1
|
Hirose Y, Oba A, Takamatsu M, Hamada T, Takeda T, Suzuki T, Maekawa A, Kitano Y, Sato S, Kobayashi K, Omiya K, Ono Y, Sato T, Ito H, Sasaki T, Ozaka M, Takeuchi K, Sasahira N, Inoue Y, Wakai T, Takahashi Y. Caveolin-1 expression is a predictor of survival and recurrence patterns in resected pancreatic ductal adenocarcinoma. Pancreatology 2024; 24:1021-1030. [PMID: 39395872 DOI: 10.1016/j.pan.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND/OBJECTIVE Caveolin-1 (Cav1) expressed in cancer cells (cCav1) or cancer-associated fibroblasts (fCav1) exerts either pro- or anti-tumorigenic effects depending on the cancer type or stage of cancer. We aimed to clarify the impact of cCav1 or fCav1 on survival, recurrence patterns, and efficacy of neoadjuvant chemotherapy (NAC) in resected pancreatic ductal adenocarcinoma (PDAC). METHODS Tissue microarrays were constructed including 615 patients who underwent curative resection for PDAC. Cav1 expression was evaluated by immunohistochemistry. Patients were divided into two groups based on Cav1 expression in cancer cells (cCav1high vs. cCav1low) or cancer-associated fibroblasts (fCav1high vs. fCav1low). RESULTS Among all 615 patients, 40.7% were cCav1high and 72.7% were fCav1high. cCav1high was associated with worse overall survival (OS) (p = 0.001) and recurrence-free survival (RFS) (p = 0.001) than cCav1low, and was an independent prognostic factor in multivariate analysis of OS and RFS (OS: p = 0.001, hazard ratio [HR] 1.361; RFS: p = 0.001, HR 1.348). Among 596 patients with resectable/borderline resectable PDAC, cCav1high patients with NAC showed better OS than those without, while there was no significant difference between cCav1low patients with NAC and those without. cCav1high was associated with early recurrence (< 6 months) and liver metastasis after resection. Multivariate analysis revealed cCav1high as an independent predictor of liver metastasis. CONCLUSIONS cCav1high correlated with worse survival, early recurrence, and liver metastasis after resection for PDAC, while NAC improved survival in cCav1high patients. The Evaluation of cCav1 status could provide additional information contributing to the personalized management of PDAC.
Collapse
Affiliation(s)
- Yuki Hirose
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tsuyoshi Hamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tatsunori Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Maekawa
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuki Kitano
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shoki Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kosuke Kobayashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kojiro Omiya
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshihiro Ono
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takafumi Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiromichi Ito
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kengo Takeuchi
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan; Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
2
|
Patritti-Cram J, Rahrmann EP, Rizvi TA, Scheffer KC, Phoenix TN, Largaespada DA, Ratner N. NF1-dependent disruption of the blood-nerve-barrier is improved by blockade of P2RY14. iScience 2024; 27:110294. [PMID: 39100928 PMCID: PMC11294707 DOI: 10.1016/j.isci.2024.110294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/17/2023] [Accepted: 06/14/2024] [Indexed: 08/06/2024] Open
Abstract
The blood-nerve-barrier (BNB) that regulates peripheral nerve homeostasis is formed by endoneurial capillaries and perineurial cells surrounding the Schwann cell (SC)-rich endoneurium. Barrier dysfunction is common in human tumorigenesis, including in some nerve tumors. We identify barrier disruption in human NF1 deficient neurofibromas, which were characterized by reduced perineurial cell glucose transporter 1 (GLUT1) expression and increased endoneurial fibrin(ogen) deposition. Conditional Nf1 loss in murine SCs recapitulated these alterations and revealed decreased tight junctions and decreased caveolin-1 (Cav1) expression in mutant nerves and in tumors, implicating reduced Cav1-mediated transcytosis in barrier disruption and tumorigenesis. Additionally, elevated receptor tyrosine kinase activity and genetic deletion of Cav1 increased endoneurial fibrin(ogen), and promoted SC tumor formation. Finally, when SC lacked Nf1, genetic loss or pharmacological inhibition of P2RY14 rescued Cav1 expression and barrier function. Thus, loss of Nf1 in SC causes dysfunction of the BNB via P2RY14-mediated G-protein coupled receptor (GPCR) signaling.
Collapse
Affiliation(s)
- Jennifer Patritti-Cram
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0713, USA
| | - Eric P. Rahrmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Tilat A. Rizvi
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Katherine C. Scheffer
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Timothy N. Phoenix
- Division of Pharmaceutical Sciences, James L. Wrinkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - David A. Largaespada
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
3
|
MOSTAFAVI SAMANEH, HASSAN ZUHAIRMOHAMMAD. The anti-neoplastic effects of metformin modulate the acquired phenotype of fibroblast cells in the breast cancer-normal fibroblast co-culture system. Oncol Res 2024; 32:477-487. [PMID: 38361760 PMCID: PMC10865743 DOI: 10.32604/or.2023.043926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 11/06/2023] [Indexed: 02/17/2024] Open
Abstract
Intracellular communications between breast cancer and fibroblast cells were reported to be involved in cancer proliferation, growth, and therapy resistance. The hallmarks of cancer-fibroblast interactions, consisting of caveolin 1 (Cav1) and mono-carboxylate transporter 4 (MCT4) (metabolic coupling markers), along with IL-6, TGFβ, and lactate secretion, are considered robust biomarkers predicting recurrence and metastasis. In order to promote a novel phenotype in normal fibroblasts, we predicted that breast cancer cells could be able to cause loss of Cav1 and increase of MCT4, as well as elevate IL-6 and TGFβ in nearby normal fibroblasts. We created a co-culture model using breast cancer (4T1) and normal fibroblast (NIH3T3) cell lines cultured under specific experimental conditions in order to directly test our theory. Moreover, we show that long-term co-culture of breast cancer cells and normal fibroblasts promotes loss of Cav1 and gain of MCT4 in adjacent fibroblasts and increase lactate secretion. These results were validated using the monoculture of each group separately as a control. In this system, we show that metformin inhibits IL-6 and TGFβ secretion and re-expresses Cav1 in both cells. However, MCT4 and lactate stayed high after treatment with metformin. In conclusion, our work shows that co-culture with breast cancer cells may cause significant alterations in the phenotype and secretion of normal fibroblasts. Metformin, however, may change this state and affect fibroblasts' acquired phenotypes. Moreover, mitochondrial inhibition by metformin after 8 days of treatment, significantly hinders tumor growth in mouse model of breast cancer.
Collapse
Affiliation(s)
- SAMANEH MOSTAFAVI
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - ZUHAIR MOHAMMAD HASSAN
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
4
|
Li B, Ming H, Qin S, Zhou L, Huang Z, Jin P, Peng L, Luo M, Zhang T, Wang K, Liu R, Liou Y, Nice EC, Jiang J, Huang C. HSPA8 Activates Wnt/β-Catenin Signaling to Facilitate BRAF V600E Colorectal Cancer Progression by CMA-Mediated CAV1 Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306535. [PMID: 37973552 PMCID: PMC10797426 DOI: 10.1002/advs.202306535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Indexed: 11/19/2023]
Abstract
BRAF V600E attracts wide attention in the treatment of colorectal cancer (CRC) as stratifying and predicting a refractory classification of CRC. Recent evidence indicates that Wnt/β-catenin signaling is broadly activated and participates in the refractoriness of BRAF V600E CRC, but the underlying molecular mechanism needs to be elucidated. Here, heat shock 70 kDa protein 8 (HSPA8), an essential regulator in chaperone-mediated autophagy (CMA), is identified as a potential therapeutic target for advanced BRAF V600E CRC. These results show that HSPA8 is transcriptionally upregulated in BRAF V600E CRC, which promotes CMA-dependent degradation of caveolin-1 (CAV1) to release β-catenin into the nucleus and thus activates the Wnt/β-catenin pathway, contributing to metastasis and progression of BRAF V600E CRC. Of note, HSPA8 directly interacts with the KIFSN motif on CAV1, the interaction can be enhanced by p38 MAPK-mediated CAV1 S168 phosphorylation. Furthermore, pharmacological targeting HSPA8 by VER155008 exhibits synergistic effects with BRAF inhibitors on CRC mouse models. In summary, these findings discover the important role of the HSPA8/CAV1/β-catenin axis in the development of refractory BRAF V600E CRC and highlight HSPA8 as a predictive biomarker and therapeutic target in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Ping Jin
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Liyuan Peng
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Maochao Luo
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Tingting Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Kui Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| | - Rui Liu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuan610041P. R. China
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingapore117543Singapore
- Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingapore117573Singapore
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVIC3800Australia
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth HospitalSichuan UniversityChengdu610041P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital and West China School of Basic Medical Sciences and Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengdu610041P. R. China
| |
Collapse
|
5
|
Kamposioras K, Dinas PC, Barriuoso J, Trachana V, Dimas K. Caveolin-1 protein expression as a prognostic biomarker of gastrointestinal tumours: A systematic review and meta-analysis. Eur J Clin Invest 2023; 53:e14065. [PMID: 37497737 DOI: 10.1111/eci.14065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Gastrointestinal (GI) cancers remain a major threat worldwide, accounting for over 30% of cancer deaths. The identification of novel prognostic biomarkers remains a challenge despite significant advances in the field. The CAV1 gene, encoding the caveolin-1 protein, remains enigmatic in cancer and carcinogenesis, as it has been proposed to act as both a tumour promoter and a tumour suppressor. METHODS To analyse the differential role of caveolin-1 expression in both tumour cells and stroma in relation to prognosis in GI tumours, we performed a systematic review and meta-analysis according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines; PROSPERO registration number: CRD42022299148. RESULTS Our analysis showed that high levels of caveolin-1 in tumour cells were associated with poor prognosis and inferior overall survival (OS) in oesophageal and pancreatic cancer and hepatocellular carcinoma (HCC), but not in gastric and colorectal cancer. Importantly, our study showed that higher stromal caveolin-1 expression was associated with significantly longer OS and disease-free survival in colorectal cancer. Analysis of stromal caveolin-1 expression in the remaining tumours showed a similar trend, although it did not reach statistical significance. CONCLUSIONS The data suggest that caveolin-1 expression in the tumour cells of oesophageal, pancreatic cancer and HCC and in the stroma of colorectal cancer may be an important novel predictive biomarker for the clinical management of these diseases in a curative setting. However, the main conclusion of our analysis is that caveolin-1 expression should always be assessed separately in stroma and tumour cells.
Collapse
Affiliation(s)
| | - Petros C Dinas
- FAME Laboratory, Department of Physical Education and Sport Science, University of Thessaly, Volos, Greece
| | - Jorge Barriuoso
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Volos, Greece
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Volos, Greece
| |
Collapse
|
6
|
Bhowmick S, Biswas T, Ahmed M, Roy D, Mondal S. Caveolin-1 and lipids: Association and their dualism in oncogenic regulation. Biochim Biophys Acta Rev Cancer 2023; 1878:189002. [PMID: 37848094 DOI: 10.1016/j.bbcan.2023.189002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023]
Abstract
Caveolin-1 (Cav-1) is a structural protein of caveolae that functions as a molecular organizer for different cellular functions including endocytosis and cellular signaling. Cancer cells take advantage of the physical position of Cav-1, as it can communicate with extracellular matrix, help to organize growth factor receptors, redistribute cholesterol and glycosphingolipids, and finally transduce signals within the cells for oncogenesis. Recent studies emphasize the exceeding involvement of Cav-1 with different lipid bodies and in altering the metabolism, especially lipid metabolism. However, the association of Cav-1 with different lipid bodies like lipid rafts, lipid droplets, cholesterols, sphingolipids, and fatty acids is remarkably dynamic. The lipid-Cav-1 alliance plays a dual role in carcinogenesis. Both cancer progression and regression are modified and affected by the type of lipid molecule's association with Cav-1. Accordingly, this Cav-1-lipid cooperation exemplifies a cancer-type-specific treatment strategy for a better prognosis of the disease. In this review, we first present Cav-1 as an oncogenic molecule and its communication via lipid raft. We discussed the involvement of Cav-1 with lipid droplets, Cholesterol, sphingolipids, gangliosides, and ceramides. Further, we describe the Cav-1-mediated altered Fatty acid metabolism in cancer and the strategic therapeutic approaches toward Cav-1 targeting.
Collapse
Affiliation(s)
- Sramana Bhowmick
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Tannishtha Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Mehnaz Ahmed
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, USA
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
7
|
Bian Q, Li B, Zhang L, Sun Y, Zhao Z, Ding Y, Yu H. Molecular pathogenesis, mechanism and therapy of Cav1 in prostate cancer. Discov Oncol 2023; 14:196. [PMID: 37910338 PMCID: PMC10620365 DOI: 10.1007/s12672-023-00813-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
Prostate cancer is the second incidence of malignant tumors in men worldwide. Its incidence and mortality are increasing year by year. Enhanced expression of Cav1 in prostate cancer has been linked to both proliferation and metastasis of cancer cells, influencing disease progression. Dysregulation of the Cav1 gene shows a notable association with prostate cancer. Nevertheless, there is no systematic review to report about molecular signal mechanism of Cav1 and drug treatment in prostate cancer. This article reviews the structure, physiological and pathological functions of Cav1, the pathogenic signaling pathways involved in prostate cancer, and the current drug treatment of prostate cancer. Cav1 mainly affects the occurrence of prostate cancer through AKT/mTOR, H-RAS/PLCε, CD147/MMPs and other pathways, as well as substance metabolism including lipid metabolism and aerobic glycolysis. Baicalein, simvastatin, triptolide and other drugs can effectively inhibit the growth of prostate cancer. As a biomarker of prostate cancer, Cav1 may provide a potential therapeutic target for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Qiang Bian
- Department of Pathophysiology, Weifang Medicine University, Weifang, 261053, Shandong, People's Republic of China
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, People's Republic of China
| | - Bei Li
- Department of Radiological Image, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, People's Republic of China
| | - Luting Zhang
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China
| | - Yinuo Sun
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China
| | - Zhankui Zhao
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, People's Republic of China
| | - Yi Ding
- Department of Pathophysiology, Weifang Medicine University, Weifang, 261053, Shandong, People's Republic of China.
| | - Honglian Yu
- Department of Biochemistry, Jining Medical University, Jining, 272067, Shandong, People's Republic of China.
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272100, Shandong, People's Republic of China.
| |
Collapse
|
8
|
Urbanek KA, Kowalska K, Habrowska-Górczyńska DE, Kozieł MJ, Domińska K, Piastowska-Ciesielska AW. Revealing the Role of Alternariol in the Local Steroidogenesis in Human Prostate Normal and Cancer Cells. Int J Mol Sci 2023; 24:ijms24119513. [PMID: 37298472 DOI: 10.3390/ijms24119513] [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: 04/28/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
The mycotoxin alternariol (AOH) can be found in food products infected by Alternaria spp. and is considered an endocrine-disruptive mycotoxin. The main mechanism of AOH toxicity is associated with DNA damage and modulation of the inflammation process. Still, AOH is considered as one of the emerging mycotoxins. In this study, we have evaluated how AOH might affect the local steroidogenesis process in the prostate, in both normal and cancer cells. We have found that AOH itself modulates the cell cycle, inflammation, and apoptosis, rather than the steroidogenesis process in prostate cancer cells; however, in the presence of another steroidogenic agent, the influence on steroidogenesis is significant. Therefore, this is the first study to report the effect of AOH on local steroidogenesis in normal and prostate cancer cells. We postulate that AOH might modulate the release of the steroid hormones and expression of the key components by interfering with the steroidogenic pathway and might be considered a steroidogenesis-altering agent.
Collapse
Affiliation(s)
- Kinga Anna Urbanek
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, 90-752 Lodz, Poland
| | - Karolina Kowalska
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, 90-752 Lodz, Poland
| | | | - Marta Justyna Kozieł
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, 90-752 Lodz, Poland
- Medical University of Lodz, BRaIn Laboratories, 92-216 Lodz, Poland
| | - Kamila Domińska
- Medical University of Lodz, Department of Comparative Endocrinology, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Agnieszka Wanda Piastowska-Ciesielska
- Medical University of Lodz, Department of Cell Cultures and Genomic Analysis, 90-752 Lodz, Poland
- Medical University of Lodz, BRaIn Laboratories, 92-216 Lodz, Poland
| |
Collapse
|
9
|
Wang Y, Li Y, Zhong J, Li M, Zhou Y, Lin Q, Zong S, Luo W, Wang J, Wang K, Wang J, Xiong L. Tumor-derived Cav-1 promotes pre-metastatic niche formation and lung metastasis in breast cancer. Theranostics 2023; 13:1684-1697. [PMID: 37056561 PMCID: PMC10086203 DOI: 10.7150/thno.79250] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/04/2023] [Indexed: 04/15/2023] Open
Abstract
Rationale: Breast cancer (BC), as one of the most frequently diagnosed cancer, has a poor prognosis due to the development of distant metastasis. Among the BC metastatic sites, lung is one of the most common sites. Caveolin-1 (Cav-1) is a functional membrane protein that plays a vital role in tumor metastasis. Although studies have revealed that Cav-1 levels were elevated in patients with advanced cancer, whether Cav-1 affects BC lung metastasis by influencing the formation of pre-metastatic niche (PMN) through exosomes has not been explored. Methods: Differential ultracentrifugation, transmission electron microscopy and nanoparticle tracking analysis were used to verify the presence of exosomes. Transwell assays were used to examine the biological effects of exosomes containing Cav-1. Both in vitro cell cultures and mammary tumor cell-induced mouse models were used to assess the lung metastasis. The regulatory mechanisms of PMN formation were revealed using western blot, flow cytometry, RT-qPCR, immunofluorescence assays, gene overexpression assays and RNA interference assays. Results: Exosomes have critical functions in transporting Cav-1 between primary BC and metastatic organ microenvironments. Cav-1 in BC-derived exosomes can act as a signaling molecule to mediate intercellular communication and regulate the PMN before lung metastasis by regulating the expression of PMN marker genes and inflammatory chemokines in lung epithelial cells, promoting the secretion of tenascin-C (TnC) in lung fibroblasts to cause extracellular matrix (ECM) deposition, and inhibiting the PTEN/CCL2/VEGF-A signaling pathway in lung macrophages to facilitate their M2-type polarization and angiogenesis. Conclusion: Our study investigated the mechanisms of lung PMN formation induced by Cav-1 in BC-derived exosomes. Our data may provide new directions for exploring the mechanisms and developing treatment strategies of BC lung metastasis.
Collapse
Affiliation(s)
- Yi Wang
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
| | - Yuqiu Li
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
- Queen Mary college, Nanchang University, Nanchang 330006, China
| | - Junpei Zhong
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
| | - Miao Li
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Youjia Zhou
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
| | - Qing Lin
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
| | - Siwen Zong
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wenting Luo
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jiayang Wang
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jie Wang
- Key laboratory of functional and clinical translational medicine, Xiamen Medical College, Fujian province university, Xiamen 361023, China
| | - Lixia Xiong
- Department of Pathophysiology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, China
- Key laboratory of functional and clinical translational medicine, Xiamen Medical College, Fujian province university, Xiamen 361023, China
- ✉ Corresponding author: Lixia Xiong
| |
Collapse
|
10
|
Huang Q, Wu L, Wang Y, Kong X, Xiao X, Huang Q, Li M, Zhai Y, Shi F, Zhao R, Zhong J, Xiong L. Caveolin-1-deficient fibroblasts promote migration, invasion, and stemness via activating the TGF-β/Smad signaling pathway in breast cancer cells. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1587-1598. [PMID: 36604141 PMCID: PMC9827800 DOI: 10.3724/abbs.2022150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) represent one of the main components in the tumor stroma and play a key role in breast cancer progression. Transforming growth factor-β (TGF-β) has been established to mediate breast cancer metastasis by regulating the epithelial-mesenchymal transition (EMT) and stemness of cancer cells. Caveolin-1 (CAV-1) is a scaffold protein of caveolae that is related to the proliferation and metabolism of cancer cells. It is now well demonstrated that CAV-1 deficiency in the tumor stroma is positively correlated with distant metastasis, but the mechanism remains unclear. Here, we explore whether CAV-1-deficient fibroblasts play an essential role in the EMT and stemness of breast cancer cells (BCCs) through TGF-β signaling. We establish a specific small interfering RNA (siRNA) to inhibit CAV-1 expression in fibroblasts and coculture them with BCCs to investigate the effect of CAV‑1-deficient fibroblasts and the tumor microenvironment on breast cancer progression. This study refreshingly points out that CAV-1 deficiency in fibroblasts enhances TGF-β1 secretion and then activates the TGF-β1/Smad signaling pathway of BCCs, thus promoting the metastasis and stemness of BCCs. Collectively, our findings indicate an unexpected role of CAV-1 deficiency in fibroblasts and the tumor microenvironment as a permissive factor, which is regulated by the TGF-β1 signaling pathway in BCCs.
Collapse
Affiliation(s)
- Qingyun Huang
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Longyuan Wu
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Yi Wang
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Xinyu Kong
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Xinhua Xiao
- The First Affiliated HospitalNanchang UniversityNanchang330006China
| | - Qiyuan Huang
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Miao Li
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Yujia Zhai
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Fuxiu Shi
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Ruichen Zhao
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Junpei Zhong
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China
| | - Lixia Xiong
- Department of PathophysiologyMedical CollegeNanchang UniversityNanchang330006China,Key Laboratory of Functional and Clinical Translational MedicineFujian Province UniversityXiamen361023China,Correspondence address. Tel: +86-791-86360565;
| |
Collapse
|
11
|
Shi X, Yang J, Deng S, Xu H, Wu D, Zeng Q, Wang S, Hu T, Wu F, Zhou H. TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J Hematol Oncol 2022; 15:135. [PMID: 36115986 PMCID: PMC9482317 DOI: 10.1186/s13045-022-01349-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/24/2022] [Indexed: 12/30/2022] Open
Abstract
AbstractTransforming growth factor-β (TGF-β) signaling has a paradoxical role in cancer progression, and it acts as a tumor suppressor in the early stages but a tumor promoter in the late stages of cancer. Once cancer cells are generated, TGF-β signaling is responsible for the orchestration of the immunosuppressive tumor microenvironment (TME) and supports cancer growth, invasion, metastasis, recurrence, and therapy resistance. These progressive behaviors are driven by an “engine” of the metabolic reprogramming in cancer. Recent studies have revealed that TGF-β signaling regulates cancer metabolic reprogramming and is a metabolic driver in the tumor metabolic microenvironment (TMME). Intriguingly, TGF-β ligands act as an “endocrine” cytokine and influence host metabolism. Therefore, having insight into the role of TGF-β signaling in the TMME is instrumental for acknowledging its wide range of effects and designing new cancer treatment strategies. Herein, we try to illustrate the concise definition of TMME based on the published literature. Then, we review the metabolic reprogramming in the TMME and elaborate on the contribution of TGF-β to metabolic rewiring at the cellular (intracellular), tissular (intercellular), and organismal (cancer-host) levels. Furthermore, we propose three potential applications of targeting TGF-β-dependent mechanism reprogramming, paving the way for TGF-β-related antitumor therapy from the perspective of metabolism.
Collapse
|
12
|
Tang Q, Wang S, Di Z, Li H, Xu K, Hu X, Di M. Identification and validation of a prognostic risk model based on caveolin family genes for breast cancer. Front Cell Dev Biol 2022; 10:822187. [PMID: 36147736 PMCID: PMC9485841 DOI: 10.3389/fcell.2022.822187] [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: 11/25/2021] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Breast cancer (BC) is the most vicious killer of women’s health and is accompanied by increased incidence and mortality rates worldwide. Many studies have demonstrated that caveolins (CAVs) were abnormally expressed in a variety of tumors and implicated in tumorigenesis and cancer progression. However, the role of CAVs in BC remains somewhat contentious.Methods: We comprehensively explored the expression and prognostic value of CAVs (CAV1-3) in BC utilizing public databases (ONCOMINE, TIMER, UALCAN, and TCGA databases). Then we constructed a prognostic model based on the expression profiles. Also, a prognostic nomogram was built to predict the overall survival (OS). We further investigated the relationship between this signature and immune cell infiltration and the mutational landscape in BC. The R package “pRRophetic” was used to predict chemotherapeutic response in BC patients. Finally, we employed loss-of-function approaches to validate the role of CAVs in BC.Results: We found that CAVs were significantly downregulated in various cancer types, especially in BC. Low CAV expression was closely related to the malignant clinicopathological characteristics and worse OS and relapse-free survival (RFS) in BC. Then we constructed a prognostic model based on the expression profiles of CAVs, which divided BC patients into two risk groups. The Kaplan–Meier analysis showed that patients in the high-risk group tend to have a poorer prognosis than those in the low-risk group. Multivariate analysis indicated that the risk score and stage were both independent prognostic factors for BC patients, suggesting a complementary value. The clinical profiles and risk module were used to construct a nomogram that could accurately predict the OS in BC. In addition, we found that patients in the low-risk group tend to have a relatively high immune status and a lower mutation event frequency compared to the high-risk group. Furthermore, this signature could predict the response to chemotherapy and immunotherapy. Finally, CAV depletion promoted the colony formation, migration, and invasion of BC cells.Conclusion: CAVs may serve as novel biomarkers and independent prognostic factors for BC patients. Also, the constructed signature based on CAVs may predict immunotherapeutic responses and provide a novel nomogram for precise outcome prediction of BC.
Collapse
Affiliation(s)
- Qiang Tang
- Department of General Surgery, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Shurui Wang
- School of Nursing Peking Union Medical College, Beijing, China
| | - Ziyang Di
- Department of General Surgery, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Huimin Li
- Department of General Surgery, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Kailiang Xu
- Department of Urology, Jingzhou Central Hospital, the Second Clinical Medical College, Yangtze University, Jingzhou, China
- *Correspondence: Maojun Di, ; Xin Hu, ; Kailiang Xu,
| | - Xin Hu
- Department of General Surgery, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Maojun Di, ; Xin Hu, ; Kailiang Xu,
| | - Maojun Di
- Department of General Surgery, Shiyan Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Maojun Di, ; Xin Hu, ; Kailiang Xu,
| |
Collapse
|
13
|
Ren L, Zhou P, Wu H, Liang Y, Xu R, Lu H, Chen Q. Caveolin-1 is a prognostic marker and suppresses the proliferation of breast cancer. Transl Cancer Res 2022; 10:3797-3810. [PMID: 35116679 PMCID: PMC8798413 DOI: 10.21037/tcr-21-1139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022]
Abstract
Background To explore the role of caveolin-1 (Cav-1) in breast cancer (BC). Methods Cav-1 expression data were downloaded from the Tumor Immune Estimation Resource (TIMER) and Gene Expression Omnibus (GEO) databases. We compared the expression of Cav-1 in different tumor tissues and between BC tissues and normal tissues (NTs), as well as the differences between different clinical traits. Kaplan-Meier survival analysis and univariate and multivariate Cox regression analyses were used to determine whether Cav-1 serves as a prognostic factor. The correlations of Cav-1 expression with the immune microenvironment and infiltrating immune cells were also analyzed. Quantitative polymerase chain reaction (qPCR) was used to detect Cav-1 mRNA expression in the MCF-7, SKB-R3, MDB-MB-231, and SUM-159 cell lines. LV-Cav-1-RNAi was transfected into MCF-7 and MDB-MB-231 cells, and the MTT assay was used to detect cell proliferation. Subsequently, MDB-MB-231 cells carrying the Cav-1-RNAi gene were used to determine the effects of Cav-1 knockdown on tumor growth in vivo using a severe combined immunodeficiency (SCID) model. Results Cav-1 was enriched in most solid tumors, and its expression was lower in BC tissues than in NT. Cav-1 expression was shown to be related to patients’ clinical outcomes. Cav-1 was expressed in the MCF-7, SKB-R3, MDB-MB-231, and SUM-159 cell lines. The MTT assay revealed that the proliferative ability of MDB-MB-231 and MCF-7 cells was accelerated. The tumor volume of SCID mice administered with LV-Cav-1-RNAi cells was increased. Conclusions These results suggest that Cav-1 may serve as a suppressor in the development of BC.
Collapse
Affiliation(s)
- Liping Ren
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peijuan Zhou
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huajia Wu
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Liang
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Xu
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hai Lu
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianjun Chen
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
14
|
Low JY, Laiho M. Caveolae-Associated Molecules, Tumor Stroma, and Cancer Drug Resistance: Current Findings and Future Perspectives. Cancers (Basel) 2022; 14:cancers14030589. [PMID: 35158857 PMCID: PMC8833326 DOI: 10.3390/cancers14030589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell membranes contain small invaginations called caveolae. They are a specialized lipid domain and orchestrate cellular signaling events, mechanoprotection, and lipid homeostasis. Formation of the caveolae depends on two classes of proteins, the caveolins and cavins, which form large complexes that allow their self-assembly into caveolae. Loss of either of these two proteins leads to distortion of the caveolae structure and disruption of many physiological processes that affect diseases of the muscle, metabolic states governing lipids, and the glucose balance as well as cancers. In cancers, the expression of caveolins and cavins is heterogenous, and they undergo alterations both in the tumors and the surrounding tumor microenvironment stromal cells. Remarkably, their expression and function has been associated with resistance to many cancer drugs. Here, we summarize the current knowledge of the resistance mechanisms and how this knowledge could be applied into the clinic in future. Abstract The discovery of small, “cave-like” invaginations at the plasma membrane, called caveola, has opened up a new and exciting research area in health and diseases revolving around this cellular ultrastructure. Caveolae are rich in cholesterol and orchestrate cellular signaling events. Within caveola, the caveola-associated proteins, caveolins and cavins, are critical components for the formation of these lipid rafts, their dynamics, and cellular pathophysiology. Their alterations underlie human diseases such as lipodystrophy, muscular dystrophy, cardiovascular disease, and diabetes. The expression of caveolins and cavins is modulated in tumors and in tumor stroma, and their alterations are connected with cancer progression and treatment resistance. To date, although substantial breakthroughs in cancer drug development have been made, drug resistance remains a problem leading to treatment failures and challenging translation and bench-to-bedside research. Here, we summarize the current progress in understanding cancer drug resistance in the context of caveola-associated molecules and tumor stroma and discuss how we can potentially design therapeutic avenues to target these molecules in order to overcome treatment resistance.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Correspondence: ; Tel.: +1-410-502-9748; Fax: +1-410-502-2821
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
15
|
Furuya K, Fujibayashi S, Wu T, Takahashi K, Takase S, Orimoto A, Sugano E, Tomita H, Kashiwagi S, Kiyono T, Ishii T, Fukuda T. Transcriptome analysis to identify the downstream genes of androgen receptor in dermal papilla cells. BMC Genom Data 2022; 23:2. [PMID: 34983378 PMCID: PMC8725446 DOI: 10.1186/s12863-021-01018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/14/2021] [Indexed: 11/10/2022] Open
Abstract
Background Testosterone signaling mediates various diseases, such as androgenetic alopecia and prostate cancer. Testosterone signaling is mediated by the androgen receptor (AR). In this study, we fortuitously found that primary and immortalized dermal papilla cells suppressed AR expression, although dermal papilla cells express AR in vivo. To analyze the AR signaling pathway, we exogenously introduced the AR gene via a retrovirus into immortalized dermal papilla cells and comprehensively compared their expression profiles with and without AR expression. Results Whole-transcriptome profiling revealed that the focal adhesion pathway was mainly affected by the activation of AR signaling. In particular, we found that caveolin-1 gene expression was downregulated in AR-expressing cells, suggesting that caveolin-1 is controlled by AR. Conclusion Our whole transcriptome data is critical resources for discovery of new therapeutic targets for testosterone-related diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12863-021-01018-6. The comprehensive gene expression profiling were obtained by RNA-Seq analysis about AR negative and AR positive dermal papilla cells. The bioinformatics analysis suggested that caveolin-1 and EGF receptors are the downstream of AR signaling. Our study showed the combination of pinpoint mutant cells and global transcriptome is effective to identify the downstream genes.
Collapse
Affiliation(s)
- Kai Furuya
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - So Fujibayashi
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Tao Wu
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Kouhei Takahashi
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Shin Takase
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Ai Orimoto
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Eriko Sugano
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Hiroshi Tomita
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan
| | - Sayo Kashiwagi
- Rohto Pharmaceutical Co., Ltd., Basic Research Development Division, 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Tohru Kiyono
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Tsuyoshi Ishii
- Rohto Pharmaceutical Co., Ltd., Basic Research Development Division, 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan.
| | - Tomokazu Fukuda
- Graduate School of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| |
Collapse
|
16
|
Goswami S, Sarkar C, Singh S, Singh AP, Chakroborty D. Racial differences in prostate tumor microenvironment: implications for disparate clinical outcomes and potential opportunities. CANCER HEALTH DISPARITIES 2022; 6:214. [PMID: 36777283 PMCID: PMC9910060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Disparities in cancer incidence and outcome are common among the racial and ethnical minorities in the United States and are of significant social and clinical concern. Prostate cancer is the most commonly diagnosed non-cutaneous malignancy in American men and exhibits substantial racial disparities with African American men bearing the highest burden in terms of incidence and mortality. A multitude of factors, including socioeconomic, behavioral, and access to healthcare, have been implicated as the underlying causes of such disparities. More recent data also suggest that there are inherent molecular and biological differences in prostate tumors of patients having distinct racial backgrounds. Tumor microenvironment has tremendous impact on the course of cancer progression and clinical outcome and may also contribute to the racial disparities observed in prostate cancer. Therefore, a better understanding of critical differences in the tumor microenvironment components may provide newer directions to study the biological causes of prostate cancer health disparities and may identify novel therapeutic targets. This review discusses the findings related to the tumor microenvironment differences between African American and Caucasian American prostate cancer patients and makes suggestion regarding their potential significance in prostate cancer disparities.
Collapse
Affiliation(s)
- Sandeep Goswami
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Chandrani Sarkar
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Ajay Pratap Singh
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Debanjan Chakroborty
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA.,Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
17
|
Leach DA, Fernandes RC, Bevan CL. Cellular specificity of androgen receptor, coregulators, and pioneer factors in prostate cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2022; 2:R112-R131. [PMID: 37435460 PMCID: PMC10259329 DOI: 10.1530/eo-22-0065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 07/13/2023]
Abstract
Androgen signalling, through the transcription factor androgen receptor (AR), is vital to all stages of prostate development and most prostate cancer progression. AR signalling controls differentiation, morphogenesis, and function of the prostate. It also drives proliferation and survival in prostate cancer cells as the tumour progresses; given this importance, it is the main therapeutic target for disseminated disease. AR is also essential in the surrounding stroma, for the embryonic development of the prostate and controlling epithelial glandular development. Stromal AR is also important in cancer initiation, regulating paracrine factors that excite cancer cell proliferation, but lower stromal AR expression correlates with shorter time to progression/worse outcomes. The profile of AR target genes is different between benign and cancerous epithelial cells, between castrate-resistant prostate cancer cells and treatment-naïve cancer cells, between metastatic and primary cancer cells, and between epithelial cells and fibroblasts. This is also true of AR DNA-binding profiles. Potentially regulating the cellular specificity of AR binding and action are pioneer factors and coregulators, which control and influence the ability of AR to bind to chromatin and regulate gene expression. The expression of these factors differs between benign and cancerous cells, as well as throughout disease progression. The expression profile is also different between fibroblast and mesenchymal cell types. The functional importance of coregulators and pioneer factors in androgen signalling makes them attractive therapeutic targets, but given the contextual expression of these factors, it is essential to understand their roles in different cancerous and cell-lineage states.
Collapse
Affiliation(s)
- Damien A Leach
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Rayzel C Fernandes
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Charlotte L Bevan
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
18
|
Rahimi HR, Mojarrad M, Moghbeli M. MicroRNA-96: A therapeutic and diagnostic tumor marker. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:3-13. [PMID: 35656454 DOI: 10.22038/ijbms.2021.59604.13226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022]
Abstract
Cancer has been always considered as one of the main human health challenges worldwide. One of the main causes of cancer-related mortality is late diagnosis in the advanced stages of the disease, which reduces the therapeutic efficiency. Therefore, novel non-invasive diagnostic methods are required for the early detection of tumors and improving the quality of life and survival in cancer patients. MicroRNAs (miRNAs) have pivotal roles in various cellular processes such as cell proliferation, motility, and neoplastic transformation. Since circulating miRNAs have high stability in body fluids, they can be suggested as efficient noninvasive tumor markers. MiR-96 belongs to the miR-183-96-182 cluster that regulates cell migration and tumor progression as an oncogene or tumor suppressor by targeting various genes in solid tumors. In the present review, we have summarized all of the studies that assessed the role of miR-96 during tumor progression. This review clarifies the molecular mechanisms and target genes recruited by miR-96 to regulate tumor progression and metastasis. It was observed that miR-96 mainly affects tumorigenesis by targeting the structural proteins and FOXO transcription factors.
Collapse
Affiliation(s)
- Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Utilizing digital pathology to quantify stromal caveolin-1 expression in malignant and benign ovarian tumors: Associations with clinicopathological parameters and clinical outcomes. PLoS One 2021; 16:e0256615. [PMID: 34813586 PMCID: PMC8610269 DOI: 10.1371/journal.pone.0256615] [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: 08/04/2021] [Accepted: 11/09/2021] [Indexed: 11/19/2022] Open
Abstract
Loss of stromal caveolin-1 (Cav-1) is a biomarker of a cancer-associated fibroblast (CAF) phenotype and is related to progression, metastasis, and poor outcomes in several cancers. The objective of this study was to evaluate the clinical significance of Cav-1 expression in invasive epithelial ovarian cancer (OvCa). Epithelial and stromal Cav-1 expression were quantified in serous OvCa and benign ovarian tissue in two, independent cohorts–one quantified expression using immunohistochemistry (IHC) and the other using multiplex immunofluorescence (IF) with digital image analysis designed to target CAF-specific expression. Cav-1 expression was significantly downregulated in OvCa stroma compared to non-neoplastic stroma using both the IHC (p = 0.002) and IF (p = 1.8x10-13) assays. OvCa stroma showed Cav-1 downregulation compared to tumor epithelium with IHC (p = 1.2x10-24). Conversely, Cav-1 expression was higher in OvCa stroma compared to tumor epithelium with IF (p = 0.002). There was moderate correlation between IHC and IF methods for stromal Cav-1 expression (r2 = 0.69, p = 0.006) whereas there was no correlation for epithelial expression (r2 = 0.006, p = 0.98). Irrespective of the staining assay, neither response to therapy or overall survival correlated with the expression level of Cav-1 in the stroma or tumor epithelium. Our findings demonstrate a loss of stromal Cav-1 expression in ovarian serous carcinomas. Studies are needed to replicate these findings and explore therapeutic implications, particularly for immunotherapy response.
Collapse
|
20
|
Bertero L, Gambella A, Barreca A, Osella-Abate S, Chiusa L, Francia di Celle P, Lista P, Papotti M, Cassoni P. Caveolin-1 expression predicts favourable outcome and correlates with PDGFRA mutations in gastrointestinal stromal tumours (GISTs). J Clin Pathol 2021; 75:825-831. [PMID: 34155091 DOI: 10.1136/jclinpath-2021-207595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/08/2021] [Indexed: 11/04/2022]
Abstract
AIMS Novel prognostic markers are warranted for gastrointestinal stromal tumours. Caveolin-1 is a multifunctional protein that proved to be associated with outcome in multiple tumour types. Aim of this study was to investigate Caveolin-1 expression and prognostic efficacy in a series of gastrointestinal stromal tumours. METHODS Caveolin-1 expression was assessed by immunohistochemistry in a retrospective series of 66 gastrointestinal stromal tumours representative of the different molecular subtypes. Correlations with clinical, histopathological and molecular features were investigated. Statistical analyses were performed as appropriate. RESULTS Thirty-five cases out of 66 (53.0%) expressed Caveolin-1. Presence of Caveolin-1 expression correlated with favourable histopathologic and clinical traits, including a lower mitotic count (p=0.003) and lower relapse rate (p=0.005). Caveolin-1 expression also resulted associated with the presence of PDGFRA mutations (p=0.010). Outcome analyses showed a favourable prognostic significance of Caveolin-1 expression in terms of relapse-free survival (HR=0.14; 95% CI=0.03 to 0.63) and overall survival (HR=0.29; 95% CI=0.11 to 0.74), even after adjusting for the mutational subgroup (relapse-free survival: HR=0.14, 95% CI=0.04 to 0.44; overall survival: HR=0.29, 95% CI=0.11 to 0.51) and imatinib treatment (relapse-free survival: HR=0.14, 95% CI=0.02 to 0.81; overall survival: HR=0.29, 95% CI=0.17 to 0.48). CONCLUSION Caveolin-1 represents a novel prognostic marker in gastrointestinal stromal tumours. Further studies are warranted to validate these results and to explore the mechanisms linking Caveolin-1 expression with the PDGFRA oncogenic pathway.
Collapse
Affiliation(s)
- Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessandro Gambella
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Antonella Barreca
- Pathology Unit, "Città della Salute e della Scienza di Torino" University Hospital, Turin, Italy
| | - Simona Osella-Abate
- Molecular Pathology Unit, "Città della Salute e della Scienza di Torino" University Hospital, Turin, Italy
| | - Luigi Chiusa
- Pathology Unit, "Città della Salute e della Scienza di Torino" University Hospital, Turin, Italy
| | - Paola Francia di Celle
- Molecular Pathology Unit, "Città della Salute e della Scienza di Torino" University Hospital, Turin, Italy
| | - Patrizia Lista
- Oncology Unit, "Città della Salute e della Scienza di Torino" University Hospital, Turin, Italy
| | - Mauro Papotti
- Pathology Unit, Department of Oncology, University of Turin, Turin, Italy
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Yang C, He B, Dai W, Zhang H, Zheng Y, Wang X, Zhang Q. The role of caveolin-1 in the biofate and efficacy of anti-tumor drugs and their nano-drug delivery systems. Acta Pharm Sin B 2021; 11:961-977. [PMID: 33996409 PMCID: PMC8105775 DOI: 10.1016/j.apsb.2020.11.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
As one of the most important components of caveolae, caveolin-1 is involved in caveolae-mediated endocytosis and transcytosis pathways, and also plays a role in regulating the cell membrane cholesterol homeostasis and mediating signal transduction. In recent years, the relationship between the expression level of caveolin-1 in the tumor microenvironment and the prognostic effect of tumor treatment and drug treatment resistance has also been widely explored. In addition, the interplay between caveolin-1 and nano-drugs is bidirectional. Caveolin-1 could determine the intracellular biofate of specific nano-drugs, preventing from lysosomal degradation, and facilitate them penetrate into deeper site of tumors by transcytosis; while some nanocarriers could also affect caveolin-1 levels in tumor cells, thereby changing certain biophysical function of cells. This article reviews the role of caveolin-1 in tumor prognosis, chemotherapeutic drug resistance, antibody drug sensitivity, and nano-drug delivery, providing a reference for the further application of caveolin-1 in nano-drug delivery systems.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ADC, antibody drug conjugates
- BBB, blood–brain barrier
- Biofate
- CAFs, cancer-associated fibroblasts
- CPT, camptothecin
- CSD, caveolin scaffolding domain
- CTB, cholera toxins B
- Cancer
- Caveolin-1
- Drug resistance
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- ER, endoplasmic reticulum
- ERK, extracellular regulated protein kinases
- FGF2, fibroblast growth factor 2
- GGT, γ-glutamyl transpeptidase
- GPI, glycosylphosphatidylinositol
- HER2, human epidermal growth factor receptor 2
- HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A
- HSA, human serum albumin
- IBC, infiltrating breast cancer
- IR, insulin receptor
- MAPK, mitogen-activated protein kinase
- MDR, multidrug resistance
- MSV, multistage nanovectors
- NPs, nanoparticles
- Nano-drug delivery systems
- PC, prostate cancer
- PDGF, platelet-derived growth factor
- PFS, progression free survival
- ROS, reactive oxygen species
- SCLC, small cell lung cancer
- SV40, simian virus 40
- Transcytosis
- cell SMA, styrene maleic acid
Collapse
|
22
|
Panic A, Reis H, Wittka A, Darr C, Hadaschik B, Jendrossek V, Klein D. The Biomarker Potential of Caveolin-1 in Penile Cancer. Front Oncol 2021; 11:606122. [PMID: 33868995 PMCID: PMC8045968 DOI: 10.3389/fonc.2021.606122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/12/2021] [Indexed: 01/11/2023] Open
Abstract
Various types of human cancers were characterized by an altered expression of epithelial or stromal caveolin-1 (CAV1). However, the clinical significance of CAV1 expression in penile cancer remains largely unknown. Here the expression patterns of CAV1 were analyzed in a retrospective cohort (n=43) of penile squamous cell carcinomas (SCC). Upon penile cancer progression, significantly increased CAV1-levels were determined within the malignant epithelium, whereas within the tumor stroma, namely the fibroblastic tumor compartment harboring activated and/or cancer associated fibroblasts, CAV1 levels significantly decline. Concerning the clinicopathological significance of CAV1 expression in penile cancer as well as respective epithelial-stromal CAV1 distributions, high expression within the tumor cells as well as low expression of CAV1 within the stromal compartment were correlated with decreased overall survival of penile cancer patients. Herein, CAV1 expressions and distributions at advanced penile cancer stages were independent of the immunohistochemically proven tumor protein p53 status. In contrast, less differentiated p16-positive tumor epithelia (indicative for human papilloma virus infection) were characterized by significantly decreased CAV1 levels. Conclusively, we provide further and new evidence that the characteristic shift in stromal‐epithelial CAV1 being functionally relevant to tumor progression even occurs in penile SCC.
Collapse
Affiliation(s)
- Andrej Panic
- Department of Urology, West German Cancer Center, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Henning Reis
- Institute of Pathology, West German Cancer Center, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Alina Wittka
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Christopher Darr
- Department of Urology, West German Cancer Center, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Boris Hadaschik
- Department of Urology, West German Cancer Center, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| |
Collapse
|
23
|
Kaya S, Wiesmann N, Goldschmitt J, Krüger M, Al-Nawas B, Heider J. Differences in the expression of caveolin-1 isoforms in cancer-associated and normal fibroblasts of patients with oral squamous cell carcinoma. Clin Oral Investig 2021; 25:5823-5831. [PMID: 33774714 PMCID: PMC8443514 DOI: 10.1007/s00784-021-03887-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES For many years, tumor development has been viewed as a cell-autonomous process; however, today we know that the tumor microenvironment (TME) and especially cancer-associated fibroblasts (CAFs) significantly contribute to tumor progression. Caveolin-1 (Cav-1) is a scaffolding protein which is involved in several cancer-associated processes as important component of the caveolae. Our goal was to shed light on the expression of the two different isoforms of Cav-1 in normal fibroblasts (NFs) and CAFs of patients with oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS Fibroblasts from normal mucosa and CAFs were isolated and propagated in vitro. Gene expression of the different Cav-1 isoforms was assessed via quantitative real-time PCR (qPCR) and supplemented by protein expression analysis. RESULTS We could show that the Cav-1β isoform is more highly expressed in NFs and CAFs compared to Cav-1α. Furthermore, the different Cav-1 isoforms tended to be differently expressed in different tumor stages. However, this trend could not be seen consistently, which is in line with the ambiguous role of Cav-1 in tumor progression described in literature. Western blotting furthermore revealed that NFs and CAFs might differ in the oligomerization profile of the Cav-1 protein. CONCLUSION These differences in expression of Cav-1 between NFs and CAFs of patients with OSCC confirm that the protein might play a role in tumor progression and is of interest for further analyses. CLINICAL RELEVANCE Our findings support a possible role of the two isoforms of Cav-1 in the malignant transformation of OSCC.
Collapse
Affiliation(s)
- S Kaya
- Department of Oral and Maxillofacial Surgery Plastic Surgery, University Medical Center of the Johannes Gutenberg-University of Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - Nadine Wiesmann
- Department of Oral and Maxillofacial Surgery Plastic Surgery, University Medical Center of the Johannes Gutenberg-University of Mainz, Augustusplatz 2, 55131, Mainz, Germany. .,Molecular Tumor Biology, Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany.
| | - J Goldschmitt
- Department of Oral and Maxillofacial Surgery Plastic Surgery, University Medical Center of the Johannes Gutenberg-University of Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - M Krüger
- Department of Oral and Maxillofacial Surgery Plastic Surgery, University Medical Center of the Johannes Gutenberg-University of Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - B Al-Nawas
- Department of Oral and Maxillofacial Surgery Plastic Surgery, University Medical Center of the Johannes Gutenberg-University of Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - J Heider
- Department of Oral and Maxillofacial Surgery Plastic Surgery, University Medical Center of the Johannes Gutenberg-University of Mainz, Augustusplatz 2, 55131, Mainz, Germany
| |
Collapse
|
24
|
Zhu G, Cao B, Liang X, Li L, Hao Y, Meng W, He C, Wang L, Li L. Small extracellular vesicles containing miR-192/215 mediate hypoxia-induced cancer-associated fibroblast development in head and neck squamous cell carcinoma. Cancer Lett 2021; 506:11-22. [PMID: 33639203 DOI: 10.1016/j.canlet.2021.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
The mechanisms underlying the hypoxic cancer cell-mediated differentiation of cancer-associated fibroblasts (CAFs) have not been elucidated yet. The present study showed that the hypoxic head and neck squamous cell carcinoma (HNSCC) cells promoted CAF-like differentiation through secreting TGF-β and small extracellular vesicles (sEVs) that contain enhanced levels of miR-192/215 family miRNAs. Caveolin-1 (CAV1), which is a target gene of miR-192/215, inhibited the TGF-β/SMAD signaling and promoted CAF-like differentiation of the fibroblasts. Restoring the levels of CAV1 inhibited the hypoxic sEV- and TGF-β-induced CAF-like differentiation. The enhanced levels of miR-192/215 encapsulated in the HNSCC tissue-derived sEVs (but not serum-derived sEVs) indicated hypoxic and aggressive cancer stroma. miR-215 in the tumor tissue-derived sEVs (but not circulating sEVs) was correlated with poor overall survival of patients with HNSCC. This study demonstrated that sEVs function as a "courier" to deliver miRNAs from the cancer cells to the fibroblasts, which promotes the remodeling of the hypoxic tumor microenvironment, and that cancer tissue-derived sEV could potentially serve as a source of biomarker.
Collapse
Affiliation(s)
- Guiquan Zhu
- Department of Head and Neck Oncology, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, Sichuan University, 610041, Chengdu, China.
| | - Bangrong Cao
- Sichuan Key Laboratory of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610041, Chengdu, China
| | - Xinhua Liang
- Department of Head and Neck Oncology, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, Sichuan University, 610041, Chengdu, China
| | - Longjiang Li
- Department of Head and Neck Oncology, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, Sichuan University, 610041, Chengdu, China
| | - Yaying Hao
- Sichuan Key Laboratory of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610041, Chengdu, China
| | - Wanrong Meng
- Sichuan Key Laboratory of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610041, Chengdu, China
| | - Chuanshi He
- Sichuan Key Laboratory of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610041, Chengdu, China
| | - Linlin Wang
- Sichuan Key Laboratory of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610041, Chengdu, China
| | - Ling Li
- Sichuan Key Laboratory of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610041, Chengdu, China.
| |
Collapse
|
25
|
Abstract
Caveolae are specialised and dynamic plasma membrane subdomains, involved in many cellular functions including endocytosis, signal transduction, mechanosensing and lipid storage, trafficking, and metabolism. Two protein families are indispensable for caveola formation and function, namely caveolins and cavins. Mutations of genes encoding these caveolar proteins cause serious pathological conditions such as cardiomyopathies, skeletal muscle diseases, and lipodystrophies. Deregulation of caveola-forming protein expression is associated with many types of cancers including prostate cancer. The distinct function of secretion of the prostatic fluid, and the unique metabolic phenotype of prostate cells relying on lipid metabolism as a main bioenergetic pathway further suggest a significant role of caveolae and caveolar proteins in prostate malignancy. Accumulating in vitro, in vivo, and clinical evidence showed the association of caveolin-1 with prostate cancer grade, stage, metastasis, and drug resistance. In contrast, cavin-1 was found to exhibit tumour suppressive roles. Studies on prostate cancer were the first to show the distinct function of the caveolar proteins depending on their localisation within the caveolar compartment or as cytoplasmic or secreted proteins. In this review, we summarise the roles of caveola-forming proteins in prostate cancer and the potential of exploiting them as therapeutic targets or biological markers.
Collapse
|
26
|
Wang Y, Lin Z, Song J, Wei S, Ye Z, Chen S, Zeng Y, Lin Z, Chen X, Chen L. MicroRNA-451a targets caveolin-1 in stomach cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2524-2533. [PMID: 33165443 PMCID: PMC7642723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/29/2020] [Indexed: 06/11/2023]
Abstract
MicroRNA-451 (miR-451) is lowly expressed in stomach cancer cells and improves their metastatic ability by down-regulating extracellular signal-regulated kinase 2 (ERK2). Many studies have found that caveolin-1 (CAV1) plays an important role in cancer progression. Additionally, miR-451 has been reported to regulate the expression of CAV1 in chronic obstructive pulmonary disease. Therefore, this study aims to determine if miR-451 regulates the biological functions of stomach cancer cells by regulating CAV1 expression. Through a bioinformatics analysis, we found that miR-451a regulates CAV1 expression, and miR-451a expression is relatively low in stomach cancer cells. Next, we confirmed that miR-451a negatively regulates CAV1 expression using a dual-luciferase reporter assay. Then MTT, 5-ethynyl-2'-deoxyuridine (EdU), propidium iodide (PI), an Annexin V-FITC/PI apoptosis kit, and transwell assays were used to measure the changes in cell proliferation, the cell cycle, apoptosis, cell migration, and invasiveness in stomach cancer cells overexpressing miR-451a or both miR-451a and CAV1. It was found that increasing the miR-451a expression in stomach cancer cells inhibits cell growth, migration, and invasiveness, and promotes apoptosis. After restoring the CAV1 expression, these biological processes resumed. In summary, in stomach cancer cells, the overexpression of miR-451a can restrain cell growth and promote apoptosis, so it is a potential treatment for stomach cancer.
Collapse
Affiliation(s)
- Yi Wang
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Zhenmeng Lin
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Jintian Song
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Shenghong Wei
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Zaisheng Ye
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Shu Chen
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Yi Zeng
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Zhitao Lin
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Xiaoling Chen
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| | - Luchuan Chen
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital Fuzhou 350014, Fujian, China
| |
Collapse
|
27
|
Ye JH, Shi JJ, Yin X, Wu HY, Xu XY, Yao YZ, Zhang WJ. Elevated Expression of CAV1 is Associated with Unfavorable Prognosis of Patients with Breast Cancer Who Undergo Surgery and Neoadjuvant Chemotherapy. Cancer Manag Res 2020; 12:8887-8892. [PMID: 33061584 PMCID: PMC7519866 DOI: 10.2147/cmar.s264673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT), which is standard treatment for locally advanced breast cancer, improves the resectability of patients with early breast cancer and reduces the extent of breast and axillary surgery. Caveolin-1 (CAV1) is implicated in human cancers, although its utility for cancer prognosis is unknown. Here, we investigated the expression of CAV1 in breast cancer tissues to evaluate its prognostic significance on patients with breast cancer administered NACT. METHODS CAV1 expression in 80 breast cancer tissue samples was evaluated using immunohistochemistry (IHC). The association between CAV1 levels and clinical factors was analyzed using the chi-square test and that between CAV1 and prognosis was evaluated using multivariate Cox regression and Kaplan-Meier analyses. RESULTS High levels of CAV1 were significantly associated with survival, and patients with overexpression of CAV1 had a poor prognosis. Adjusted multivariate Cox regression analyses revealed that a high level of CAV1 expression was an independent, significant prognostic factor for patients with breast cancer treated with NACT. DISCUSSION Overexpression of CAV1 in patients with breast cancer administered NACT was associated with shorter disease-free survival and overall survival. Therefore, high levels of CAV1 may serve as a prognostic biomarker for such patients.
Collapse
Affiliation(s)
- Jia-Hui Ye
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Jia-Jun Shi
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xi Yin
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Hong-Yan Wu
- Pathology Department, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xin-Yun Xu
- Pathology Department, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Yong-Zhong Yao
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Wei-Jie Zhang
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
28
|
Low JY, Brennen WN, Meeker AK, Ikonen E, Simons BW, Laiho M. Stromal CAVIN1 Controls Prostate Cancer Microenvironment and Metastasis by Modulating Lipid Distribution and Inflammatory Signaling. Mol Cancer Res 2020; 18:1414-1426. [PMID: 32493699 DOI: 10.1158/1541-7786.mcr-20-0364] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/01/2020] [Accepted: 05/28/2020] [Indexed: 11/16/2022]
Abstract
Lipid uptake occurs through caveolae, plasma membrane invaginations formed by caveolins (CAV) and caveolae-associated protein 1 (CAVIN1). Genetic alterations of CAV1N1 and CAV1 modify lipid metabolism and underpin lipodystrophy syndromes. Lipids contribute to tumorigenesis by providing fuel to cancer metabolism and supporting growth and signaling. Tumor stroma promotes tumor proliferation, invasion, and metastasis, but how stromal lipids influence these processes remain to be defined. Here, we show that stromal CAVIN1 regulates lipid abundance in the prostate cancer microenvironment and suppresses metastasis. We show that depletion of CAVIN1 in prostate stromal cells markedly reduces their lipid droplet accumulation and increases inflammation. Stromal cells lacking CAVIN1 enhance prostate cancer cell migration and invasion. Remarkably, they increase lipid uptake and M2 inflammatory macrophage infiltration in the primary tumors and metastasis to distant sites. Our data support the concept that stromal cells contribute to prostate cancer aggressiveness by modulating lipid content and inflammation in the tumor microenvironment. IMPLICATIONS: This study showed that stromal CAVIN1 suppresses prostate cancer metastasis by modulating tumor microenvironment, lipid content, and inflammatory response.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - W Nathaniel Brennen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan K Meeker
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elina Ikonen
- Faculty of Medicine, Anatomy and Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Brian W Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
29
|
Caveolin-1 regulates the ASMase/ceramide-mediated radiation response of endothelial cells in the context of tumor-stroma interactions. Cell Death Dis 2020; 11:228. [PMID: 32273493 PMCID: PMC7145831 DOI: 10.1038/s41419-020-2418-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/21/2022]
Abstract
The integral membrane protein caveolin-1 (CAV1) plays a central role in radioresistance-mediating tumor–stroma interactions of advanced prostate cancer (PCa). Among the tumor–stroma, endothelial cells (EC) evolved as critical determinants of the radiation response. CAV1 deficiency in angiogenic EC was already shown to account for increased apoptosis rates of irradiated EC. This study explores the potential impact of differential CAV1 levels in EC on the acid sphingomyelinase (ASMase)/ceramide pathway as a key player in the regulation of EC apoptosis upon irradiation and cancer cell radioresistance. Enhanced apoptosis sensitivity of CAV1-deficient EC was associated with increased ASMase activity, ceramide generation, formation of large lipid platforms, and finally an altered p38 mitogen-activated protein kinase (MAPK)/heat-shock protein 27 (HSP27)/AKT (protein kinase B, PKB) signaling. CAV1-deficient EC increased the growth delay of LNCaP and PC3 PCa cells upon radiation treatment in direct 3D spheroid co-cultures. Exogenous C6 and C16 ceramide treatment in parallel increased the growth delay of PCa spheroids and induced PCa cell apoptosis. Analysis of the respective ceramide species in PCa cells with increased CAV1 levels like those typically found in radio-resistant advanced prostate tumors further revealed an upregulation of unsaturated C24:1 ceramide that might scavenge the effects of EC-derived apoptosis-inducing C16 ceramide. Higher ASMase as well as ceramide levels could be confirmed by immunohistochemistry in human advanced prostate cancer specimen bearing characteristic CAV1 tumor–stroma alterations. Conclusively, CAV1 critically regulates the generation of ceramide-dependent (re-)organization of the plasma membrane that in turn affects the radiation response of EC and adjacent PCa cells. Understanding the CAV1-dependent crosstalk between tumor cells and the host-derived tumor microvasculature and its impact on radiosensitivity may allow to define a rational strategy for overcoming tumor radiation resistance improving clinical outcomes by targeting CAV1.
Collapse
|
30
|
Nikas JB, Mitanis NT, Nikas EG. Whole Exome and Transcriptome RNA-Sequencing Model for the Diagnosis of Prostate Cancer. ACS OMEGA 2020; 5:481-486. [PMID: 31956794 PMCID: PMC6964263 DOI: 10.1021/acsomega.9b02995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 06/02/2023]
Abstract
In our previous study, we developed a genome-wide DNA methylation model for the diagnosis of prostate cancer, and we pointed out that a considerable average error is associated with the current method for the diagnosis of prostate cancer, which is predicated on pathological assessment of biopsied tissue. In this study, we utilized whole exome and transcriptome RNA-sequencing (RNA-seq) data that were derived from 468 tumor samples and 51 normal samples of prostatic tissue, and we analyzed over 20,000 genes per sample. We were able to develop a mathematical model that classified tumor tissue versus normal tissue with a high accuracy. The overall sensitivity was 97.01%, and the overall specificity was 94.12%. The input variables to the model were the mRNA expression values of the following nine genes: ANGPT1, MED21, AOX1, PLP2, HPN, HPN-AS1, EPHA10, NKX2-3, and LRFN1. The model was validated with unknown samples, with a 10-fold cross-validation, and a leave-one-out cross-validation. We present here a genomic model, based on a whole exome and transcriptome RNA-seq analysis of biopsied prostatic tissue, that could be utilized in the diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Jason B. Nikas
- Research
& Development, Genomix Inc., Minneapolis, Minnesota 55364, United States
| | - Nikos T. Mitanis
- Department
of Mathematics, University of the Aegean, Samos 83200, Greece
| | - Emily G. Nikas
- School
of Mathematics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
31
|
Lin CJ, Yun EJ, Lo UG, Tai YL, Deng S, Hernandez E, Dang A, Chen YA, Saha D, Mu P, Lin H, Li TK, Shen TL, Lai CH, Hsieh JT. The paracrine induction of prostate cancer progression by caveolin-1. Cell Death Dis 2019; 10:834. [PMID: 31685812 PMCID: PMC6828728 DOI: 10.1038/s41419-019-2066-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/21/2022]
Abstract
A subpopulation of cancer stem cells (CSCs) plays a critical role of cancer progression, recurrence, and therapeutic resistance. Many studies have indicated that castration-resistant prostate cancer (CRPC) is associated with stem cell phenotypes, which could further promote neuroendocrine transdifferentiation. Although only a small subset of genetically pre-programmed cells in each organ has stem cell capability, CSCs appear to be inducible among a heterogeneous cancer cell population. However, the inductive mechanism(s) leading to the emergence of these CSCs are not fully understood in CRPC. Tumor cells actively produce, release, and utilize exosomes to promote cancer development and metastasis, cancer immune evasion as well as chemotherapeutic resistance; the impact of tumor-derived exosomes (TDE) and its cargo on prostate cancer (PCa) development is still unclear. In this study, we demonstrate that the presence of Cav-1 in TDE acts as a potent driver to induce CSC phenotypes and epithelial-mesenchymal transition in PCa undergoing neuroendocrine differentiation through NFκB signaling pathway. Furthermore, Cav-1 in mCRPC-derived exosomes is capable of inducing radio- and chemo-resistance in recipient cells. Collectively, these data support Cav-1 as a critical driver for mCRPC progression.
Collapse
Affiliation(s)
- Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Eun-Jin Yun
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Division of Integrative Bioscience and Biotechnology, POSTECH, Pohang, 37673, Republic of Korea
| | - U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yu-Ling Tai
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Su Deng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Elizabeth Hernandez
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew Dang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yu-An Chen
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ping Mu
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tsai-Kun Li
- Department and Graduate Institute of Microbiology, National Taiwan University, Taipei, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
32
|
Yan C, Sun C, Ding X, Rizeq FK, Ren M, Yang F, Chen Y, Wang B. Association of CAV1 polymorphisms with the risks of breast cancer: A systematic review and meta-analysis. Pathol Res Pract 2019; 215:152518. [PMID: 31303379 DOI: 10.1016/j.prp.2019.152518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Caveolin-1 (CAV1) polymorphisms have been shown to correlated with breast cancer risk in previous studies. However, the role of CAV1 polymorphisms still remained indecisive, and dual functions of CAV1 was demonstrated in breast cancer development. Consequently, a meta-analysis to evaluate and summarize the association of the CAV1 polymorphisms with breast cancer susceptibility. MATERIAL AND METHODS Extensive search was performed in PubMed, Web of Science, Google scholar, EMBASE.com, CNKI and Wanfang searching platform up to March 2019. The Newcastle-Ottawa Scale (NOS) were used to evaluate the quality of each study. The Odds ratios (ORs) and the 95% confidence intervals (CIs) were analyzed to evaluate the strength of the associations in five genetic models. Inter-study heterogeneity was quantified using the I-squared (I2) test. In addition, the Egger's test and Begg's test were applied to evaluate the publication bias. RESULTS 4 case-control studies with 2115 cases and 2138 controls were enrolled into this analysis. There was a significant association between rs3807987 polymorphism of CAV1 and breast cancer in allele comparison (A vs. G: OR = 1.288, 95%CI = 1.162-1.428, P < 0.001), heterozygote comparison (AG vs. GG: OR= 1.422, 95%CI=1.233-1.639, P < 0.001), and dominant comparison (AA+AG vs. GG: OR=1.395, 95%CI=1.228-1.586, P < 0.001). A significant association of rs3807987 polymorphism in allele comparison (A vs. G: OR=1.238, 95%CI=1.109-1.383, P < 0.001), heterozygote comparison (AG VS. GG: OR=1.466, 95%CI=1.267-1.697, P < 0.05), and dominant comparison (AA+AG vs. GG: OR=1.384, 95%CI=1.209-1.585, P < 0.001) was also founded amongst Chinese population. A significant association between rs7804372 polymorphism and breast cancer amongst Chinese population in recessive comparison (AA vs. AT + TT: OR = 0.730, 95%CI = 0.567-0.940, P = 0.015) was identified. No significant association between breast cancer risk and rs1997623 was found. CONCLUSION CAV1 rs3807987 and rs7804372 polymorphisms are associated with the change of breast cancer risk. More well-designed and large studies in various populations are needed to further elaborate these associations.
Collapse
Affiliation(s)
- Cunye Yan
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL 60657, USA
| | - Xiuxiu Ding
- Lianhua Community Health Service Centre, The Second Affiliated Hospital of Anhui Medical University, 217 Furong Street, Hefei, Anhui, PR China
| | - Feras Kamel Rizeq
- Avalon University School of Medicine, Santa Rosaweg 122-124, Willemstad, Curaçao
| | - Min Ren
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China
| | - Fan Yang
- Maternal and Chile Health Care Hospital of Anhui Province, No.15 Yimin Street, Hefei, 230001, Anhui, PR China
| | - Ying Chen
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China
| | - Benzhong Wang
- Department of Breast Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, PR China.
| |
Collapse
|
33
|
Caveolin-1 enhances brain metastasis of non-small cell lung cancer, potentially in association with the epithelial-mesenchymal transition marker SNAIL. Cancer Cell Int 2019; 19:171. [PMID: 31297035 PMCID: PMC6599320 DOI: 10.1186/s12935-019-0892-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023] Open
Abstract
Background Caveolin-1 (Cav-1) plays an important role in the development of various human cancers. We investigated the relationship between Cav-1 expression and non-small cell lung cancer (NSCLC) progression in the context of brain metastasis (BM). Methods Cav-1 expression was investigated in a series of 102 BM samples and 49 paired primary NSCLC samples, as well as 162 unpaired primary NSCLC samples with (63 cases) or without (99 cases) metastasis to distant organs. Human lung cancer cell lines were used for in vitro functional analysis. Results High Cav-1 expression in tumor cells was observed in 52% (38/73) of squamous cell carcinomas (SQCs) and 33% (45/138) of non-SQCs. In SQC, high Cav-1 expression was increased after BM in both paired and unpaired samples of lung primary tumors and BM (53% vs. 84% in paired samples, P = 0.034; 52% vs. 78% in unpaired samples, P = 0.020). Although the difference in median overall survival in patients NSCLC was not statistically significant, high Cav-1 expression in tumor cells (P = 0.005, hazard ratio 1.715, 95% confidence index 1.175–2.502) was independent prognostic factors of overall survival on multivariate Cox regression analyses, in addition to the presence of BM and non-SQC type. In vitro assays revealed that Cav-1 knockdown inhibited the invasion and migration of lung cancer cells. Genetic modulation of Cav-1 was consistently associated with SNAIL up- and down-regulation. These findings were supported by increased SNAIL and Cav-1 expression in BM samples of SQC. Conclusions Cav-1 plays an important role in the BM of NSCLC, especially in SQC. The mechanism may be linked to SNAIL regulation. Electronic supplementary material The online version of this article (10.1186/s12935-019-0892-0) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
Zhang Y, Fan W, Wu J, Dong J, Cui Z. Association of caveolin-1 protein expression with hepatocellular carcinoma: a meta-analysis and literature review. Cancer Manag Res 2019; 11:5113-5122. [PMID: 31239768 PMCID: PMC6553953 DOI: 10.2147/cmar.s194033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/07/2019] [Indexed: 01/08/2023] Open
Abstract
Background: Aberrant expression of caveolin-1 (CAV-1) is involved in the pathogenesis of hepatocellular carcinoma (HCC); however, the results have been inconsistent due to the small size of sample in the individual study. Methods: We performed a meta-analysis and evaluated the association of CAV-1 protein overexpression and clinicopathological significance by using Review Manager 5.2. Pooled ORs and HR with corresponding CIs were calculated. Results: Nine studies were included in the meta-analysis with 810 HCC and 172 cirrhosis patients. CAV-1 protein overexpression was correlated with the risk of cirrhosis; OR was 3.25, p=0.01. Furthermore, the rate of CAV-1 protein overexpression was significantly higher in HCC with cirrhosis than HCC without cirrhosis, suggesting that the CAV-1 protein overexpression likely initiated carcinogenesis in liver with cirrhosis and subsequently contributed to the progression of HCC. In addition, CAV-1 protein overexpression was strongly associated with poor differentiated HCC and invasion; ORs were 2.61 and 2.71, respectively. CAV-1 protein overexpression was strongly correlated with poor overall survival in patients with HCC; HR was 0.4, p=0.03. Conclusions: In summary, CAV-1 protein overexpression is at risk for liver cirrhosis and HCC derived from cirrhosis, and CAV-1 is also a promising prognostic predictor in HCC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng 475000, People's Republic of China
| | - Wenjuan Fan
- Medical Bioengineering Key Laboratory, Luohe Medical College, Luohe 462002, People's Republic of China
| | - Jiang Wu
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng 475000, People's Republic of China
| | - Jinglong Dong
- Department of Pathology, Huaihe Hospital, Henan University, Kaifeng 475000, People's Republic of China
| | - Zhanjun Cui
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, People's Republic of China
| |
Collapse
|
35
|
Lopes MB, Casimiro S, Vinga S. Twiner: correlation-based regularization for identifying common cancer gene signatures. BMC Bioinformatics 2019; 20:356. [PMID: 31238876 PMCID: PMC6593597 DOI: 10.1186/s12859-019-2937-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/06/2019] [Indexed: 12/27/2022] Open
Abstract
Background Breast and prostate cancers are typical examples of hormone-dependent cancers, showing remarkable similarities at the hormone-related signaling pathways level, and exhibiting a high tropism to bone. While the identification of genes playing a specific role in each cancer type brings invaluable insights for gene therapy research by targeting disease-specific cell functions not accounted so far, identifying a common gene signature to breast and prostate cancers could unravel new targets to tackle shared hormone-dependent disease features, like bone relapse. This would potentially allow the development of new targeted therapies directed to genes regulating both cancer types, with a consequent positive impact in cancer management and health economics. Results We address the challenge of extracting gene signatures from transcriptomic data of prostate adenocarcinoma (PRAD) and breast invasive carcinoma (BRCA) samples, particularly estrogen positive (ER+), and androgen positive (AR+) triple-negative breast cancer (TNBC), using sparse logistic regression. The introduction of gene network information based on the distances between BRCA and PRAD correlation matrices is investigated, through the proposed twin networks recovery (twiner) penalty, as a strategy to ensure similarly correlated gene features in two diseases to be less penalized during the feature selection procedure. Conclusions Our analysis led to the identification of genes that show a similar correlation pattern in BRCA and PRAD transcriptomic data, and are selected as key players in the classification of breast and prostate samples into ER+ BRCA/AR+ TNBC/PRAD tumor and normal tissues, and also associated with survival time distributions. The results obtained are supported by the literature and are expected to unveil the similarities between the diseases, disclose common disease biomarkers, and help in the definition of new strategies for more effective therapies.
Collapse
Affiliation(s)
- Marta B Lopes
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, Lisboa, 1049-001, Portugal. .,INESC-ID, Instituto Superior Técnico, Universidade de Lisboa, Rua Alves Redol 9, Lisboa, 1000-029, Portugal.
| | - Sandra Casimiro
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, 1649-028, Portugal
| | - Susana Vinga
- INESC-ID, Instituto Superior Técnico, Universidade de Lisboa, Rua Alves Redol 9, Lisboa, 1000-029, Portugal.,IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, Lisboa, 1049-001, Portugal
| |
Collapse
|
36
|
Liu B, Zhang J, Yang D. miR-96-5p promotes the proliferation and migration of ovarian cancer cells by suppressing Caveolae1. J Ovarian Res 2019; 12:57. [PMID: 31228941 PMCID: PMC6588920 DOI: 10.1186/s13048-019-0533-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the second most common gynaecological malignancy. MicroRNAs (miRNAs) have been found to be aberrantly expressed in OC tissue and have been proposed as biomarkers and therapeutic targets for OC. RESULTS In this study, we found that miR-96-5p was up-regulated in OC tissues and OC cells compared to normal ovarian tissues and epithelial cell line. And, miR-96-5p was also up-regulated in the serum samples from OC patients compared to health participants. In addition, there was a positive correlation of miR-96-5p levels between OC tissues and serum samples. At the cellular level, overexpression of miR-96-5p promoted cell proliferation and migration in OC cells. Moreover, we further validated Caveolae1 (CAV1) as the direct target of miR-96-5p in OC cells through luciferase activity assays and western blot. CAV1 was obvious low expression in OC tissues. The overexpression of CAV1 abrogated the promotion of miR-96-5p on the OC cells proliferation and migration. Finally, we found that AKT signaling pathway was involved in this process. MiR-96-5p inhibited the phosphorylation of AKT and expression of down-stream proteins Cyclin D1 and P70 by targeting CAV1. CONCLUSIONS The above findings suggested that targeting miR-96-5p may be a promising strategy for OC treatment.
Collapse
Affiliation(s)
- Bo Liu
- Department of gynaecology & obstetrics, Jinan Maternal & Children Health Care Hospital, No 2 Jianguo Xiaojing Three road, Jinan, 250001, Shandong, People's Republic of China
| | - Jinglu Zhang
- Department of gynaecology & obstetrics, Jinan Maternal & Children Health Care Hospital, No 2 Jianguo Xiaojing Three road, Jinan, 250001, Shandong, People's Republic of China
| | - Dongxia Yang
- Department of gynaecology & obstetrics, Jinan Maternal & Children Health Care Hospital, No 2 Jianguo Xiaojing Three road, Jinan, 250001, Shandong, People's Republic of China.
| |
Collapse
|
37
|
Ketteler J, Panic A, Reis H, Wittka A, Maier P, Herskind C, Yagüe E, Jendrossek V, Klein D. Progression-Related Loss of Stromal Caveolin 1 Levels Mediates Radiation Resistance in Prostate Carcinoma via the Apoptosis Inhibitor TRIAP1. J Clin Med 2019; 8:jcm8030348. [PMID: 30871022 PMCID: PMC6462938 DOI: 10.3390/jcm8030348] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/16/2022] Open
Abstract
Tumour resistance to chemo- and radiotherapy, as well as molecularly targeted therapies, limits the effectiveness of current cancer treatments. We previously reported that the radiation response of human prostate tumours is critically regulated by CAV1 expression in stromal fibroblasts and that loss of stromal CAV1 expression in advanced tumour stages may contribute to tumour radiotherapy resistance. Here we investigated whether fibroblast secreted anti-apoptotic proteins could induce radiation resistance of prostate cancer cells in a CAV1-dependent manner and identified TRIAP1 (TP53 Regulated Inhibitor of Apoptosis 1) as a resistance-promoting CAV1-dependent factor. TRIAP1 expression and secretion was significantly higher in CAV1-deficient fibroblasts and secreted TRIAP1 was able to induce radiation resistance of PC3 and LNCaP prostate cancer cells in vitro, as well as of PC3 prostate xenografts derived from co-implantation of PC3 cells with TRIAP1-expressing fibroblasts in vivo. Immunohistochemical analyses of irradiated PC3 xenograft tumours, as well as of human prostate tissue specimen, confirmed that the characteristic alterations in stromal-epithelial CAV1 expression were accompanied by increased TRIAP1 levels after radiation in xenograft tumours and within advanced prostate cancer tissues, potentially mediating resistance to radiation treatment. In conclusion, we have determined the role of CAV1 alterations potentially induced by the CAV1-deficient, and more reactive, stroma in radio sensitivity of prostate carcinoma at a molecular level. We suggest that blocking TRIAP1 activity and thus avoiding drug resistance may offer a promising drug development strategy for inhibiting resistance-promoting CAV1-dependent signals.
Collapse
Affiliation(s)
- Julia Ketteler
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Virchowstrasse 173, 45122 Essen, Germany.
| | - Andrej Panic
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Virchowstrasse 173, 45122 Essen, Germany.
- Department of Urology and Urooncology, University of Duisburg-Essen, University Hospital, Essen, Hufelandstr. 55, 45122 Essen, Germany.
| | - Henning Reis
- Institute of Pathology, University of Duisburg-Essen, University Hospital, Hufelandstr. 55, 45122 Essen, Germany.
| | - Alina Wittka
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Virchowstrasse 173, 45122 Essen, Germany.
| | - Patrick Maier
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| | - Carsten Herskind
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| | - Ernesto Yagüe
- Cancer Research Center, Division of Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Virchowstrasse 173, 45122 Essen, Germany.
| | - Diana Klein
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Virchowstrasse 173, 45122 Essen, Germany.
| |
Collapse
|
38
|
Gupta VK, Sharma NS, Kesh K, Dauer P, Nomura A, Giri B, Dudeja V, Banerjee S, Bhattacharya S, Saluja A, Banerjee S. Metastasis and chemoresistance in CD133 expressing pancreatic cancer cells are dependent on their lipid raft integrity. Cancer Lett 2018; 439:101-112. [PMID: 30290209 DOI: 10.1016/j.canlet.2018.09.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/25/2018] [Accepted: 09/21/2018] [Indexed: 02/01/2023]
Abstract
Metabolic rewiring is an integral part of tumor growth. Among metabolic pathways, the Mevalonic-Acid-Pathway (MVAP) plays a key role in maintaining membrane architecture through cholesterol synthesis, thereby affecting invasiveness. In the current study, we show for the first time that CD133Hi pancreatic tumor initiating cells (TIC) have increased expression of MVAP enzymes, cholesterol-content and Caveolin expression. Further, we show that CD133 in these cells is localized in the lipid-rafts (characterized by Cav-1-cholesterol association). Disruption of lipid-rafts by either depleting Cav-1 or by inhibiting MVAP by lovastatin decreased metastatic-potential and chemoresistance in CD133Hi cells while not affecting the CD133lo cells. Additionally, disruption of lipid-raft results in deregulation of FAK-signaling, decreasing invasiveness in pancreatic-TICs. Furthermore, this also inhibits ABC-transporter activity resulting in sensitizing TICs to standard chemotherapeutic agents. Repurposing existing drugs for new clinical applications is one of the safest and least resource intensive approaches to improve therapeutic options. In this context, our study is extremely timely as it shows that targeting lipid-rafts with statins can sensitize the normally resistant pancreatic TICHi-cells to standard chemotherapy and decrease metastasis, thereby defining a novel strategy for targeting the TICHi-PDAC.
Collapse
Affiliation(s)
| | - Nikita S Sharma
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Kousik Kesh
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Patricia Dauer
- Department of Surgery, University of Miami, Miami, FL, 33136, USA; Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Alice Nomura
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Bhuwan Giri
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Vikas Dudeja
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Santanu Banerjee
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | | | - Ashok Saluja
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Sulagna Banerjee
- Department of Surgery, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
39
|
Comparing the expression profiles of steroid hormone receptors and stromal cell markers in prostate cancer at different Gleason scores. Sci Rep 2018; 8:14326. [PMID: 30254333 PMCID: PMC6156570 DOI: 10.1038/s41598-018-32711-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/12/2018] [Indexed: 12/19/2022] Open
Abstract
The recent developments in anti-angiogenic and immunomodulatory drugs show that the tumour micro-environment (TME) becomes increasingly important in cancer research. Here we investigated the correlation between the Gleason score (GS) and the TME by comparing tissue expression profiles of steroid hormone receptors, cancer activated fibroblast (CAF) markers and vessel densities between different GS groups. Therefore, matched patient cohorts were composed for different GS (6-7-8). Tissue micro-arrays with 6 samples/patient were processed for immunohistochemistry. Stained slides were digitised, stroma and epithelium were selectively annotated, and all selected areas were quantitatively analysed for marker expression. The most striking findings were decreased stromal expression levels of several steroid hormone receptors, increased CAF-phenotypes and increased vessel densities in high GS prostate cancer compared to low GS prostate cancer and paired prostate non-tumour tissue. The present data reveal a complex correlation between prostate cancer differentiation and TME components and suggest that different GS can be associated with different possible actionable targets in the TME. The use of standardised digital image analysis tools generated robust and reproducible quantitative data, which is novel and more informative compared to the classic semi-quantitative and observer-dependent visual scoring of immunohistochemistry.
Collapse
|
40
|
Gillard M, Javier R, Ji Y, Zheng SL, Xu J, Brendler CB, Crawford SE, Pierce BL, Griend DJV, Franco OE. Elevation of Stromal-Derived Mediators of Inflammation Promote Prostate Cancer Progression in African-American Men. Cancer Res 2018; 78:6134-6145. [PMID: 30181178 DOI: 10.1158/0008-5472.can-17-3810] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/23/2018] [Accepted: 08/29/2018] [Indexed: 11/16/2022]
Abstract
Progress in prostate cancer racial disparity research has been hampered by a lack of appropriate research tools and better understanding of the tumor biology. Recent gene expression studies suggest that the tumor microenvironment (TME) may contribute to racially disparate clinical outcomes in prostate cancer. Analysis of the prostate TME has shown increased reactive stroma associated with chronic inflammatory infiltrates in African-American (AA) compared with European-American (EA) patients with prostate cancer. To better understand stromal drivers of changes in TME, we isolated prostate fibroblasts (PrF) from AA (PrF-AA) and EA (PrF-EA) prostate cancer tissues and studied their functional characteristics. PrF-AA showed increased growth response to androgens FGF2 and platelet-derived growth factor. Compared with PrF-EA, conditioned media from PrF-AA significantly enhanced the proliferation and motility of prostate cancer cell lines. Expression of markers associated with myofibroblast activation (αSMA, vimentin, and tenascin-C) was elevated in PrF-AA In vivo tumorigenicity of an AA patient-derived prostatic epithelial cell line E006AA was significantly increased in the presence of PrF-AA compared with PrF-EA, and RNA-seq data and cytokine array analysis identified a panel of potential proinflammatory paracrine mediators (BDNF, CHI3L1, DPPIV, FGF7, IL18BP, IL6, and VEGF) to be enriched in PrF-AA E006AA cell lines showed increased responsiveness to BDNF ligand compared with EA-derived LNCaP and C4-2B cells. Addition of a TrkB-specific antagonist significantly reduced the protumorigenic effects induced by PrF-AA compared with PrF-EA These findings suggest that fibroblasts in the TME of AA patients may contribute to the health disparity observed in the incidence and progression of prostate cancer tumors.Significance: These findings suggest that stromal cells in the tumor microenvironment of African-American men promote progression of prostate cancer by increasing levels of a specific set of pro-inflammatory molecules compared with European-American men.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/21/6134/F1.large.jpg Cancer Res; 78(21); 6134-45. ©2018 AACR.
Collapse
Affiliation(s)
- Marc Gillard
- Department of Surgery, Section of Urology, The University of Chicago, Chicago, Illinois
| | - Rodrigo Javier
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Yuan Ji
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - S Lilly Zheng
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Jianfeng Xu
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Charles B Brendler
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Susan E Crawford
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois
| | - Brandon L Pierce
- Department of Public Health Sciences, The University of Chicago, Chicago, Illinois
| | | | - Omar E Franco
- Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, Illinois.
| |
Collapse
|
41
|
Li S, Chen Y, Zhang Y, Jiang X, Jiang Y, Qin X, Yang H, Wu C, Liu Y. Shear stress promotes anoikis resistance of cancer cells via caveolin-1-dependent extrinsic and intrinsic apoptotic pathways. J Cell Physiol 2018; 234:3730-3743. [PMID: 30171601 DOI: 10.1002/jcp.27149] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/05/2018] [Indexed: 02/02/2023]
Abstract
Circulating tumor cells (CTCs) need to acquire resistance to anoikis to survive after they experience fluid shear stress in the circulatory and lymphatic systems. However, the mechanism by which tumor cells resist anoikis under shear stress conditions remains unknown. Here, we found that the application of low shear stress (LSS; 2 dyn/cm2 ) to human breast carcinoma cells (MDA-MB-231) resulted in increased anoikis resistance when tumor cells were grown under anchorage-independent conditions. Caveolin-1 (Cav-1), the major component of plasma membrane caveolae, was overexpressed in LSS-treated cells and prevented tumor cells from anoikis, while depletion of Cav-1 restored sensitivity to anoikis. LSS-induced dissociation of Cav-1-Fas inhibited formation of the death-inducing signaling complex, caspase-8 activation, and rendered tumor cells resistant to anoikis. Likewise, LSS blocked the mitochondrial pathway through promotion of integrin β1-focal adhesion kinase-mediated multicellular aggregation and suppression of truncated BID translocation mediated crosstalk between the extrinsic and intrinsic apoptotic pathways. Our findings provide insights into the mechanisms by which LSS induces anoikis resistance in breast carcinoma cells through inhibition of Cav-1-dependent extrinsic and intrinsic apoptotic pathways, and serves as a potential therapeutic target for CTCs and metastatic breast cancer.
Collapse
Affiliation(s)
- Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuehui Zhang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaomin Jiang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Jiang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,The Institute of Cancer Research, School of Clinical Medicine/The Affiliated Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
42
|
Codrici E, Albulescu L, Popescu ID, Mihai S, Enciu AM, Albulescu R, Tanase C, Hinescu ME. Caveolin-1-Knockout Mouse as a Model of Inflammatory Diseases. J Immunol Res 2018; 2018:2498576. [PMID: 30246033 PMCID: PMC6136523 DOI: 10.1155/2018/2498576] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/05/2018] [Indexed: 02/07/2023] Open
Abstract
Caveolin-1 (CAV1) is the scaffold protein of caveolae, which are minute invaginations of the cell membrane that are involved in endocytosis, cell signaling, and endothelial-mediated inflammation. CAV1 has also been reported to have a dual role as either a tumor suppressor or tumor promoter, depending on the type of cancer. Inflammation is an important player in tumor progression, but the role of caveolin-1 in generating an inflammatory milieu remains poorly characterized. We used a caveolin-1-knockout (CAV1-/-) mouse model to assess the inflammatory status via the quantification of the pro- and anti-inflammatory cytokine levels, as well as the ability of circulating lymphocytes to respond to nonspecific stimuli by producing cytokines. Here, we report that the CAV1-/- mice were characterized by a low-grade systemic proinflammatory status, with a moderate increase in the IL-6, TNF-α, and IL-12p70 levels. CAV1-/- circulating lymphocytes were more prone to cytokine production upon nonspecific stimulation than the wild-type lymphocytes. These results show that CAV1 involvement in cell homeostasis is more complex than previously revealed, as it plays a role in the inflammatory process. These findings indicate that the CAV1-/- mouse model could prove to be a useful tool for inflammation-related studies.
Collapse
Affiliation(s)
- Elena Codrici
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Lucian Albulescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | | | - Simona Mihai
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Ana-Maria Enciu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050047 Bucharest, Romania
| | - Radu Albulescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- National Institute for Chemical Pharmaceutical R&D, Bucharest, Romania
| | - Cristiana Tanase
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Mihail E. Hinescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, 050047 Bucharest, Romania
| |
Collapse
|
43
|
Herek TA, Robinson JE, Heavican TB, Amador C, Iqbal J, Cutucache CE. Caveolin-1 is dispensable for early lymphoid development, but plays a role in the maintenance of the mature splenic microenvironment. BMC Res Notes 2018; 11:470. [PMID: 30005686 PMCID: PMC6043983 DOI: 10.1186/s13104-018-3583-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 07/06/2018] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE Caveolin-1 (CAV1) is known for its role as both a tumor suppressor and an oncogene, harboring a highly context-dependent role within a myriad of malignancies and cell types. In an immunological context, dysregulation of CAV1 expression has been shown to alter immunological signaling functions and suggests a pivotal role for CAV1 in the facilitation of proper immune responses. Nonetheless, it is still unknown how Cav1-deficiency and heterozygosity would impact the development and composition of lymphoid organs in mice. Herein, we investigated the impacts of Cav1-dysregulation on the lymphoid organs in young (12 weeks) and aged (36 weeks) Cav1+/+, Cav1+/-, and Cav1-/- mice. RESULTS We observed that only Cav1-deficiency is associated with persistent splenomegaly at all timepoints. Furthermore, no differences in overall body weight were detected (and without sexual dimorphisms). Both aged Cav1+/- and Cav1-/- mice present with decreased CD19+CD22+ B cells and secondary-follicle atrophy, specifically in the spleen, compared with wild-type controls and irrespective of splenomegaly status. Consequently, the demonstrated effects on B cell homeostasis and secondary follicle characteristics prompted our investigation into follicle-derived human B-cell lymphomas. Our investigation points toward CAV1 as a dysregulated protein in follicle-derived B-cell malignancies without harboring a differential expression between more aggressive and indolent hematological malignancies.
Collapse
Affiliation(s)
- Tyler A Herek
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jacob E Robinson
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge St, Omaha, NE, 68182, USA
| | - Tayla B Heavican
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Catalina Amador
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Christine E Cutucache
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge St, Omaha, NE, 68182, USA.
| |
Collapse
|
44
|
Protective effect of stromal Dickkopf-3 in prostate cancer: opposing roles for TGFBI and ECM-1. Oncogene 2018; 37:5305-5324. [PMID: 29858602 PMCID: PMC6160402 DOI: 10.1038/s41388-018-0294-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/12/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022]
Abstract
Aberrant transforming growth factor-β (TGF-β) signaling is a hallmark of the stromal microenvironment in cancer. Dickkopf-3 (Dkk-3), shown to inhibit TGF-β signaling, is downregulated in prostate cancer and upregulated in the stroma in benign prostatic hyperplasia, but the function of stromal Dkk-3 is unclear. Here we show that DKK3 silencing in WPMY-1 prostate stromal cells increases TGF-β signaling activity and that stromal cell-conditioned media inhibit prostate cancer cell invasion in a Dkk-3-dependent manner. DKK3 silencing increased the level of the cell-adhesion regulator TGF-β-induced protein (TGFBI) in stromal and epithelial cell-conditioned media, and recombinant TGFBI increased prostate cancer cell invasion. Reduced expression of Dkk-3 in patient tumors was associated with increased expression of TGFBI. DKK3 silencing reduced the level of extracellular matrix protein-1 (ECM-1) in prostate stromal cell-conditioned media but increased it in epithelial cell-conditioned media, and recombinant ECM-1 inhibited TGFBI-induced prostate cancer cell invasion. Increased ECM1 and DKK3 mRNA expression in prostate tumors was associated with increased relapse-free survival. These observations are consistent with a model in which the loss of Dkk-3 in prostate cancer leads to increased secretion of TGFBI and ECM-1, which have tumor-promoting and tumor-protective roles, respectively. Determining how the balance between the opposing roles of extracellular factors influences prostate carcinogenesis will be key to developing therapies that target the tumor microenvironment.
Collapse
|
45
|
Stromal Caveolin-1 and Caveolin-2 Expression in Primary Tumors and Lymph Node Metastases. Anal Cell Pathol (Amst) 2018; 2018:8651790. [PMID: 29850392 PMCID: PMC5914130 DOI: 10.1155/2018/8651790] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/23/2018] [Indexed: 12/26/2022] Open
Abstract
The expression of caveolin-1 (CAV1) in both tumor cell and cancer-associated fibroblasts (CAFs) has been found to correlate with tumor aggressiveness in different epithelial tumor entities, whereas less is known for caveolin-2 (CAV2). The aim of this study was to investigate the clinicopathological significance and prognostic value of stromal CAV1 and CAV2 expression in lung cancer. The expression of these two genes was investigated at protein level on a tissue microarray (TMA) consisting of 161 primary tumor samples. 50.7% of squamous cell lung cancer (SCC) tumors showed strong expression of CAV1 in the tumor-associated stromal cells, whereas only 15.1% of adenocarcinomas (AC) showed a strong CAV1 expression (p < 0.01). A strong CAV2 stromal expression was found in 46.0% of the lung tumor specimens, with no significant difference between the subtypes. Neither CAV1 nor CAV2 stromal expression was associated with any other clinicopathological factor including survival. When the stromal expression in matched primary tumors and lymph node metastases was compared, both CAV1 and CAV2 expressions were frequently found lost in the corresponding stroma of the lymph node metastasis (40.6%, p = 0.003 and 38.4%, p = 0.001, resp.). Loss of stromal CAV2 in the lymph node metastases was also significantly associated with earlier death (p = 0.011). In conclusion, in contrast to the expression patterns in the tumor tissue of lung cancer, stromal expression of CAV1 in primary tumors was not associated with clinical outcome whereas the stromal expression of especially CAV2 in the metastatic lymph nodes could be associated with lung cancer pathogenesis.
Collapse
|
46
|
Abstract
Resistance of solid tumors to chemo- and radiotherapy remains a major obstacle in anti-cancer treatment. Herein, the membrane protein caveolin-1 (CAV1) came into focus as it is highly expressed in many tumors and high CAV1 levels were correlated with tumor progression, invasion and metastasis, and thus a worse clinical outcome. Increasing evidence further indicates that the heterogeneous tumor microenvironment, also known as the tumor stroma, contributes to therapy resistance resulting in poor clinical outcome. Again, CAV1 seems to play an important role in modulating tumor host interactions by promoting tumor growth, metastasis, therapy resistance and cell survival. However, the mechanisms driving stroma-mediated tumor growth and radiation resistance remain to be clarified. Understanding these interactions and thus, targeting CAV1 may offer a novel strategy for preventing cancer therapy resistance and improving clinical outcomes. In this review, we will summarize the resistance-promoting effects of CAV1 in tumors, and emphasize its role in the tumor-stroma communication as well as the resulting malignant phenotype of epithelial tumors.
Collapse
Affiliation(s)
- Julia Ketteler
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
47
|
Huang K, Fang C, Yi K, Liu X, Qi H, Tan Y, Zhou J, Li Y, Liu M, Zhang Y, Yang J, Zhang J, Li M, Kang C. The role of PTRF/Cavin1 as a biomarker in both glioma and serum exosomes. Theranostics 2018; 8:1540-1557. [PMID: 29556340 PMCID: PMC5858166 DOI: 10.7150/thno.22952] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/05/2017] [Indexed: 12/30/2022] Open
Abstract
Exosomes play critical roles in intercellular communication in both nearby and distant cells in individuals and organs. Polymerase I and transcript release factor (PTRF), also known as Cavin1, has previously been described as a critical factor in caveola formation, and aberrant PTRF expression has been reported in various malignancies. However, the function of PTRF in tumor progression remains controversial, and its role in glioma is poorly understood. In this study, we report that PTRF is associated with malignancy grade and poor prognosis in glioma patients. Our previous study using two proteomics methods, tandem mass tag (TMT) and data-independent acquisition (DIA), showed that EGFRvIII overexpression increased PTRF expression at the protein level. In contrast, blocking PI3K and AKT using LY294002 and MK-2206, respectively, decreased PTRF expression, showing that PTRF is regulated in the EGFR/PI3K/AKT pathway. ChIP-PCR analysis showed that PTRF is transcriptionally regulated by the H3K4me3 and H3K27me3 modifications. Furthermore, PTRF overexpression increased exosome secretion and induced cell growth in vitro. More importantly, overexpressing PTRF induced the malignancy of nearby cells in vivo, suggesting that PTRF alters the microenvironment through intercellular communication via exosomes. Furthermore, analysis of clinical samples showed a positive correlation between tumor grade and PTRF expression in both tumor tissues and exosomes isolated from blood harvested from glioma patients, and PTRF expression in exosomes isolated from the sera of GBM patients was decreased after surgery. In conclusion, PTRF serves as a promising biomarker in both tumor samples and serum exosomes, thus facilitating the detection of glioma and potentially serving as a therapeutic target for glioblastoma multiforme.
Collapse
Affiliation(s)
- Kai Huang
- Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Chuan Fang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Neurosurgery, Hebei University Affiliated Hospital, Baoding 071000, China
| | - Kaikai Yi
- Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Capital Medical University,Beijing,100050,China
| | - Hongzhao Qi
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University , Tianjin 300072, China
| | - Yanli Tan
- College of Fundamental Medicine, Hebei University, Baoding 071000, China
| | - Junhu Zhou
- Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Ying Li
- Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Mingyang Liu
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yuqing Zhang
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Min Li
- Department of Medicine, Department of Surgery, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Chunsheng Kang
- Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| |
Collapse
|
48
|
Webber JP, Spary LK, Mason MD, Tabi Z, Brewis IA, Clayton A. Prostate stromal cell proteomics analysis discriminates normal from tumour reactive stromal phenotypes. Oncotarget 2018; 7:20124-39. [PMID: 26934553 PMCID: PMC4991442 DOI: 10.18632/oncotarget.7716] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/27/2016] [Indexed: 01/05/2023] Open
Abstract
Changes within interstitial stromal compartments often accompany carcinogenesis, and this is true of prostate cancer. Typically, the tissue becomes populated by myofibroblasts that can promote progression. Not all myofibroblasts exhibit the same negative influence, however, and identifying the aggressive form of myofibroblast may provide useful information at diagnosis. A means of molecularly defining such myofibroblasts is unknown. We compared protein profiles of normal and diseased stroma isolated from prostate cancer patients to identify discriminating hallmarks of disease-associated stroma. We included the stimulation of normal stromal cells with known myofibroblast inducers namely soluble TGFβ and exosome-associated-TGFβ and compared the function and protein profiles arising. In all 6-patients examined, diseased stroma exhibited a pro-angiogenic influence on endothelial cells, generating large multicellular vessel-like structures. Identical structures were apparent following stimulation of normal stroma with exosomes (5/6 patients), but TGFβ-stimulation generated a non-angiogenic stroma. Proteomics highlighted disease-related cytoskeleton alterations such as elevated Transgelin (TAGLN). Many of these were also changed following TGFβ or exosome stimulation and did not well discriminate the nature of the stimulus. Soluble TGFβ, however triggered differential expression of proteins related to mitochondrial function including voltage dependent ion channels VDAC1 and 2, and this was not found in the other stromal types studied. Surprisingly, Aldehyde Dehydrogenase (ALDH1A1), a stem-cell associated protein was detected in normal stromal cells and found to decrease in disease. In summary, we have discovered a set of proteins that contribute to defining disease-associated myofibroblasts, and emphasise the similarity between exosome-generated myofibroblasts and those naturally arising in situ.
Collapse
Affiliation(s)
- Jason P Webber
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Lisa K Spary
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Malcolm D Mason
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Zsuzsanna Tabi
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Ian A Brewis
- Institute of Translation, Innovation, Methodology and Engagement (TIME), Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Aled Clayton
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| |
Collapse
|
49
|
Yang W, Xu T, Qiu P, Xu G. Caveolin-1 promotes pituitary adenoma cells migration and invasion by regulating the interaction between EGR1 and KLF5. Exp Cell Res 2018; 367:7-14. [PMID: 29309750 DOI: 10.1016/j.yexcr.2018.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/29/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
Caveolin-1 (Cav-1) is a principal structural protein of caveolae. Cav-1 has been implicated in cancer progression, but its precise functional roles in pituitary adenoma cells remain largely unclear. In this study, we evidenced that the level of cav-1 was elevated in the invasive pituitary adenoma. Cav-1 knockdown restrained the migration and invasion of pituitary adenoma cells. In cav-1-depleting cells, the expression of miR-145, miR-124 and miR-183 were up-regulated. Further investigation showed that cav-1 knockdown inhibited the nuclear translocation of EGR1, reducing the interaction between EGR1 and KLF5. The resulting free KLF5 promoted the expression of miR-145, miR-124 and miR-183 by binding to their promoters, which was blocked by EGR1. Luciferase reporter assay indicated that miR-145 targeted FSCN1, miR-124 targeted PTTG1IP, and miR-183 targeted EZR in pituitary adenoma cells, respectively. Knockdown of FSCN1, PTTG1IP or EZR suppressed the migration and invasion of pituitary adenoma cells. In conclusion, our data suggested that the elevated cav-1 promoted pituitary adenoma cells migration and invasion by regulating the interaction between EGR1 and KLF5.
Collapse
Affiliation(s)
- Wei Yang
- Department of neurosurgery, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Tongjiang Xu
- Department of neurosurgery, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Peng Qiu
- Department of neurosurgery, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Guangming Xu
- Department of neurosurgery, Shandong provincial Hospital Affiliated to Shandong University, Jinan 250021, China.
| |
Collapse
|
50
|
Liu Z, Yu J, Wu R, Tang S, Cai X, Guo G, Chen S. Rho/ROCK Pathway Regulates Migration and Invasion of Esophageal Squamous Cell Carcinoma by Regulating Caveolin-1. Med Sci Monit 2017; 23:6174-6185. [PMID: 29288243 PMCID: PMC5757863 DOI: 10.12659/msm.905820] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 07/07/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a common cancer with poor prognosis. Caveolin-1 (Cav1) and Rho/ROCK pathway play important roles in tumor metastasis, separately. However, less research was focused on the relationship between Cav1 and Rho/ROCK in ECSS metastasis. Therefore, we investigated the relationship between Cav1 and Rho/ROCK pathway in ESCC metastasis. MATERIAL AND METHODS Cav1 and phosphorylated Cav1 (PY14Cav1) were examined in ESCC and in adjacent and non-tumorous tissues from ESCC patients by immunohistochemistry (IHC). Small interfering RNA (siRNA) targeting Cav1 or Rho/ROCK inhibitor was used to treat EC109, Eca109, TE1, and TE13 cells. Western blotting (WB) was used to detect Cav1 and PY14Cav1 expression. The wound healing scratch test and transwell assays were used to assess migration and invasion. RESULTS Cav1 and PY14Cav1 were gradually expressed at higher levels in ECSS than in adjacent and non-tumor tissues as ESCC stage and lymphatic metastasis increased, and this difference was significant (P<0.05). Cav1 was expressed at higher levels in TE1 and TE13 than in EC109 and Eca109, while PY14Cav1 was enhanced in TE1 and TE13 cells but not in EC109 and Eca109, and the difference was significant (P<0.05). TE1 and TE13 had significantly (P<0.05) stronger motility, migratory, and invasion abilities than EC109 and Eca109 cells. Silencing Cav1 decreased PY14Cav1 expression in TE1 and TE13 cells, as well as suppressing the migration and invasion of all ECSS cells, and these differences were significant (P<0.05). Suppressing the Rho/ROCK pathway obviously inhibited Cav1 and PY14Cav1 expressions, as well as significantly (P<0.05) decreasing migration and invasion of ESCC cells. CONCLUSIONS Cav1 and PY14Cav1 were positively correlated with ESCC lymphatic metastasis and cancer stages. Rho/ROCK pathway activation promoted ESCC metastasis by regulating Cav1.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Jing Yu
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Ruinuan Wu
- Department of Pathology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Shengxin Tang
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Xiaoman Cai
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Guanghua Guo
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
- Corresponding Authors: Guanghua Guo, e-mail: , Suzuan Chen, e-mail:
| | - Suzuan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, P.R. China
- Corresponding Authors: Guanghua Guo, e-mail: , Suzuan Chen, e-mail:
| |
Collapse
|