disproportionate incidence of PCa and its clinical outcomes.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shalie Malik
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA; Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Upender Manne
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Manoj Mishra
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
39
|
Danziger O, Pupko T, Bacharach E, Ehrlich M. Interleukin-6 and Interferon-α Signaling via JAK1-STAT Differentially Regulate Oncolytic versus Cytoprotective Antiviral States. Front Immunol 2018; 9:94. [PMID: 29441069 PMCID: PMC5797546 DOI: 10.3389/fimmu.2018.00094] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/12/2018] [Indexed: 12/17/2022] Open
Abstract
Malignancy-induced alterations to cytokine signaling in tumor cells differentially regulate their interactions with the immune system and oncolytic viruses. The abundance of inflammatory cytokines in the tumor microenvironment suggests that such signaling plays key roles in tumor development and therapy efficacy. The JAK-STAT axis transduces signals of interleukin-6 (IL-6) and interferons (IFNs), mediates antiviral responses, and is frequently altered in prostate cancer (PCa) cells. However, how activation of JAK-STAT signaling with different cytokines regulates interactions between oncolytic viruses and PCa cells is not known. Here, we employ LNCaP PCa cells, expressing (or not) JAK1, activated (or not) with IFNs (α or γ) or IL-6, and infected with RNA viruses of different oncolytic potential (EHDV-TAU, hMPV-GFP, or HIV-GFP) to address this matter. We show that in JAK1-expressing cells, IL-6 sensitized PCa cells to viral cell death in the presence or absence of productive infection, with dependence on virus employed. Contrastingly, IFNα induced a cytoprotective antiviral state. Biochemical and genetic (knockout) analyses revealed dependency of antiviral state or cytoprotection on STAT1 or STAT2 activation, respectively. In IL-6-treated cells, STAT3 expression was required for anti-proliferative signaling. Quantitative proteomics (SILAC) revealed a core repertoire of antiviral IFN-stimulated genes, induced by IL-6 or IFNs. Oncolysis in the absence of productive infection, induced by IL-6, correlated with reduction in regulators of cell cycle and metabolism. These results call for matching the viral features of the oncolytic agent, the malignancy-induced genetic-epigenetic alterations to JAK/STAT signaling and the cytokine composition of the tumor microenvironment for efficient oncolytic virotherapy.
Collapse
Affiliation(s)
- Oded Danziger
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tal Pupko
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eran Bacharach
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Yen MC, Huang YC, Kan JY, Kuo PL, Hou MF, Hsu YL. S100B expression in breast cancer as a predictive marker for cancer metastasis. Int J Oncol 2017; 52:433-440. [PMID: 29345293 DOI: 10.3892/ijo.2017.4226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/05/2017] [Indexed: 11/06/2022] Open
Abstract
In the tumor microenvironment, soluble molecules play important role in the establishment of a pre-metastatic niche. The S100 calcium-binding protein family are inflammatory molecules that contribute to the development of a pro-inflammatory tumor microenvironment. S100B belongs to the S100 family and serum S100B (also known as S100beta) serves as a marker for metastasis in lung cancer, ovarian cancer and melanoma. However, the association between S100B and the metastasis of breast cancer is not yet well understood. In the present study, a relatively low S100B expression was observed in the tumor samples compared to normal breast tissue among online microarray datasets. When the estrogen receptor (ER)-negative breast cancer cell lines, MDA-MB-231 and Hs578T, were treated with recombinant human S100B, cell migration was significantly inhibited and epithelial cadherin expression was increased. Our results revealed that a high S100B expression predicted a good overall survival in patients with ER-negative breast cancer, and good distant metastases-free survival in all patients with breast cancer via the analysis of the KM plotter and SurvExpress databases. Although previous studies have indicated that the interaction of S100B with wild-type p53 inhibits p53 function, a high S100B expression is associated with a good prognosis in patients with p53 mutant and p53 wild-type breast cancers. On the whole, our findings demonstrate that S100B treatment suppresses the migratory capacity of ER-negative breast cancer and that S100B expression may serve a predictive marker for metastasis in breast cancer.
Collapse
Affiliation(s)
- Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Yung-Chi Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Jung-Yu Kan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ming-Feng Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ya-Ling Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
41
|
Dominguez C, David JM, Palena C. Epithelial-mesenchymal transition and inflammation at the site of the primary tumor. Semin Cancer Biol 2017; 47:177-184. [PMID: 28823497 PMCID: PMC5698091 DOI: 10.1016/j.semcancer.2017.08.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Tumor growth and progression are the products of complex signaling networks between different cell types within the tumor and its surrounding stroma. In particular, established tumors are known to stimulate an inflammatory reaction via the secretion of cytokines, chemokines, and growth factors that favor the recruitment of a range of infiltrating immune cell populations into the tumor microenvironment. While potentially able to exert tumor control, this inflammatory reaction is typically seized upon by the tumor to promote its own growth and progression towards metastasis. This review focuses on recent advances in understanding how an established tumor can initiate an inflammatory response via the release of pro-inflammatory mediators, such as IL-6 and IL-8, and their roles in cancer metastasis. In particular, the role of the epithelial-mesenchymal transition (EMT), a phenotypic switch observed in carcinomas that promotes progression towards metastasis, is discussed here in relation to cancer inflammation.
Collapse
Affiliation(s)
- Charli Dominguez
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Justin M David
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States.
| |
Collapse
|
42
|
High miR-205 expression in normal epithelium is associated with biochemical failure - an argument for epithelial crosstalk in prostate cancer? Sci Rep 2017; 7:16308. [PMID: 29176717 PMCID: PMC5701197 DOI: 10.1038/s41598-017-16556-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 11/14/2017] [Indexed: 12/31/2022] Open
Abstract
Due to insufficient prognostic tools, failure to predict aggressive prostate cancer (PC) has left patient selection for radical treatment an unsolved challenge. This has resulted in overtreatment with radical therapy. Better prognostic tools are urgently warranted. MicroRNAs (miRs) have emerged as important regulators of cellular pathways, resulting in altered gene expressions. miR-205 has previously been observed downregulated in PC, acting as tumor suppressor. Herein, the expression of miR-205 in prostate tissue was examined in a large, well-described cohort of 535 Norwegian prostatectomy patients. Using in situ hybridization, miR-205 expression was semiquantatively measured in normal and tumor tissues from radical prostatectomy specimens. Associations with clinicopathological data and PC relapse were calculated. Expression of miR-205 was lower in tumor epithelium compared to normal epithelium. No association was observed between miR-205 expression in primary tumor epithelium and cancer relapse. In contrast, high expression of miR-205 in normal epithelium was independently associated with biochemical relapse (HR = 1.64, p = 0.003). A prognostic importance of miR-205 expression was only found in the normal epithelium, raising the hypothesis of epithelial crosstalk between normal and tumor epithelium in PC. This finding supports the proposed novel hypothesis of an anti-cancerogenous function of normal epithelium in tumor tissue.
Collapse
|
43
|
Liu L, Li E, Luo L, Zhao S, Li F, Wang J, Luo J, Zhao Z. PSCA regulates IL-6 expression through p38/NF-κB signaling in prostate cancer. Prostate 2017; 77:1389-1400. [PMID: 28845520 DOI: 10.1002/pros.23399] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein. We previously reported that PSCA involved in proliferation and invasion of PCa cells, however, the underlying mechanisms are unknown. In this study, we aimed to explore the regulating role of PSCA gene expression in interleukin-6 (IL-6) autocrine of PCa cells. METHODS The stable knockdown-PSCA and ectopically overexpressed-PSCA vector were constructed and transfected into human PCa DU145 and PC-3M cells. The effects of PSCA overexpression or knockdown were determined in proliferation, invasion, and metastasis assays. The effect of PSCA on the expression and secretion of IL-6 was evaluated by immunoblotting and ELISA. Subcellular localization and expression pattern of PSCA and IL-6 protein were examined by immunohistochemistry. Its clinical significance was statistically analyzed. RESULTS The results showed that stable knockdown of PSCA delayed proliferation, migration, and invasion while overexpressing PSCA enhanced the proliferation, migration, and invasion in vitro and the lung metastasis in vivo of PCa cells. Importantly, the PSCA involved in the IL-6 secretion and positively regulated p38/NF-κB/IL-6 signaling, leading to enhanced PCa cell invasion and metastasis. Both the expression of PSCA and IL-6 were significantly associated with poor biochemical recurrence-free survival of patients with PCa. PSCA protein expression showed a prognostic value in overall survival as indicated by Kaplan-Meier analysis. CONCLUSIONS These results indicate that PSCA regulates the expression and secretion of IL-6 in human PCa cells through p38/NF-κB signaling pathways. PSCA may be a potential diagnostic marker and therapeutic target for PSCA-positive PCa.
Collapse
Affiliation(s)
- Luhao Liu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ermao Li
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| | - Lianmin Luo
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| | - Shankun Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| | - Futian Li
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| | - Jiamin Wang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| | - Jintai Luo
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| | - Zhigang Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of GuangZhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Katsura A, Tamura Y, Hokari S, Harada M, Morikawa M, Sakurai T, Takahashi K, Mizutani A, Nishida J, Yokoyama Y, Morishita Y, Murakami T, Ehata S, Miyazono K, Koinuma D. ZEB1-regulated inflammatory phenotype in breast cancer cells. Mol Oncol 2017; 11:1241-1262. [PMID: 28618162 PMCID: PMC5579340 DOI: 10.1002/1878-0261.12098] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/04/2017] [Indexed: 12/20/2022] Open
Abstract
Zinc finger E‐box binding protein 1 (ZEB1) and ZEB2 induce epithelial‐mesenchymal transition (EMT) and enhance cancer progression. However, the global view of transcriptional regulation by ZEB1 and ZEB2 is yet to be elucidated. Here, we identified a ZEB1‐regulated inflammatory phenotype in breast cancer cells using chromatin immunoprecipitation sequencing and RNA sequencing, followed by gene set enrichment analysis (GSEA) of ZEB1‐bound genes. Knockdown of ZEB1 and/or ZEB2 resulted in the downregulation of genes encoding inflammatory cytokines related to poor prognosis in patients with cancer, including IL6 and IL8, therefore suggesting that ZEB1 and ZEB2 have similar functions in terms of the regulation of production of inflammatory cytokines. Antibody array and ELISA experiments confirmed that ZEB1 controlled the production of the IL‐6 and IL‐8 proteins. The secretory proteins regulated by ZEB1 enhanced breast cancer cell proliferation and tumor growth. ZEB1 expression in breast cancer cells also affected the growth of fibroblasts in cell culture, and the accumulation of myeloid‐derived suppressor cells in tumors in vivo. These findings provide insight into the role of ZEB1 in the progression of cancer, mediated by inflammatory cytokines, along with the initiation of EMT.
Collapse
Affiliation(s)
- Akihiro Katsura
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yusuke Tamura
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Satoshi Hokari
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan.,Department of Respiratory Medicine and Infectious Disease, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Mayumi Harada
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan.,Department of Metabolic Care and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Japan
| | - Masato Morikawa
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Tsubasa Sakurai
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Kei Takahashi
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Anna Mizutani
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Jun Nishida
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yuichiro Yokoyama
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yasuyuki Morishita
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Takashi Murakami
- Department of Microbiology, Faculty of Medicine, Saitama Medical University, Moroyama, Japan
| | - Shogo Ehata
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
45
|
Kolosenko I, Yu Y, Busker S, Dyczynski M, Liu J, Haraldsson M, Palm Apergi C, Helleday T, Tamm KP, Page BDG, Grander D. Identification of novel small molecules that inhibit STAT3-dependent transcription and function. PLoS One 2017. [PMID: 28636670 PMCID: PMC5479526 DOI: 10.1371/journal.pone.0178844] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Activation of Signal Transducer and Activator of Transcription 3 (STAT3) has been linked to several processes that are critical for oncogenic transformation, cancer progression, cancer cell proliferation, survival, drug resistance and metastasis. Inhibition of STAT3 signaling has shown a striking ability to inhibit cancer cell growth and therefore, STAT3 has become a promising target for anti-cancer drug development. The aim of this study was to identify novel inhibitors of STAT-dependent gene transcription. A cellular reporter-based system for monitoring STAT3 transcriptional activity was developed which was suitable for high-throughput screening (Z’ = 0,8). This system was used to screen a library of 28,000 compounds (the ENAMINE Drug-Like Diversity Set). Following counter-screenings and toxicity studies, we identified four hit compounds that were subjected to detailed biological characterization. Of the four hits, KI16 stood out as the most promising compound, inhibiting STAT3 phosphorylation and transcriptional activity in response to IL6 stimulation. In silico docking studies showed that KI16 had favorable interactions with the STAT3 SH2 domain, however, no inhibitory activity could be observed in the STAT3 fluorescence polarization assay. KI16 inhibited cell viability preferentially in STAT3-dependent cell lines. Taken together, using a targeted, cell-based approach, novel inhibitors of STAT-driven transcriptional activity were discovered which are interesting leads to pursue further for the development of anti-cancer therapeutic agents.
Collapse
Affiliation(s)
- Iryna Kolosenko
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (IK); (DG)
| | - Yasmin Yu
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Sander Busker
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Matheus Dyczynski
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jianping Liu
- Karolinska High-Throughput Center, Department of Medical Biochemistry and Biophysics, Division of Functional Genomics, Karolinska Institutet Stockholm, Sweden
| | - Martin Haraldsson
- Chemical Biology Consortium Sweden, Department of Medical Biochemistry and Biophysics, Division of Translational Medicine and Chemical Biology, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Palm Apergi
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Helleday
- Department of Medical Biochemistry and Biophysics, Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Katja Pokrovskaja Tamm
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Brent D. G. Page
- Department of Medical Biochemistry and Biophysics, Division of Translational Medicine and Chemical Biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Dan Grander
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (IK); (DG)
| |
Collapse
|
46
|
Liu VWS, Yau WL, Tam CW, Yao KM, Shiu SYW. Melatonin Inhibits Androgen Receptor Splice Variant-7 (AR-V7)-Induced Nuclear Factor-Kappa B (NF-κB) Activation and NF-κB Activator-Induced AR-V7 Expression in Prostate Cancer Cells: Potential Implications for the Use of Melatonin in Castration-Resistant Prostate Cancer (CRPC) Therapy. Int J Mol Sci 2017; 18:E1130. [PMID: 28561752 PMCID: PMC5485954 DOI: 10.3390/ijms18061130] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/09/2017] [Accepted: 05/17/2017] [Indexed: 01/09/2023] Open
Abstract
A major current challenge in the treatment of advanced prostate cancer, which can be initially controlled by medical or surgical castration, is the development of effective, safe, and affordable therapies against progression of the disease to the stage of castration resistance. Here, we showed that in LNCaP and 22Rv1 prostate cancer cells transiently overexpressing androgen receptor splice variant-7 (AR-V7), nuclear factor-kappa B (NF-κB) was activated and could result in up-regulated interleukin (IL)-6 gene expression, indicating a positive interaction between AR-V7 expression and activated NF-κB/IL-6 signaling in castration-resistant prostate cancer (CRPC) pathogenesis. Importantly, both AR-V7-induced NF-κB activation and IL-6 gene transcription in LNCaP and 22Rv1 cells could be inhibited by melatonin. Furthermore, stimulation of AR-V7 mRNA expression in LNCaP cells by betulinic acid, a pharmacological NF-κB activator, was reduced by melatonin treatment. Our data support the presence of bi-directional positive interactions between AR-V7 expression and NF-κB activation in CRPC pathogenesis. Of note, melatonin, by inhibiting NF-κB activation via the previously-reported MT₁ receptor-mediated antiproliferative pathway, can disrupt these bi-directional positive interactions between AR-V7 and NF-κB and thereby delay the development of castration resistance in advanced prostate cancer. Apparently, this therapeutic potential of melatonin in advanced prostate cancer/CRPC management is worth translation in the clinic via combined androgen depletion and melatonin repletion.
Collapse
Affiliation(s)
| | - Wing Lung Yau
- Division of Nursing and Health Studies, School of Science and Technology, Open University of Hong Kong, Hong Kong, China.
| | - Chun Wai Tam
- Division of Nursing and Health Studies, School of Science and Technology, Open University of Hong Kong, Hong Kong, China.
| | - Kwok-Ming Yao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.
| | | |
Collapse
|
47
|
Wang LJ, Chang YC, Sun R, Li L. A multichannel smartphone optical biosensor for high-throughput point-of-care diagnostics. Biosens Bioelectron 2017; 87:686-692. [DOI: 10.1016/j.bios.2016.09.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 02/07/2023]
|
48
|
Divella R, De Luca R, Abbate I, Naglieri E, Daniele A. Obesity and cancer: the role of adipose tissue and adipo-cytokines-induced chronic inflammation. J Cancer 2016; 7:2346-2359. [PMID: 27994674 PMCID: PMC5166547 DOI: 10.7150/jca.16884] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue in addition to its ability to keep lipids is now recognized as a real organ with both metabolic and endocrine functions. Recent studies demonstrated that in obese animals is established a status of adipocyte hypoxia and in this hypoxic state interaction between adipocytes and stromal vascular cells contribute to tumor development and progression. In several tumors such as breast, colon, liver and prostate, obesity represents a poor predictor of clinical outcomes. Dysfunctional adipose tissue in obesity releases a disturbed profile of adipokines with elevated levels of pro-inflammatory factors and a consequent alteration of key signaling mediators which may be an active local player in establishing the peritumoral environment promoting tumor growth and progression. Therefore, adipose tissue hypoxia might contribute to cancer risk in the obese population. To date the precise mechanisms behind this obesity-cancer link is not yet fully understood. In the light of information provided in this review that aims to identify the key mechanisms underlying the link between obesity and cancer we support that inflammatory state specific of obesity may be important in obesity-cancer link.
Collapse
Affiliation(s)
- Rosa Divella
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Raffaele De Luca
- Department of Surgery Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Ines Abbate
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Emanuele Naglieri
- Department of Medical Oncology, Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Antonella Daniele
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| |
Collapse
|
49
|
Rudnicka C, Mochizuki S, Okada Y, McLaughlin C, Leedman PJ, Stuart L, Epis M, Hoyne G, Boulos S, Johnson L, Schlaich M, Matthews V. Overexpression and knock-down studies highlight that a disintegrin and metalloproteinase 28 controls proliferation and migration in human prostate cancer. Medicine (Baltimore) 2016; 95:e5085. [PMID: 27749584 PMCID: PMC5059087 DOI: 10.1097/md.0000000000005085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is one of the most prevalent cancers in men. It is critical to identify and characterize oncogenes that drive the pathogenesis of human prostate cancer. The current study builds upon previous research showing that a disintegrin and metallproteinase (ADAM)28 is involved in the pathogenesis of numerous cancers. Our novel study used overexpression, pharmacological, and molecular approaches to investigate the biological function of ADAM28 in human prostate cancer cells, with a focus on cell proliferation and migration. The results of this study provide important insights into the role of metalloproteinases in human prostate cancer.The expression of ADAM28 protein levels was assessed within human prostate tumors and normal adjacent tissue by immunohistochemistry. Immunocytochemistry and western blotting were used to assess ADAM28 protein expression in human prostate cancer cell lines. Functional assays were conducted to assess proliferation and migration in human prostate cancer cells in which ADAM28 protein expression or activity had been altered by overexpression, pharmacological inhibition, or by siRNA gene knockdown.The membrane bound ADAM28 was increased in human tumor biopsies and prostate cancer cell lines. Pharmacological inhibition of ADAM28 activity and/or knockdown of ADAM28 significantly reduced proliferation and migration of human prostate cancer cells, while overexpression of ADAM28 significantly increased proliferation and migration.ADAM28 is overexpressed in primary human prostate tumor biopsies, and it promotes human prostate cancer cell proliferation and migration. This study supports the notion that inhibition of ADAM28 may be a potential novel therapeutic strategy for human prostate cancer.
Collapse
Affiliation(s)
| | - Satsuki Mochizuki
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | | | - Peter J. Leedman
- Centre for Medical Research, The University of Western Australia, Perth
- Harry Perkins Institute of Medical Research, Nedlands
- School of Medicine and Pharmacology – Royal Perth Hospital Unit, The University of Western Australia
| | - Lisa Stuart
- Centre for Medical Research, The University of Western Australia, Perth
- Harry Perkins Institute of Medical Research, Nedlands
| | - Michael Epis
- Centre for Medical Research, The University of Western Australia, Perth
- Harry Perkins Institute of Medical Research, Nedlands
| | - Gerard Hoyne
- The University of Notre Dame Australia, Fremantle Campus
| | - Sherif Boulos
- Western Australian Neuromuscular Research Institute, Perth, Western Australia
| | - Liam Johnson
- School of Medicine and Pharmacology – Royal Perth Hospital Unit, The University of Western Australia
| | - Markus Schlaich
- School of Medicine and Pharmacology – Royal Perth Hospital Unit, The University of Western Australia
| | - Vance Matthews
- Harry Perkins Institute of Medical Research, Nedlands
- School of Medicine and Pharmacology – Royal Perth Hospital Unit, The University of Western Australia
- Correspondence: Vance Matthews, School of Medicine and Pharmacology – Royal Perth Hospital Unit, Level 3, Medical Research Foundation Building, Rear 50 Murray Street, Perth 6000, Western Australia (e-mail: )
| |
Collapse
|
50
|
Wang S, Clarke PAG, Davis R, Mumuni S, Kwabi-Addo B. Sex steroid-induced DNA methylation changes and inflammation response in prostate cancer. Cytokine 2016; 86:110-118. [PMID: 27500645 DOI: 10.1016/j.cyto.2016.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/08/2016] [Accepted: 07/09/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Sex steroid hormones have been reported to induce inflammation causing dysregulation of cytokines in prostate cancer cells. However, the underlying epigenetic mechanism has not well been studied. The objective of this study was to evaluate the effect of sex steroid hormones on epigenetic DNA methylation changes in prostate cancer cells using a signature PCR methylation array panel that correspond to 96 genes with biological function in the human inflammatory and autoimmune signals in prostate cancer. Of the 96-gene panel, 32 genes showed at least 10% differentially methylation level in response to hormonal treatment when compared to untreated cells. Genes that were hypomethylated included CXCL12, CXCL5, CCL25, IL1F8, IL13RAI, STAT5A, CXCR4 and TLR5; and genes that were hypermethylated included ELA2, TOLLIP, LAG3, CD276 and MALT1. Quantitative RT-PCR analysis of select genes represented in a cytokine expression array panel showed inverse association between DNA methylation and gene expression for TOLLIP, CXCL5, CCL18 and IL5 genes and treatment of prostate cancer cells with 5'-aza-2'-deoxycytidine with or without trichostatin A induced up-regulation of TOLLIP expression. Further analysis of relative gene expression of matched prostate cancer tissues when compared to benign tissues from individual patients with prostate cancer showed increased and significant expression for CCL18 (2.6-fold; p<0.001), a modest yet significant increase in IL5 expression (1.17-fold; p=0.015), and a modest increase in CXCL5 expression (1.4-fold; p=0.25). In conclusion, our studies demonstrate that sex steroid hormones can induce aberrant gene expression via differential methylation changes in prostate carcinogenesis.
Collapse
Affiliation(s)
- Songping Wang
- Department of Biochemistry and Molecular Biology, Howard University, 2041 Georgia Ave N.W., Washington, DC 20060, USA
| | - Pamela A G Clarke
- Department of Biochemistry and Molecular Biology, Howard University, 2041 Georgia Ave N.W., Washington, DC 20060, USA
| | - Roderick Davis
- Department of Biochemistry and Molecular Biology, Howard University, 2041 Georgia Ave N.W., Washington, DC 20060, USA
| | - Salma Mumuni
- Department of Biochemistry and Molecular Biology, Howard University, 2041 Georgia Ave N.W., Washington, DC 20060, USA
| | - Bernard Kwabi-Addo
- Department of Biochemistry and Molecular Biology, Howard University, 2041 Georgia Ave N.W., Washington, DC 20060, USA.
| |
Collapse
|