1
|
Jackson Cullison SR, Flemming JP, Karagoz K, Wermuth PJ, Mahoney MG. Mechanisms of extracellular vesicle uptake and implications for the design of cancer therapeutics. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70017. [PMID: 39483807 PMCID: PMC11522837 DOI: 10.1002/jex2.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024]
Abstract
The translation of pre-clinical anti-cancer therapies to regulatory approval has been promising, but slower than hoped. While innovative and effective treatments continue to achieve or seek approval, setbacks are often attributed to a lack of efficacy, failure to achieve clinical endpoints, and dose-limiting toxicities. Successful efforts have been characterized by the development of therapeutics designed to specifically deliver optimal and effective dosing to tumour cells while minimizing off-target toxicity. Much effort has been devoted to the rational design and application of synthetic nanoparticles to serve as targeted therapeutic delivery vehicles. Several challenges to the successful application of this modality as delivery vehicles include the induction of a protracted immune response that results in their rapid systemic clearance, manufacturing cost, lack of stability, and their biocompatibility. Extracellular vesicles (EVs) are a heterogeneous class of endogenous biologically produced lipid bilayer nanoparticles that mediate intercellular communication by carrying bioactive macromolecules capable of modifying cellular phenotypes to local and distant cells. By genetic, chemical, or metabolic methods, extracellular vesicles (EVs) can be engineered to display targeting moieties on their surface while transporting specific cargo to modulate pathological processes following uptake by target cell populations. This review will survey the types of EVs, their composition and cargoes, strategies employed to increase their targeting, uptake, and cargo release, and their potential as targeted anti-cancer therapeutic delivery vehicles.
Collapse
Affiliation(s)
| | - Joseph P. Flemming
- Rowan‐Virtua School of Osteopathic MedicineRowan UniversityStratfordNew JerseyUSA
| | - Kubra Karagoz
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Mỹ G. Mahoney
- Departments of PharmacologyPhysiology, and Cancer Biology, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Otolaryngology – Head and Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Villa-Martínez E, Rios A, Gutiérrez-Vidal R, Escalante B. Potentiation of anti-angiogenic eNOS-siRNA transfection by ultrasound-mediated microbubble destruction in ex vivo rat aortic rings. PLoS One 2024; 19:e0308075. [PMID: 39088581 PMCID: PMC11293687 DOI: 10.1371/journal.pone.0308075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/16/2024] [Indexed: 08/03/2024] Open
Abstract
Nitric oxide (NO) regulates vascular homeostasis and plays a key role in revascularization and angiogenesis. The endothelial nitric oxide synthase (eNOS) enzyme catalyzes NO production in endothelial cells. Overexpression of the eNOS gene has been implicated in pathologies with dysfunctional angiogenic processes, such as cancer. Therefore, modulating eNOS gene expression using small interfering RNAs (siRNAs) represents a viable strategy for antitumor therapy. siRNAs are highly specific to the target gene, thus reducing off-target effects. Given the widespread distribution of endothelium and the crucial physiological role of eNOS, localized delivery of nucleic acid to the affected area is essential. Therefore, the development of an efficient eNOS-siRNA delivery carrier capable of controlled release is imperative for targeting specific vascular regions, particularly those associated with tumor vascular growth. Thus, this study aims to utilize ultrasound-mediated microbubble destruction (UMMD) technology with cationic microbubbles loaded with eNOS-siRNA to enhance transfection efficiency and improve siRNA delivery, thereby preventing sprouting angiogenesis. The efficiency of eNOS-siRNA transfection facilitated by UMMD was assessed using bEnd.3 cells. Synthesis of nitric oxide and eNOS protein expression were also evaluated. The silencing of eNOS gene in a model of angiogenesis was assayed using the rat aortic ring assay. The results showed that from 6 to 24 h, the transfection of fluorescent siRNA with UMMD was twice as high as that of lipofection. Moreover, transfection of eNOS-siRNA with UMMD enhanced the knockdown level (65.40 ± 4.50%) compared to lipofectamine (40 ± 1.70%). Silencing of eNOS gene with UMMD required less amount of eNOS-siRNA (42 ng) to decrease the level of eNOS protein expression (52.30 ± 0.08%) to the same extent as 79 ng of eNOS-siRNA using lipofectamine (56.30 ± 0.10%). NO production assisted by UMMD was reduced by 81% compared to 67% reduction transfecting with lipofectamine. This diminished NO production led to higher attenuation of aortic ring outgrowth. Three-fold reduction compared to lipofectamine transfection. In conclusion, we propose the combination of eNOS-siRNA and UMMD as an efficient, safe, non-viral nucleic acid transfection strategy for inhibition of tumor progression.
Collapse
Affiliation(s)
- Elisa Villa-Martínez
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| | - Amelia Rios
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| | - Roxana Gutiérrez-Vidal
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
- Programa de Investigadoras e Investigadores por México, CONAHCyT/Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| | - Bruno Escalante
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, México
| |
Collapse
|
3
|
Lin MC, Kuo WH, Chen SY, Hsu JY, Lu LY, Wang CC, Chen YJ, Tsai JS, Li HJ. Ago2/CAV1 interaction potentiates metastasis via controlling Ago2 localization and miRNA action. EMBO Rep 2024; 25:2441-2478. [PMID: 38649663 PMCID: PMC11094075 DOI: 10.1038/s44319-024-00132-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024] Open
Abstract
Ago2 differentially regulates oncogenic and tumor-suppressive miRNAs in cancer cells. This discrepancy suggests a secondary event regulating Ago2/miRNA action in a context-dependent manner. We show here that a positive charge of Ago2 K212, that is preserved by SIR2-mediated Ago2 deacetylation in cancer cells, is responsible for the direct interaction between Ago2 and Caveolin-1 (CAV1). Through this interaction, CAV1 sequesters Ago2 on the plasma membranes and regulates miRNA-mediated translational repression in a compartment-dependent manner. Ago2/CAV1 interaction plays a role in miRNA-mediated mRNA suppression and in miRNA release via extracellular vesicles (EVs) from tumors into the circulation, which can be used as a biomarker of tumor progression. Increased Ago2/CAV1 interaction with tumor progression promotes aggressive cancer behaviors, including metastasis. Ago2/CAV1 interaction acts as a secondary event in miRNA-mediated suppression and increases the complexity of miRNA actions in cancer.
Collapse
Affiliation(s)
- Meng-Chieh Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, 100229, Taiwan
| | - Shih-Yin Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jing-Ya Hsu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Li-Yu Lu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chen-Chi Wang
- Department of Surgery, National Taiwan University Hospital, Taipei, 100229, Taiwan
| | - Yi-Ju Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Jia-Shiuan Tsai
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Hua-Jung Li
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan.
- Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung City, 402, Taiwan.
| |
Collapse
|
4
|
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
|
5
|
Hwang N, Yoon BK, Chun KH, Kim H, Lee Y, Kim JW, Jeon H, Kim TH, Kim MY, Fang S, Cheong JH, Kim JW. Caveolin-1 mediates the utilization of extracellular proteins for survival in refractory gastric cancer. Exp Mol Med 2023; 55:2461-2472. [PMID: 37919422 PMCID: PMC10689497 DOI: 10.1038/s12276-023-01109-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 11/04/2023] Open
Abstract
Despite advances in cancer therapy, the clinical outcome of patients with gastric cancer remains poor, largely due to tumor heterogeneity. Thus, finding a hidden vulnerability of clinically refractory subtypes of gastric cancer is crucial. Here, we report that chemoresistant gastric cancer cells rely heavily on endocytosis, facilitated by caveolin-1, for survival. caveolin-1 was highly upregulated in the most malignant stem-like/EMT/mesenchymal (SEM)-type gastric cancer cells, allowing caveolin-1-mediated endocytosis and utilization of extracellular proteins via lysosomal degradation. Downregulation of caveolin-1 alone was sufficient to induce cell death in SEM-type gastric cancer cells, emphasizing its importance as a survival mechanism. Consistently, chloroquine, a lysosomal inhibitor, successfully blocked caveolin-1-mediated endocytosis, leading to the marked suppression of tumor growth in chemorefractory gastric cancer cells in vitro, including patient-derived organoids, and in vivo. Together, our findings suggest that caveolin-1-mediated endocytosis is a key metabolic pathway for gastric cancer survival and a potential therapeutic target.
Collapse
Affiliation(s)
- Nahee Hwang
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo Kyung Yoon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Hye Chun
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeonhui Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoseob Lee
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Won Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeonuk Jeon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hyun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi-Young Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jae-Ho Cheong
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of R&D, Veraverse Inc., Seoul, Republic of Korea.
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
6
|
López-Rodulfo IM, Villa-Martínez E, Rios A, Escalante B. Caveolin Delivered by Ultrasound-Mediated Microbubble Destruction Prevents Endothelial Cell Proliferation. Cell Mol Bioeng 2023; 16:219-229. [PMID: 37456788 PMCID: PMC10338419 DOI: 10.1007/s12195-023-00763-1] [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: 08/11/2022] [Accepted: 03/29/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction The nitric oxide synthase (eNOS) is an important regulator of vascular homeostasis. eNOS is modulated by intracellular mechanisms that include protein-protein interaction with Caveolin-1 (Cav). Cav binds to and impairs eNOS activation reducing vascular permeability and angiogenesis. Blocking of eNOS by Cav has been proposed as therapeutic antiangiogenic approach. However, the efficient and controlled delivery of the peptide requires to be solved. Methods The effect of antennapedia (AP)-Cav loaded into microbubbles (MBs) and delivered by ultrasound-mediated microbubble destruction (UMMD) into brain endothelial cells (bEnd.3 cells) was evaluated on NO production using DAF2-DA, cell migration assessed by the wound healing assay, cell proliferation with BrdU, and ex-vivo angiogenesis in rat aortic rings. Results An enhanced inhibitory effect of AP-Cav was observed on cells treated with UMMD. MBs and ultrasound disruption delivery of AP-Cav increased acetylcholine-induced NO release, wound healing, cell proliferation, and angiogenesis inhibition on bEnd.3 cells, compared to free AP-Cav administration. Conclusion We demonstrated that the delivery of Cav via AP-Cav-loaded MBs and UMMD may be an administration method for Cav that would increase its therapeutic potential by enhancing efficacy and cellular specificity.
Collapse
Affiliation(s)
- Iván M. López-Rodulfo
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
- Present Address: Aarhus Universitet, Nordre Ringgade 1, 8000 Aarhus C, Denmark
| | - Elisa Villa-Martínez
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
| | - Amelia Rios
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
| | - Bruno Escalante
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
| |
Collapse
|
7
|
Lee S, Jung Park M, Joo Lee H, Kil Joo J, Soo Suh D, Un Choi K, Hyung Kim K, Chul Kim S. Decreased expression of caveolin-1 have relevance to promoted senescence in preeclamptic placenta. Pregnancy Hypertens 2022; 30:59-67. [PMID: 36007380 DOI: 10.1016/j.preghy.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVES To assess the association between altered expression of caveolin-1 and p53/p21, as indicatives of cellular senescence, in preeclamptic placenta. STUDY DESIGN Placental tissues and serum were collected from rats (Sham and reduced uterine perfusion pressure group) at 18.5 days post coitum and humans (normotensive pregnant and preeclampsia groups). The concentration and expression of caveolin-1 were measured in the collected tissues, and the correlation between p53 and p21 expression was evaluation. MAIN OUTCOME MEASURES Placental mRNA expression and serum concentration of caveolin-1 were measured using qRT-PCR and ELISA, respectively. Altered expressions of caveolin-1 and p53/p21 were revealed and quantified by immunohistochemistry. The association between these changes was investigated using correlation analysis. RESULTS Placental mRNA expressions and serum concentrations of caveolin-1 were significantly decreased in reduced uterine perfusion pressure and preeclampsia groups. The expressions of caveolin-1 and p53/ p21 were significantly altered in placenta complicated with preeclampsia. Correlation analysis revealed a significant inverse association between changes in caveolin-1 and p53/p21. Subsequently, these results were obtained by investigating the preeclampsia onset time. CONCLUSION These results revealed that the expression of caveolin-1 profoundly decreases in the placenta and serum of preeclampsia. These factors contribute to the mechanism of accelerated cellular senescence in placenta, which is one of the various etiologies of preeclampsia.
Collapse
Affiliation(s)
- Sul Lee
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Republic of Korea; Biomedical Research Institute Pusan National University Hospital, Republic of Korea
| | - Min Jung Park
- The Korea Institute for Public Sperm Bank, Republic of Korea
| | - Hyun Joo Lee
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Republic of Korea; Biomedical Research Institute Pusan National University Hospital, Republic of Korea
| | - Jong Kil Joo
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Republic of Korea; Biomedical Research Institute Pusan National University Hospital, Republic of Korea
| | - Dong Soo Suh
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Republic of Korea; Biomedical Research Institute Pusan National University Hospital, Republic of Korea
| | - Kyung Un Choi
- Biomedical Research Institute Pusan National University Hospital, Republic of Korea; Department of Pathology, Pusan National University School of Medicine, Republic of Korea
| | - Ki Hyung Kim
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Republic of Korea; Biomedical Research Institute Pusan National University Hospital, Republic of Korea
| | - Seung Chul Kim
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Republic of Korea; Biomedical Research Institute Pusan National University Hospital, Republic of Korea.
| |
Collapse
|
8
|
Yang C, Fan S, Wang X, Liu W, Yang L, He B, Dai W, Zhang H, Wang X, Zhang Q. Optimize the combination regimen of Trastuzumab and Nab-paclitaxel in HER2-positive tumors via modulating Caveolin-1 expression by lovastatin. Asian J Pharm Sci 2022; 17:697-712. [PMID: 36382307 PMCID: PMC9640369 DOI: 10.1016/j.ajps.2022.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/12/2022] [Accepted: 06/25/2022] [Indexed: 11/02/2022] Open
Abstract
The combination regimen of trastuzumab (Tras) plus Nab-paclitaxel (Nab) is recommended to treat HER2-positive (HER2+) cancers. However, they exert effects in different mechanisms: Tras need to stay on cell membranes, while Nab need to be endocytosed, therefore the concurrent combination regimen may not be the best one in HER2+ tumors treatment. Caveolin-1 (Cav-1) is a key player in mediating their endocytosis and is associated with their efficacy, but few researches noticed the opposite effect of Cav-1 expression on the combination efficacy. Herein, we systematically studied the Cav-1 expression level on the combination efficacy and proposed an optimized and clinically feasible combination regimen for HER2+ Cav-1High tumor treatment. In the regimen, lovastatin (Lova) was introduced to modulate the Cav-1 expression and the results indicated that Lova could downregulate Cav-1 expression, increase Tras retention on cell membrane and enhance the in vitro cytotoxicity of Tras in HER2+ Cav-1High cells but not in HER2+ Cav-1Low cells. Therefore, by exchanging the dosing sequence of Nab and Tras, and by adding Lova at appropriate time points, the precise three-drug-sequential regimen (PTDS, Nab(D1)-Lova(D2)-Lova & Tras(D2+12 h)) was established. Compared with the concurrent regimen, the PTDS regimen exhibited a higher in vitro cytotoxicity and a stronger tumor growth inhibition in HER2+ Cav-1High tumors, which might be a promising combination regimen for these patients in clinics.
Collapse
Affiliation(s)
- Canyu Yang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
| | - Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xing Wang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
| | - Wei Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100083, China
| | - Long Yang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Science, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
9
|
Deficiency of Urokinase-Type Plasminogen Activator Receptor Is Associated with the Development of Perivascular Fibrosis in Mouse Heart. Bull Exp Biol Med 2022; 173:5-9. [PMID: 35622258 DOI: 10.1007/s10517-022-05480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Indexed: 10/18/2022]
Abstract
It was suggested that the urokinase system plays a certain role in the regulation of activity of the endothelial-mesenchymal transition and in the development of perivascular fibrosis. Urokinase (uPA), the key component of the urokinase system, is a serine protease that binds to its receptor on the cell surface (uPAR) and affects the cell microenvironment components through the formation of plasmin, remodeling of the extracellular matrix, release of growth factors, and initiation of intracellular signals. The heart of PLAUR gene knockout C57BL/129 (uPAR-/-) mice showed signs of vasculopathy: reduced number of capillaries/arterioles, signs of endothelial-mesenchymal transition in endothelial cells, vascular wall remodeling, and deposition of extracellular matrix components. These changes were combined with enhanced expression of urokinase and active forms of TGF-β1. Apparently, uPAR is a part of a multicomponent system that provides multifaceted regulatory effects on the components of forming vessels and vascular wall cells, which allows considering it as a possible target for targeted antifibrotic therapy.
Collapse
|
10
|
Jones JH, Minshall RD. Endothelial Transcytosis in Acute Lung Injury: Emerging Mechanisms and Therapeutic Approaches. Front Physiol 2022; 13:828093. [PMID: 35431977 PMCID: PMC9008570 DOI: 10.3389/fphys.2022.828093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury (ALI) is characterized by widespread inflammation which in its severe form, Acute Respiratory Distress Syndrome (ARDS), leads to compromise in respiration causing hypoxemia and death in a substantial number of affected individuals. Loss of endothelial barrier integrity, pneumocyte necrosis, and circulating leukocyte recruitment into the injured lung are recognized mechanisms that contribute to the progression of ALI/ARDS. Additionally, damage to the pulmonary microvasculature by Gram-negative and positive bacteria or viruses (e.g., Escherichia coli, SARS-Cov-2) leads to increased protein and fluid permeability and interstitial edema, further impairing lung function. While most of the vascular leakage is attributed to loss of inter-endothelial junctional integrity, studies in animal models suggest that transendothelial transport of protein through caveolar vesicles, known as transcytosis, occurs in the early phase of ALI/ARDS. Here, we discuss the role of transcytosis in healthy and injured endothelium and highlight recent studies that have contributed to our understanding of the process during ALI/ARDS. We also cover potential approaches that utilize caveolar transport to deliver therapeutics to the lungs which may prevent further injury or improve recovery.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,*Correspondence: Richard D. Minshall,
| |
Collapse
|
11
|
Johar D, Elmehrath AO, Khalil RM, Elberry MH, Zaky S, Shalabi SA, Bernstein LH. Protein networks linking Warburg and reverse Warburg effects to cancer cell metabolism. Biofactors 2021; 47:713-728. [PMID: 34453457 DOI: 10.1002/biof.1768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
It was 80 years after the Otto Warburg discovery of aerobic glycolysis, a major hallmark in the understanding of cancer. The Warburg effect is the preference of cancer cell for glycolysis that produces lactate even when sufficient oxygen is provided. "reverse Warburg effect" refers to the interstitial tissue communications with adjacent epithelium, that in the process of carcinogenesis, is needed to be explored. Among these cell-cell communications, the contact between epithelial cells; between epithelial cells and matrix; and between fibroblasts and inflammatory cells in the underlying matrix. Cancer involves dysregulation of Warburg and reverse Warburg cellular metabolic pathways. How these gene and protein-based regulatory mechanisms have functioned has been the basis for this review. The importance of the Warburg in oxidative phosphorylation suppression, with increased glycolysis in cancer growth and proliferation is emphasized. Studies that are directed at pathways that would be expected to shift cell metabolism to an increased oxidation and to a decrease in glycolysis are emphasized. Key enzymes required for oxidative phosphorylation, and affect the inhibition of fatty acid metabolism and glutamine dependence are conferred. The findings are of special interest to cancer pharmacotherapy. Studies described in this review are concerned with the effects of therapeutic modalities that are intimately related to the Warburg effect. These interactions described may be helpful as adjuvant therapy in controlling the process of proliferation and metastasis.
Collapse
Affiliation(s)
- Dina Johar
- Department of Biochemistry and Nutrition, Faculty of Women for Arts, Sciences and Education, Ain Shams University, Heliopolis, Cairo, Egypt
| | | | - Rania M Khalil
- Department of Biochemistry, Pharmacy College, Delta University for Science and Technology, Gamasa, Egypt
| | - Mostafa H Elberry
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Samy Zaky
- Hepatogastroenterology and Infectious Diseases, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Samy A Shalabi
- Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Consultant Pathologist, Kuwait, Kuwait
| | - Larry H Bernstein
- Emeritus Prof. Department of Pathology, Yale University, Connecticut, USA
- Triplex Consulting Pharmaceuticals, 54 Firethorn Lane Northampton, MA 01060, USA
| |
Collapse
|
12
|
Zhu Y, Tian J, Peng X, Wang X, Yang N, Ying P, Wang H, Li B, Li Y, Zhang M, Cai Y, Lu Z, Niu S, Li Y, Zhong R, Chang J, Miao X. A genetic variant conferred high expression of CAV2 promotes pancreatic cancer progression and associates with poor prognosis. Eur J Cancer 2021; 151:94-105. [PMID: 33975060 DOI: 10.1016/j.ejca.2021.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
AIM This study aimed to identify the functional genes and genetic variants associated with the prognosis of pancreatic ductal adenocarcinoma (PDAC) and reveal the mechanism underlying their prognostic roles. METHODS First, we implement a two-stage exome-wide association study in a total of 1070 patients to identify the genetic variant correlated with PDAC prognosis. Then we performed fine mapping through bioinformatics analysis and dual-luciferase reporter assays to reveal the causal functional variant and prognostic gene. Next, we established the gene knockdown, knockout, and overexpression cell lines with small interfering RNA, CRISPR/Cas9, and lentivirus, respectively, and investigated the gene function on cell proliferation and migration in vivo and in vitro. Finally, we performed the RNA-seq to elucidate downstream genes and mechanisms altering PDAC prognosis. RESULTS We identified the CAV1-CAV2 locus tagged by rs8940 was significantly associated with PDAC prognosis, and rs10249656 in the 3'untranslated region of CAV2 was the real functional variant, which upregulated CAV2 expression through abolishing miR-548s binding. We observed upregulated CAV2 in PDAC and the higher expression correlated with worse prognosis. Transient knockdown of CAV2 inhibited PDAC migration without affecting proliferation rate. Knockout of CAV2 suppressed PDAC progression and metastasis, whereas stable overexpression of CAV2 promoted. Overexpressed CAV2 promoted the PDAC progression and metastasis via perturbing genes in the focal adhesion (CCND1, IGTA1, and ZYX) and extracellular matrix organisation (PLOD2, CAST, and ITGA1) pathways mechanically. CONCLUSION These findings shed light on an important role of CAV2 on PDAC progression and the prognostic impact of its genetic variation.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xiating Peng
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xiaoyang Wang
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Nan Yang
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Pingting Ying
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Haoxue Wang
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yue Li
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yimin Cai
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Siyuan Niu
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yao Li
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rong Zhong
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jiang Chang
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.
| |
Collapse
|
13
|
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: 27] [Impact Index Per Article: 6.8] [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
|
14
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Moghadam ER, Owrang M, Hashemi F, Makvandi P, Goharrizi MASB, Najafi M, Khan H. Lung cancer cells and their sensitivity/resistance to cisplatin chemotherapy: Role of microRNAs and upstream mediators. Cell Signal 2021; 78:109871. [PMID: 33279671 DOI: 10.1016/j.cellsig.2020.109871] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CP) is a well-known chemotherapeutic agent with excellent clinical effects. The anti-tumor activity of CP has been demonstrated in different cancers such as breast, cervical, reproductive, lung, brain, and prostate cancers. However, resistance of cancer cells to CP chemotherapy has led to its failure in eradication of cancer cells, and subsequent death of patients with cancer. Fortunately, much effort has been put to identify molecular pathways and mechanisms involved in CP resistance/sensitivity. It seems that microRNAs (miRs) are promising candidates in mediating CP resistance/sensitivity, since they participate in different biological aspects of cells such as proliferation, migration, angiogenesis, and differentiation. In this review, we focus on miRs and their regulation in CP chemotherapy of lung cancer, as the most malignant tumor worldwide. Oncogenic miRs trigger CP resistance in lung cancer cells via targeting various pathways such as Wnt/β-catenin, Rab6, CASP2, PTEN, and Apaf-1. In contrast, onco-suppressor miRs inhibit oncogene pathways such as STAT3 to suppress CP resistance. These topics are discussed to determine the role of miRs in CP resistance/sensitivity. We also describe the upstream modulators of miRs such as lncRNAs, circRNAs, NF-κB, SOX2 and TRIM65 and their association with CP resistance/sensitivity in lung cancer cells. Finally, the effect of anti-tumor plant-derived natural compounds on miR expression during CP sensitivity of lung cancer cells is discussed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Owrang
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
15
|
Abstract
Caveolae are bulb-like invaginations made up of two essential structural proteins, caveolin-1 and cavins, which are abundantly present at the plasma membrane of vertebrate cells. Since their discovery more than 60 years ago, the function of caveolae has been mired in controversy. The last decade has seen the characterization of new caveolae components and regulators together with the discovery of additional cellular functions that have shed new light on these enigmatic structures. Early on, caveolae and/or caveolin-1 have been involved in the regulation of several parameters associated with cancer progression such as cell migration, metastasis, angiogenesis, or cell growth. These studies have revealed that caveolin-1 and more recently cavin-1 have a dual role with either a negative or a positive effect on most of these parameters. The recent discovery that caveolae can act as mechanosensors has sparked an array of new studies that have addressed the mechanobiology of caveolae in various cellular functions. This review summarizes the current knowledge on caveolae and their role in cancer development through their activity in membrane tension buffering. We propose that the role of caveolae in cancer has to be revisited through their response to the mechanical forces encountered by cancer cells during tumor mass development.
Collapse
Affiliation(s)
- Vibha Singh
- UMR3666, INSERM U1143, Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, CNRS, 75005, Paris, France
| | - Christophe Lamaze
- UMR3666, INSERM U1143, Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, CNRS, 75005, Paris, France.
| |
Collapse
|
16
|
Chen P, Zhang YL, Xue B, Xu GY. Association of Caveolin-1 Expression With Prostate Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2021; 10:562774. [PMID: 33489874 PMCID: PMC7820696 DOI: 10.3389/fonc.2020.562774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/23/2020] [Indexed: 11/29/2022] Open
Abstract
Purpose The prognostic value of caveolin-1 in prostate cancer remains uncertain. Hence, this meta-analysis was performed to evaluate the prognostic value of caveolin-1 in prostate cancer, as well as ascertain the relationship between caveolin-1 expression and clinicopathological characteristics of prostate cancer patients. Methods The PubMed, Embase, Chinese National Knowledge Infrastructure and Chinese Biology Medicine databases were electronically searched to retrieve published studies on caveolin-1 expression in prostate cancer. After study selection and data extraction, the meta-analysis was conducted using Review manager 5.3 software. Odds ratio (OR) with 95% confidence interval (CI) was used to estimate the pooled effect. Funnel plot was used to assess publication bias. Results A total of ten studies were enrolled, which included 3976 cases of prostate cancer, 72 cases of high-grade intraepithelial neoplasia (HGPIN), and 157 normal controls. Results of the meta-analysis showed that the positive rate of caveolin-1 expression in prostate cancer was 18.28 times higher than that in normal control (OR= 18.28, 95% CI: 9.02–37.04, p<0.01), and 4.73 times higher than that in HGPIN (OR= 4.73, 95% CI: 2.38–9.42, p<0.01). The relationship between caveolin-1 and clinicopathological characteristics of prostate cancer showed that the differences in caveolin-1 expression in patients with prostate-specific antigen (PSA) >10 vs. ≤ 10 (OR=2.09, 95% CI: 1.35–3.22, p<0.01), differentiation degree low vs. medium/high (OR=2.74, 95% CI: 1.84–4.08, p<0.01), TNM stage T3+T4 vs. T1+T2 (OR=2.77, 95% CI: 1.78–4.29, p<0.01), and lymph node metastasis present vs. absent (OR=2.61, 95% CI: 1.84–3.69, p<0.01) were statistically significant. The correlation analysis between caveolin-1 and the survival time of patients with prostate cancer demonstrated that caveolin-1 was closely related to the prognosis of prostate cancer patients (HR=1.50, 95% CI: 1.28–1.76, p<0.01). Conclusion Caveolin-1 is overexpressed in prostate cancer, which can serve as a risk factor and adverse clinicopathological feature of prostate cancer. Caveolin-1 can also predict poor survival in prostate cancer patients after radical prostatectomy.
Collapse
Affiliation(s)
- Pei Chen
- Department of Basic Medicine, Jiangsu College of Nursing, Huai'an, China
| | - Yu-Ling Zhang
- Department of Basic Medicine, Jiangsu College of Nursing, Huai'an, China
| | - Bai Xue
- Department of Medical Technology, Jiangsu College of Nursing, Huai'an, China
| | - Guo-Ying Xu
- Department of Medical Technology, Jiangsu College of Nursing, Huai'an, China
| |
Collapse
|
17
|
Caveolin-1-mediated sphingolipid oncometabolism underlies a metabolic vulnerability of prostate cancer. Nat Commun 2020; 11:4279. [PMID: 32855410 PMCID: PMC7453025 DOI: 10.1038/s41467-020-17645-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Plasma and tumor caveolin-1 (Cav-1) are linked with disease progression in prostate cancer. Here we report that metabolomic profiling of longitudinal plasmas from a prospective cohort of 491 active surveillance (AS) participants indicates prominent elevations in plasma sphingolipids in AS progressors that, together with plasma Cav-1, yield a prognostic signature for disease progression. Mechanistic studies of the underlying tumor supportive onco-metabolism reveal coordinated activities through which Cav-1 enables rewiring of cancer cell lipid metabolism towards a program of 1) exogenous sphingolipid scavenging independent of cholesterol, 2) increased cancer cell catabolism of sphingomyelins to ceramide derivatives and 3) altered ceramide metabolism that results in increased glycosphingolipid synthesis and efflux of Cav-1-sphingolipid particles containing mitochondrial proteins and lipids. We also demonstrate, using a prostate cancer syngeneic RM-9 mouse model and established cell lines, that this Cav-1-sphingolipid program evidences a metabolic vulnerability that is targetable to induce lethal mitophagy as an anti-tumor therapy.
Collapse
|
18
|
Chen BW, He YC, Sung SY, Le TTH, Hsieh CL, Chen JY, Wei ZH, Yao DJ. Synthesis and characterization of magnetic nanoparticles coated with polystyrene sulfonic acid for biomedical applications. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2020; 21:471-481. [PMID: 32939172 PMCID: PMC7476547 DOI: 10.1080/14686996.2020.1790032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/28/2020] [Accepted: 06/28/2020] [Indexed: 05/31/2023]
Abstract
The development of novel magnetic nanoparticles (MNPs) with satisfactory biocompatibility for biomedical applications has been the subject of extensive exploration over the past two decades. In this work, we synthesized superparamagnetic iron oxide MNPs coated with polystyrene sulfonic acid (PSS-MNPs) and with a conventional co-precipitation method. The core size and hydrodynamic diameter of the PSS-MNPs were determined as 8-18 nm and 50-200 nm with a transmission electron microscopy and dynamic light scattering, respectively. The saturation magnetization of the particles was measured as 60 emu g-1 with a superconducting quantum-interference-device magnetometer. The PSS content in the PSS-MNPs was 17% of the entire PSS-MNPs according to thermogravimetric analysis. Fourier-transform infrared spectra were recorded to detect the presence of SO3 - groups, which confirmed a successful PSS coating. The structural properties of the PSS-MNPs, including the crystalline lattice, composition and phases, were characterized with an X-ray powder diffractometer and 3D nanometer-scale Raman microspectrometer. MTT assay and Prussian-blue staining showed that, although PSS-MNPs caused no cytotoxicity in both NIH-3T3 mouse fibroblasts and SK-HEP1 human liver-cancer cells up to 1000 μg mL-1, SK-HEP1 cells exhibited significantly greater uptake of PSS-MNPs than NIH-3T3 cells. The low cytotoxicity and high biocompatibility of PSS-MNPs in human cancer cells demonstrated in the present work might have prospective applications for drug delivery.
Collapse
Affiliation(s)
- Bo-Wei Chen
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu, Taiwan
| | - Yun-Chi He
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Shian-Ying Sung
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Trang Thi Huynh Le
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Hsieh
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jiann-Yeu Chen
- Center of Nanoscience and Nanotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Zung-Hang Wei
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu, Taiwan
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Da-Jeng Yao
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu, Taiwan
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
19
|
Philips BJ, Kumar A, Burki S, Ryan JP, Noda K, D'Cunha J. Triptolide-induced apoptosis in non-small cell lung cancer via a novel miR204-5p/Caveolin-1/Akt-mediated pathway. Oncotarget 2020; 11:2793-2806. [PMID: 32733649 PMCID: PMC7367654 DOI: 10.18632/oncotarget.27672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is one of the most prevalent malignancies world-wide with non-small cell lung cancer (NSCLC) comprising nearly 80% of all cases. Unfortunately, many lung cancer patients are diagnosed at advanced stages of the disease with an associated poor prognosis. Recently, the Chinese herb root extract Triptolide/Minnelide (TL) has shown significant promise as a therapeutic agent for NSCLC treatment both in vitro and in vivo. The aim of this study was to investigate the underlying mechanism(s) of action regarding TL-induced cytotoxicity in NSCLC. We demonstrate that triptolide treatment of A549 and H460 NSCLC cells decreases Caveolin-1 (CAV-1) mRNA/protein expression, resulting in activation of the Akt/Bcl-2-mediated mitochondrial apoptosis pathway. CAV-1 down-regulation was triggered by Micro-RNA 204-5p (miR204-5p) up-regulation and could be significantly blocked by pre-treatment with both Sirt-1/Sirt-3 specific siRNA and SIRT-1/SIRT-3 enzyme inhibitors, EX-527 and nicotinamide. Overall, our results provide evidence for a novel mechanism by which TL exerts its cytotoxic effects on NSCLC via CAV-1 down-regulation. Furthermore, these findings demonstrate a pivotal role for TL induction of the Akt/Bax pathway in apoptosis of human lung cancer.
Collapse
Affiliation(s)
- Brian J Philips
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ajay Kumar
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah Burki
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - John P Ryan
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kentaro Noda
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
20
|
Deng X, Chen Y, Liu Z, Xu J. MiR-124-3p.1 Sensitizes Ovarian Cancer Cells to Mitochondrial Apoptosis Induced by Carboplatin. Onco Targets Ther 2020; 13:5375-5386. [PMID: 32606755 PMCID: PMC7294572 DOI: 10.2147/ott.s242342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/09/2020] [Indexed: 12/16/2022] Open
Abstract
Background Carboplatin is a platinum-based chemotherapeutic drug that is commonly used as a treatment for ovarian cancer. However, high doses and repeated use of carboplatin usually reduce the sensitivity of cancer cells to the drug. There is an urgent need to develop strategies to increase the sensitivity of ovarian cancer cells to carboplatin. Materials and Methods Quantitative reverse-transcriptase real-time PCR was used to detect miR-124-3p.1 levels in ovarian cancer tissues and cell lines. Transfection with miR-124-3p.1 and caveolin-1 (CAV1) was used for gain-of-function experiments. Western blot and immunoprecipitation assays were performed to evaluate the expression and function of CAV1, AKT, Bad, and Bcl-xl. Flow cytometry analysis was used to measure the apoptosis rates of SKOV3 and A2780 cells. Results Expression levels of miR-124-3p.1 were decreased in ovarian cancer tissues and cell lines. Furthermore, overexpression of miR-124-3p.1 enhanced carboplatin-induced apoptotic cell death of ovarian cancer cell lines. Regarding the mechanism of this effect, we showed that CAV1 was the target of miR-124-3p.1 in ovarian cancer. Overexpression of miR-124-3p.1 suppressed the expression of CAV1, thereby reducing the activation of AKT and phosphorylation of Bad. As a result, the function of Bcl-xl was inhibited and carboplatin-induced mitochondrial apoptosis was enhanced. Conclusion miR-124-3p.1 sensitizes carboplatin-induced mitochondrial apoptosis through suppression of CAV1 in ovarian cancer. Increasing miR-124-3p.1 expression may represent a novel strategy to improve carboplatin sensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Xiaohong Deng
- Department of Gynecology, Northwest Women and Children's Hospital, Xi'an City, Shanxi Province 710061, People's Republic of China
| | - Yi Chen
- Department of Surgery, Affiliated Hospital of Xi'an Jiao Tong University, Chang'an District Hospital, Xi'an City, Shanxi Province 710119, People's Republic of China
| | - Zhao Liu
- Department of Surgery, Xi'an Chest Hospital, Xi'an TB and Thoracic Tumor Hospital, Xi'an City, Shanxi Province 710100, People's Republic of China
| | - Jingning Xu
- Department of Obstetrics and Gynecology, Northwest Women and Children's Hospital, Xi'an City, Shanxi Province 710061, People's Republic of China
| |
Collapse
|
21
|
Liu S, Wang W, Zhao Y, Liang K, Huang Y. Identification of Potential Key Genes for Pathogenesis and Prognosis in Prostate Cancer by Integrated Analysis of Gene Expression Profiles and the Cancer Genome Atlas. Front Oncol 2020; 10:809. [PMID: 32547947 PMCID: PMC7277826 DOI: 10.3389/fonc.2020.00809] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Prostate cancer (PCa)is a malignancy of the urinary system with a high incidence, which is the second most common male cancer in the world. There are still huge challenges in the treatment of prostate cancer. It is urgent to screen out potential key biomarkers for the pathogenesis and prognosis of PCa. Methods: Multiple gene differential expression profile datasets of PCa tissues and normal prostate tissues were integrated analysis by R software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the overlapping Differentially Expressed Genes (DEG) were performed. The STRING online database was used in conjunction with Cytospace software for protein-protein interaction (PPI) network analysis to define hub genes. The relative mRNA expression of hub genes was detected in Gene Expression Profiling Interactive Analysis (GEPIA) database. A prognostic gene signature was identified by Univariate and multivariate Cox regression analysis. Results: Three hundred twelve up-regulated genes and 85 down-regulated genes were identified from three gene expression profiles (GSE69223, GSE3325, GSE55945) and The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) dataset. Seven hub genes (FGF2, FLNA, FLNC, VCL, CAV1, ACTC1, and MYLK) further were detected, which related to the pathogenesis of PCa. Seven prognostic genes (BCO1, BAIAP2L2, C7, AP000844.2, ASB9, MKI67P1, and TMEM272) were screened to construct a prognostic gene signature, which shows good predictive power for survival by the ROC curve analysis. Conclusions: We identified a robust set of new potential key genes in PCa, which would provide reliable biomarkers for early diagnosis and prognosis and would promote molecular targeting therapy for PCa.
Collapse
Affiliation(s)
- Shuang Liu
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China
| | | | - Yan Zhao
- Xuzhou Central Hospital, Xuzhou, China
| | - Kaige Liang
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China
| | - Yaojiang Huang
- Beijing Engineering Research Center of Food Environment and Public Health, Minzu University of China, Beijing, China.,Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
22
|
MiR-204 reduces cisplatin resistance in non-small cell lung cancer through suppression of the caveolin-1/AKT/Bad pathway. Aging (Albany NY) 2020; 11:2138-2150. [PMID: 30981205 PMCID: PMC6503872 DOI: 10.18632/aging.101907] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/03/2019] [Indexed: 12/17/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common and lethal human malignant tumor worldwide. Platinum-based chemotherapy is still the mainstay of treatment for NSCLC. However, long-term chemotherapy usually induces serious drug resistance in NSCLC cells. Accordingly, treatment strategies that reverse the resistance of NSCLC cells against platinum-based drugs may have considerable clinical value. In the present study, we observed significant upregulation of CAV-1 expression and a significant decrease of miR-204 expression in cisplatin-resistant A549 (CR-A549) and cisplatin-resistant PC9 (CR-PC9) cells compared to their parental A549 and PC9 cells. Furthermore, we demonstrated that the downregulation of miR-204 expression was responsible for CAV-1 overexpression in these cisplatin-resistant NSCLC cells. We then found that enforced expression of miR-204 can resensitize CR-A549 and CR-PC9 cells to cisplatin-induced mitochondrial apoptosis through suppression of the caveolin-1/AKT/Bad pathway. We demonstrated that dysregulation of miR-204/caveolin-1 axis is an important mechanism for NSCLC cells to develop the chemoresistance.
Collapse
|
23
|
Jaiprasart P, Dogra S, Neelakantan D, Devapatla B, Woo S. Identification of signature genes associated with therapeutic resistance to anti-VEGF therapy. Oncotarget 2020; 11:99-114. [PMID: 32002127 PMCID: PMC6967771 DOI: 10.18632/oncotarget.27307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/04/2019] [Indexed: 12/31/2022] Open
Abstract
VEGF-mediated tumor angiogenesis is a validated clinical target in many cancers, but modest efficacy and rapid development of resistance are major challenges of VEGF-targeted therapies. To establish a molecular signature of this resistance in ovarian cancer, we developed preclinical tumor models of adaptive resistance to chronic anti-VEGF treatment. We performed RNA-seq analysis and reverse-phase protein array to compare changes in gene and protein expressions in stroma and cancer cells from resistant and responsive tumors. We identified a unique set of stromal-specific genes that were strongly correlated with resistance phenotypes against two different anti-VEGF treatments, and selected the apelin/APJ signaling pathway for further in vitro validation. Using various functional assays, we showed that activation of apelin/APJ signaling reduces the efficacy of a VEGF inhibitor in endothelial cells. In patients with ovarian cancer treated with bevacizumab, increased expression of apelin was associated with significantly decreased disease-free survival. These findings link signature gene expressions with anti-VEGF response, and may thus provide novel targetable mechanisms of clinical resistance to anti-VEGF therapies.
Collapse
Affiliation(s)
- Pharavee Jaiprasart
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Samrita Dogra
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Deepika Neelakantan
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Bharat Devapatla
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Gynecologic Cancers Research Program, Peggy and Charles Stephenson Cancer Center, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
24
|
Gong Y, Yang Y, Tian S, Chen H. Different Role of Caveolin-1 Gene in the Progression of Gynecological Tumors. Asian Pac J Cancer Prev 2019; 20:3259-3268. [PMID: 31759347 PMCID: PMC7062999 DOI: 10.31557/apjcp.2019.20.11.3259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Indexed: 12/13/2022] Open
Abstract
Caveolin-1 (Cav-1), an integral membrane protein, is a principal component of caveolae and has been reported to play a promoting or inhibiting role in cancer progression. Gynecologic tumor is a group of tumors that affect the tissue and organs of the female reproductive system, especially cervical cancer. Cervical cancer, as one of the most common cancers, severely affects female health in developing countries in particular because of its high morbidity and mortality. This review summarizes some mechanisms of Cav-1 in the development and progression of gynecological tumors. The role of Cav-1 in tumorigenesis, including dysregulation of cell cycle, apoptosis and autophagy, adhesion, invasion, and metastasis, such as the formation of invadopodia and matrix metalloproteinase degradation are presented in detail. In addition, Cav-1 modulates autophagy and the formation of invadopodia and target regulated by miRNAs to affect tumor progress. Taken together, we find that, no matter Cav-1 expression in the tumor or stromal cells , Cav-1 has paradoxical role in different types of gynecological tumors in vivo or in vitro and even in the same tumor from the same organ.
Collapse
Affiliation(s)
- Yan Gong
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| | - Yuhan Yang
- Department of Pathology, School of Basic Medical Science, Wuhan University, Wuhan, P. R. China
| | - Sufang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| | - Honglei Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, P. R. China
| |
Collapse
|
25
|
Chen P, Feng Y, Zhang H, Shi X, Li B, Ju W, Yu X, Zhang N, Luo X. MicroRNA‑192 inhibits cell proliferation and induces apoptosis in human breast cancer by targeting caveolin 1. Oncol Rep 2019; 42:1667-1676. [PMID: 31485620 PMCID: PMC6775803 DOI: 10.3892/or.2019.7298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/25/2019] [Indexed: 12/11/2022] Open
Abstract
It has been demonstrated that microRNA-192 (miR-192) serves important roles in different cancer types, including breast cancer, prostate cancer and colorectal cancer. However, its biological role and function in breast cancer remains largely unknown. The present study aimed to determine the role of miR-192 in breast cancer. In the present study, one normal breast and two breast tumor cells lines were used, which included the normal mammary fibroblast cell line Hs578Bst, a more aggressive breast tumor cell line MDA-MB-231 and a less aggressive breast tumor cell line MCF-7. The effect of miR-192 on proliferation of breast cancer cells was detected with an MTT assay. Western blot analysis was performed to determine protein expression of caveolin 1 (CAV1). A lentiviral vector that overexpresses pre-miR-192 and control lentiviral packaging plasmids were used in the present study. The Student's t-test was performed to analyze the significance of differences between samples. In the present study, it was determined that the expression of miR-192 is downregulated in breast cancer, compared with the adjacent normal tissues. Overexpression of miR-192 significantly inhibited cell proliferation, and induced cell apoptosis and cell cycle arrest in MCF7 and MDA-MB-231 cells. Using a bioinformatics method, CAV1 was considered a potential target of miR-192. Furthermore, it was demonstrated that CAV1 is a direct target of miR-192 and its protein expression is negatively regulated by miR-192. Therefore, the present study demonstrated that miR-192 serves an important role as a regulator in breast cancer and the miR-192/CAV1 axis has a potential as a therapeutic target for treatment of breast cancer.
Collapse
Affiliation(s)
- Pinjia Chen
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Yun Feng
- Surgical Department, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Haige Zhang
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Xinpeng Shi
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Bin Li
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Wencui Ju
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Xiurong Yu
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Nan Zhang
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Xiaoyong Luo
- Department of Oncology, The Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang, Henan 471000, P.R. China
| |
Collapse
|
26
|
Okada S, Raja SA, Okerblom J, Boddu A, Horikawa Y, Ray S, Okada H, Kawamura I, Murofushi Y, Murray F, Patel HH. Deletion of caveolin scaffolding domain alters cancer cell migration. Cell Cycle 2019; 18:1268-1280. [PMID: 31116089 DOI: 10.1080/15384101.2019.1618118] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Caveolin-1 (Cav-1) is an integral membrane protein that plays an important role in proliferative and terminally differentiated cells. As a structural component of Caveolae, Cav-1 interacts with signaling molecules via a caveolin scaffolding domain (CSD) regulating cell signaling. Recent reports have shown that Cav-1 is a negative regulator in tumor metastasis. Therefore, we hypothesize that Cav-1 inhibits cell migration through its CSD. HeLa cells were engineered to overexpress Cav-1 (Cav-1 OE), Cav-1 without a functional CSD (∆CSD), or enhanced green fluorescent protein (EGFP) as a control. HeLa cell migration was suppressed in Cav-1 OE cells while ∆CSD showed increased migration, which corresponded to a decrease in the tight junction protein, zonula occludens (ZO-1). The migration phenotype was confirmed in multiple cancer cell lines. Phosphorylated STAT-3 was decreased in Cav-1 OE cells compared to control and ∆CSD cells; reducing STAT-3 expression alone decreased cell migration. ∆CSD blunted HeLa proliferation by increasing the number of cells in the G2/M phase of the cell cycle. Overexpressing the CSD peptide alone suppressed HeLa cell migration and inhibited pSTAT3. These findings suggest that Cav-1 CSD may be critical in controlling the dynamic phenotype of cancer cells by facilitating the interaction of specific signal transduction pathways, regulating STAT3 and participating in a G2/M checkpoint. Modulating the CSD and targeting specific proteins may offer potential new therapies in the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Sunaho Okada
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| | - Sadaf A Raja
- c Department of Biosciences , COMSATS Institute of Information Technology , Islamabad , Pakistan
| | - Jonathan Okerblom
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| | - Aayush Boddu
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| | - Yousuke Horikawa
- d Department of Pediatrics , Sharp Rees-Stealy Medical Group , San Diego , CA , USA.,e Department of Anesthesiology , Tokushima University , Tokushima , Japan
| | | | - Hideshi Okada
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,g Department of Anesthesiology and Medicine , UCSD School of Medicine , San Diego , CA , USA.,h Department of Emergency and Disaster Medicine , Gifu University Graduate School of Medicine , Gifu , Japan
| | - Itta Kawamura
- i Department of Cardiovascular Medicine , Gifu Heart Center , Gifu , Japan
| | - Yoshiteru Murofushi
- g Department of Anesthesiology and Medicine , UCSD School of Medicine , San Diego , CA , USA
| | - Fiona Murray
- j Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences , University of Aberdeen , Aberdeen , Scotland
| | - Hemal H Patel
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| |
Collapse
|
27
|
Codenotti S, Faggi F, Ronca R, Chiodelli P, Grillo E, Guescini M, Megiorni F, Marampon F, Fanzani A. Caveolin-1 enhances metastasis formation in a human model of embryonal rhabdomyosarcoma through Erk signaling cooperation. Cancer Lett 2019; 449:135-144. [DOI: 10.1016/j.canlet.2019.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/08/2019] [Accepted: 02/10/2019] [Indexed: 11/15/2022]
|
28
|
Gao Y, Li L, Li T, Ma L, Yuan M, Sun W, Cheng HL, Niu L, Du Z, Quan Z, Fan Y, Fan J, Luo C, Wu X. Simvastatin delays castration‑resistant prostate cancer metastasis and androgen receptor antagonist resistance by regulating the expression of caveolin‑1. Int J Oncol 2019; 54:2054-2068. [PMID: 31081050 PMCID: PMC6521936 DOI: 10.3892/ijo.2019.4774] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/22/2019] [Indexed: 12/19/2022] Open
Abstract
The failure of androgen deprivation therapy in prostate cancer treatment mainly results from drug resistance to androgen receptor antagonists. Although an aberrant caveolin‑1 (Cav‑1) expression has been reported in multiple tumor cell lines, it is unknown whether it is responsible for the progression of castration‑resistant prostate cancer (CRPC). Thus, the aim of the present study was to determine whether Cav‑1 can be used as a key molecule for the prevention and treatment of CRPC, and to explore its mechanism of action in CRPC. For this purpose, tissue and serum samples from patients with primary prostate cancer and CRPC were analyzed using immunohistochemistry and enzyme‑linked immunosorbent assay, which revealed that Cav‑1 was overexpressed in CRPC. Furthermore, Kaplan‑Meier survival analysis and univariate Cox proportional hazards regression analysis demonstrated that Cav‑1 expression in tumors was an independent risk factor for the occurrence of CRPC and was associated with a shorter recurrence‑free survival time in patients with CRPC. Receiver operating characteristic curves suggested that serum Cav‑1 could be used as a diagnostic biomarker for CRPC (area under the curve, 0.876) using a cut‑off value of 0.68 ng/ml (with a sensitivity of 82.1% and specificity of 80%). In addition, it was determined that Cav‑1 induced the invasion and migration of CRPC cells by the activation of the H‑Ras/phosphoinositide‑specific phospholipase Cε signaling cascade in the cell membrane caveolae. Importantly, simvastatin was able to augment the anticancer effects of androgen receptor antagonists by downregulating the expression of Cav‑1. Collectively, the findings of this study provide evidence that Cav‑1 is a promising predictive biomarker for CRPC and that lowering cholesterol levels with simvastatin or interfering with the expression of Cav‑1 may prove to be a useful strategy with which to prevent and/or treat CRPC.
Collapse
Affiliation(s)
- Yingying Gao
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Luo Li
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Ting Li
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Lei Ma
- Department of Laboratory Diagnosis, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154000, P.R. China
| | - Mengjuan Yuan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 408000, P.R. China
| | - Wei Sun
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 408000, P.R. China
| | - Hong Lin Cheng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 408000, P.R. China
| | - Lingfang Niu
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Zhongbo Du
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 408000, P.R. China
| | - Zhen Quan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 408000, P.R. China
| | - Yanru Fan
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Jiaxin Fan
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Chunli Luo
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 408000, P.R. China
| | - Xiaohou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 408000, P.R. China
| |
Collapse
|
29
|
Liu R, Kong Y, Sun P, Li F, Shi X. Correlation between methylation of the caveolin‐1 gene and of caveolin‐1 messenger ribonucleic acid, and protein levels and human epidermal growth factor receptor 2 protein expression in adenocarcinomas of the esophagogastric junction. PRECISION RADIATION ONCOLOGY 2019. [DOI: 10.1002/pro6.61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Ruizhen Liu
- The First People's Hospital of Wu'an Wu'an Hebei China
| | - Yi Kong
- The First People's Hospital of Wu'an Wu'an Hebei China
| | - Pengbo Sun
- The First People's Hospital of Wu'an Wu'an Hebei China
| | - Faliang Li
- The First People's Hospital of Wu'an Wu'an Hebei China
| | - Xiaopeng Shi
- The First People's Hospital of Wu'an Wu'an Hebei China
| |
Collapse
|
30
|
Dai X, Zhou X, Liao C, Yao Y, Yu Y, Zhang S. A nanodrug to combat cisplatin-resistance by protecting cisplatin with p-sulfonatocalix[4]arene and regulating glutathione S-transferases with loaded 5-fluorouracil. Chem Commun (Camb) 2019; 55:7199-7202. [DOI: 10.1039/c9cc03012c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A nanodrug that can effectively combat cisplatin-resistant A549/CDDP cells was developed by protecting cisplatin from glutathione (GSH) detoxification trough a host–guest interaction between cisplatin and p-sulfonatocalix[4]arene.
Collapse
Affiliation(s)
- Xin Dai
- National Engineering Research Centre for Biomaterials, and College of Chemistry
- Sichuan University
- Chengdu 610064
- China
- Zunyi Medical and Pharmaceutical College
| | - Xueying Zhou
- National Engineering Research Centre for Biomaterials, and College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Chunyan Liao
- National Engineering Research Centre for Biomaterials, and College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Yongchao Yao
- National Engineering Research Centre for Biomaterials, and College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Yunlong Yu
- National Engineering Research Centre for Biomaterials, and College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| | - Shiyong Zhang
- National Engineering Research Centre for Biomaterials, and College of Chemistry
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
31
|
Huang Q, Zhong W, Hu Z, Tang X. A review of the role of cav-1 in neuropathology and neural recovery after ischemic stroke. J Neuroinflammation 2018; 15:348. [PMID: 30572925 PMCID: PMC6302517 DOI: 10.1186/s12974-018-1387-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke starts a series of pathophysiological processes that cause brain injury. Caveolin-1 (cav-1) is an integrated protein and locates at the caveolar membrane. It has been demonstrated that cav-1 can protect blood–brain barrier (BBB) integrity by inhibiting matrix metalloproteases (MMPs) which degrade tight junction proteins. This article reviews recent developments in understanding the mechanisms underlying BBB dysfunction, neuroinflammation, and oxidative stress after ischemic stroke, and focuses on how cav-1 modulates a series of activities after ischemic stroke. In general, cav-1 reduces BBB permeability mainly by downregulating MMP9, reduces neuroinflammation through influencing cytokines and inflammatory cells, promotes nerve regeneration and angiogenesis via cav-1/VEGF pathway, reduces apoptosis, and reduces the damage mediated by oxidative stress. In addition, we also summarize some experimental results that are contrary to the above and explore possible reasons for these differences.
Collapse
Affiliation(s)
- Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China.
| |
Collapse
|
32
|
Ren J, Li X, Sun G, Li S, Liang S, Li Z, Li B, Xia M. Protective effect of leptin-mediated caveolin-1 expression on neurons after spinal cord injury. Cell Calcium 2018; 76:122-128. [PMID: 30469142 DOI: 10.1016/j.ceca.2018.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/18/2018] [Accepted: 11/12/2018] [Indexed: 10/27/2022]
Abstract
Spinal cord injury (SCI) causes long-term disability and has no effective clinical treatment. After SCI, extracellular adenosine triphosphate (ATP) leads to an influx of extracellular Ca2+, and this Ca2+ overload causes neuronal toxicosis and apoptosis. The biological functions of leptin have been widely investigated in the central nervous system. In this study, we discovered that the administration of leptin could improve locomotor recovery following SCI. The aim of this study was to determine the neuroprotective mechanism of leptin in vivo and in vitro. The neuronal apoptosis and Ca2+ imaging signal induced by ATP were suppressed by leptin, due to elevated caveolin-1 expression. In vivo two-photon observations revealed that leptin reduced the neuronal Ca2+ imaging signal in the exposed spinal cords of live Thy1-YFP mice. In conclusion, leptin promotes locomotor functional recovery and suppresses neuronal impairment after SCI, suggesting that leptin has a promising clinical therapeutic value for treatment of SCI.
Collapse
Affiliation(s)
- Jiaan Ren
- Department of Orthopaedics, First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | - Xiaowei Li
- Laboratory Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Guangfeng Sun
- Department of Orthopaedics, First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | - Shuai Li
- Laboratory Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Shanshan Liang
- Laboratory Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Zexiong Li
- Laboratory Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Baoman Li
- Laboratory Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Maosheng Xia
- Department of Orthopaedics, First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
33
|
Kamibeppu T, Yamasaki K, Nakahara K, Nagai T, Terada N, Tsukino H, Mukai S, Kamoto T. Caveolin-1 and -2 regulate cell motility in castration-resistant prostate cancer. Res Rep Urol 2018; 10:135-144. [PMID: 30324095 PMCID: PMC6174915 DOI: 10.2147/rru.s173377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Caveolin (Cav)-1 and Cav-2 are cell membrane proteins, which are structural proteins of caveolae and are reported to be positive regulators of cell survival and metastasis in prostate cancer (PC). In a previous study, we reported that elevated levels of Cav-1 and Cav-2 were significantly associated with PC progression. However, their functions in PC have not yet been clarified. In this study, we examined the function of Cav-1 and Cav-2 in PC cell invasiveness and motility. Materials and methods We introduced Cav-1- and Cav-2-specific small interfering into PC3 cells to knock-down (KD) both molecules. We also performed cell proliferation assay, wound healing assay, migration assay, and invasion assay using PC3 cells and compared the results between Cav-1-KD, Cav-2-KD, and negative control PC3 cells. In addition, we performed real-time quantitative PCR (RT-qPCR) and RT2 Profiler PCR Array analysis to identify factors influencing migration. Results We observed no significant difference in the proliferative and invasive activities of Cav-1-KD and Cav-2-KD PC3 cells; however, the cell motility was significantly decreased compared with negative control PC3 cells. RT-qPCR revealed that the expression of vimentin and N-cadherin was downregulated in Cav-1-KD PC3 cells. In addition, PCR array revealed a decreased expression of MGAT5, MMP13, and MYCL in Cav-1-KD PC3 and ETV4, FGFR4, and SRC in Cav-2-KD PC3. Conclusion Cav-1 and Cav-2 may positively contribute to the upregulation of castration-resistant PC cell migration. Cav-induced regulation of several molecules including vimentin, N-cadherin, MGAT5, MMP13, MYCL, ETV4, FGFR4, and SRC may have an important role in PC3 cell motility. However, further examination will be required.
Collapse
Affiliation(s)
- Toyoharu Kamibeppu
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Koji Yamasaki
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Kozue Nakahara
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Takahiro Nagai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Naoki Terada
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Hiromasa Tsukino
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Shoichiro Mukai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| | - Toshiyuki Kamoto
- Department of Urology, Faculty of Medicine, University of Miyazaki, Kiyotake, Japan,
| |
Collapse
|
34
|
Guerrero S, Díaz-García VM, Contreras-Orellana P, Lara P, Palma S, Guzman F, Lobos-Gonzalez L, Cárdenas A, Rojas-Silva X, Muñoz L, Leyton L, Kogan MJ, Quest AF. Gold nanoparticles as tracking devices to shed light on the role of caveolin-1 in early stages of melanoma metastasis. Nanomedicine (Lond) 2018; 13:1447-1462. [PMID: 29972676 DOI: 10.2217/nnm-2017-0390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM To track early events during lung metastasis, we labeled cells expressing (B16F10CAV1) or lacking CAV1 (B16F10mock) with gold nanoparticles conjugated to the peptide TAT (AuNPs-PEG-TAT). METHODS B16F10 expressing or lacking CAV1 were labeled with AuNPs-PEG-TAT. The physicochemical properties and cytotoxicity of these nanoparticles, as well as their effects on migration and invasiveness of B16F10 cells in vitro were evaluated. Ex vivo lung distribution of the labeled cells after tail vein injection into C57BL/6 mice was examined. RESULTS AuNPs-PEG-TAT did not affect B16F10 viability, migration and invasiveness. The metastatic and tumorigenic capability of the labeled B16F10 was also not modified in comparison to unlabeled B16F10 cells. CAV1 expression favored the retention of B16F10 cells in the lungs of mice 2 h post injection, suggesting CAV1 promoted adherence to endothelial cells and transendothelial migration. CONCLUSIONS We developed a protocol to label B16F10 cells with AuNPs-PEG-TAT that permits subsequent tracking of cells in mice. CAV1 overexpression was found to increase retention and transendothelial migration of B16F10 cells in the lung.
Collapse
Affiliation(s)
- Simón Guerrero
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Victor Manuel Díaz-García
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Lientur 1457, Concepción 4080871, Chile
| | - Pamela Contreras-Orellana
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Pablo Lara
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Sujey Palma
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Fanny Guzman
- Núcleo de Biotecnología Curauma (NBC), Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Valparaíso, Chile
| | - Lorena Lobos-Gonzalez
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Avenida Las Condes 12.438, Lo Barnechea Santiago, Chile
| | - Areli Cárdenas
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Escuela de Obstetricia y Puericultura, Facultad de Salud, Universidad Bernardo OHiggins, Avenida Viel 1497, Santiago, Chile
| | - Ximena Rojas-Silva
- Laboratorio de Análisis por Activación Neutrónica, Comisión Chilena de Energía Nuclear (CChEN), Nueva Bilbao 12501, Santiago, Chile
| | - Luis Muñoz
- Laboratorio de Análisis por Activación Neutrónica, Comisión Chilena de Energía Nuclear (CChEN), Nueva Bilbao 12501, Santiago, Chile
| | - Lisette Leyton
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Andrew Fg Quest
- Laboratory of Cellular Communication, Program of Cell & Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.,Center for Studies on Exercise Metabolism & Cancer (CEMC), University of Chile, Av. Independencia 1027, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| |
Collapse
|
35
|
Li M, Chen H, Diao L, Zhang Y, Xia C, Yang F. Caveolin-1 and VEGF-C promote Lymph Node Metastasis in the Absence of Intratumoral Lymphangiogenesis in Non-small Cell Lung Cancer. TUMORI JOURNAL 2018; 96:734-43. [DOI: 10.1177/030089161009600516] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aims and background Caveolin-1 is a key component of membrane caveolae which plays an important role in cell transformation, cell migration, metastasis and angiogenesis. The mechanism of caveolin-1 and VEGF-C in lymphatic metastasis of non-small cell lung cancer (NSCLC) is still unclear. This study aimed to define the caveolin-1 and VEGF-C expression and lymph vessel density in NSCLC and look for correlations with clinicopathological features in NSCLC. Methods Caveolin-1, VEGF-C, and D2–40 protein expression were assessed by immunohistochemistry in a tissue microarray constructed from 70 NSCLCs and 12 normal lungs. Results Caveolin-1 expression was detected in 31 of 70 (44.3%) NSCLCs, which was significantly lower than its expression in normal lungs (9 of 12, 75%; P = 0.049). Expression of VEGF-C was detected in 49 of 70 (70%) NSCLCs and 4 of 12 (33.3%) normal lungs (P = 0.022). Both caveolin-1 and VEGF-C expression were correlated with lymph node metastasis of NSCLC (P = 0.001; P = 0.028). Moreover, caveolin-1 expression was correlated with tumor stage, histological type, and differentiation grade (P = 0.012; P = 0.038; P = 0.002). VEGF-C expression was correlated only with histological type (P = 0.020). There was no correlation between intratumoral lymph vessel density and any clinicopathological parameters including lymph node status. Furthermore, there was no correlation between caveolin-1 expression, VEGF-C expression, and lymph vessel density. Conclusions These findings indicated a reduction of caveolin-1 expression in NSCLC and suggested that caveolin-1 as well as VEGF-C might be involved in lymph node metastasis of NSCLC. The role of caveolin-1 in lymphatic metastasis and intratumoral lymphangiogenesis in NSCLC needs further study. Free full text available at www.tumorionline.it
Collapse
Affiliation(s)
- Minhua Li
- Department of Pathology, School of Basic Medicine Science, Wuhan University, Wuhan
- Department of Pathology, Shaoxing People's Hospital & The First Affiliated Hospital of Shaoxing University, Shaoxing
| | - Honglei Chen
- Department of Pathology, School of Basic Medicine Science, Wuhan University, Wuhan
| | - Luming Diao
- Department of Pathology, School of Basic Medicine Science, Wuhan University, Wuhan
| | - Yuxia Zhang
- Department of Pathology, School of Basic Medicine Science, Wuhan University, Wuhan
| | - Cong Xia
- Medical school, Jianghan University, Wuhan, China
| | - Fei Yang
- Department of Pathology, School of Basic Medicine Science, Wuhan University, Wuhan
| |
Collapse
|
36
|
Xu H, Zhang L, Chen W, Xu J, Zhang R, Liu R, Zhou L, Hu W, Ju R, Lee C, Lu W, Kumar A, Li X, Tang Z. Inhibitory effect of caveolin-1 in vascular endothelial cells, pericytes and smooth muscle cells. Oncotarget 2017; 8:76165-76173. [PMID: 29100301 PMCID: PMC5652695 DOI: 10.18632/oncotarget.19191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 06/19/2017] [Indexed: 11/25/2022] Open
Abstract
Caveolin-1 (Cav1) is the principle structural protein of caveolae. It plays important roles in the vascular system under both physiological and pathological conditions. Although Cav1 has been shown to inhibit microvascular permeability and has been considered as a tumor-suppressor for years, the underlying cellular mechanism has yet to be discovered. Here, we systematically investigated Cav1 functions in the main types of vascular cells, including endothelial cells (ECs), pericytes (PCs) and smooth muscle cells (SMCs). We synthesized a cell-permeable peptide called cavtratin that is derived from the Cav1 scaffolding domain. We found that cavtratin inhibited ECs in all assays, including survival, proliferation, migration and permeability assays. It also inhibited the proliferation of PCs and SMCs but had no effect on their survival or migration. The inhibitory effect of cavtratin on the proliferation of all vascular cells suggests that Cav1 plays important roles in vascular development and angiogenesis. Under physiological condition, the main function of Cav1 is to inhibit EC permeability.
Collapse
Affiliation(s)
- Hongping Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Liwei Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Jiazhou Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Ruting Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Ran Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Lan Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Wenjie Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Weisi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Zhongshu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, P. R. China
| |
Collapse
|
37
|
Gao C, Wang Q, Chung SK, Shen J. Crosstalk of metabolic factors and neurogenic signaling in adult neurogenesis: Implication of metabolic regulation for mental and neurological diseases. Neurochem Int 2017; 106:24-36. [DOI: 10.1016/j.neuint.2017.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/10/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022]
|
38
|
The metastatic suppressor NDRG1 inhibits EMT, migration and invasion through interaction and promotion of caveolin-1 ubiquitylation in human colorectal cancer cells. Oncogene 2017; 36:4323-4335. [PMID: 28346422 PMCID: PMC5537633 DOI: 10.1038/onc.2017.74] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 01/15/2017] [Accepted: 02/14/2017] [Indexed: 02/07/2023]
Abstract
N-myc downstream-regulated gene 1 (NDRG1) has been reported to act as a key regulatory molecule in tumor progression-related signaling pathways, especially in tumor metastasis. However, the related mechanism has not been fully discovered yet. Herein we demonstrated that the novel molecule of cell migration and invasion, caveolin-1, has direct interaction with NDRG1 in human colorectal cancer (CRC) cells. Moreover, we discovered that NDRG1 reduces caveolin-1 protein expression through promoting its ubiquitylation and subsequent degradation via the proteasome in CRC cells. In addition, caveolin-1 mediates the suppressive function of NDRG1 in epithelial–mesenchymal transition, migration and invasion in vitro and metastasis in vivo. These results help to fulfill the potential mechanisms of NDRG1 in anti-metastatic treatment for human colorectal cancer.
Collapse
|
39
|
Spencer A, Yu L, Guili V, Reynaud F, Ding Y, Ma J, Jullien J, Koubi D, Gauthier E, Cluet D, Falk J, Castellani V, Yuan C, Rudkin BB. Nerve Growth Factor Signaling from Membrane Microdomains to the Nucleus: Differential Regulation by Caveolins. Int J Mol Sci 2017; 18:E693. [PMID: 28338624 PMCID: PMC5412279 DOI: 10.3390/ijms18040693] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 11/16/2022] Open
Abstract
Membrane microdomains or "lipid rafts" have emerged as essential functional modules of the cell, critical for the regulation of growth factor receptor-mediated responses. Herein we describe the dichotomy between caveolin-1 and caveolin-2, structural and regulatory components of microdomains, in modulating proliferation and differentiation. Caveolin-2 potentiates while caveolin-1 inhibits nerve growth factor (NGF) signaling and subsequent cell differentiation. Caveolin-2 does not appear to impair NGF receptor trafficking but elicits prolonged and stronger activation of MAPK (mitogen-activated protein kinase), Rsk2 (ribosomal protein S6 kinase 2), and CREB (cAMP response element binding protein). In contrast, caveolin-1 does not alter initiation of the NGF signaling pathway activation; rather, it acts, at least in part, by sequestering the cognate receptors, TrkA and p75NTR, at the plasma membrane, together with the phosphorylated form of the downstream effector Rsk2, which ultimately prevents CREB phosphorylation. The non-phosphorylatable caveolin-1 serine 80 mutant (S80V), no longer inhibits TrkA trafficking or subsequent CREB phosphorylation. MC192, a monoclonal antibody towards p75NTR that does not block NGF binding, prevents exit of both NGF receptors (TrkA and p75NTR) from lipid rafts. The results presented herein underline the role of caveolin and receptor signaling complex interplay in the context of neuronal development and tumorigenesis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- CREB-Binding Protein/metabolism
- Caveolin 1/antagonists & inhibitors
- Caveolin 1/genetics
- Caveolin 1/metabolism
- Caveolin 2/antagonists & inhibitors
- Caveolin 2/genetics
- Caveolin 2/metabolism
- Cell Differentiation/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Ganglia, Spinal/cytology
- Ganglia, Spinal/metabolism
- Membrane Microdomains/metabolism
- Mice
- Nerve Growth Factor/pharmacology
- Nerve Tissue Proteins
- PC12 Cells
- Phosphorylation/drug effects
- Protein Binding
- Protein Transport/drug effects
- RNA Interference
- RNA, Small Interfering/metabolism
- Rats
- Receptor, Nerve Growth Factor/metabolism
- Receptor, trkA/chemistry
- Receptor, trkA/immunology
- Receptor, trkA/metabolism
- Receptors, Growth Factor
- Receptors, Nerve Growth Factor/chemistry
- Receptors, Nerve Growth Factor/immunology
- Receptors, Nerve Growth Factor/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Ambre Spencer
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
- East China Normal University, School of Life Sciences, Laboratory of Molecular and Cellular Neurophysiology, Shanghai 200062, China.
| | - Lingli Yu
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
- East China Normal University, School of Life Sciences, Laboratory of Molecular and Cellular Neurophysiology, Shanghai 200062, China.
| | - Vincent Guili
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
| | - Florie Reynaud
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, CGphiMC UMR5534, 69622 Villeurbanne Cedex, France.
| | - Yindi Ding
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
- East China Normal University, School of Life Sciences, Laboratory of Molecular and Cellular Neurophysiology, Shanghai 200062, China.
| | - Ji Ma
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- East China Normal University, School of Life Sciences, Laboratory of Molecular and Cellular Neurophysiology, Shanghai 200062, China.
| | - Jérôme Jullien
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
| | - David Koubi
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
| | - Emmanuel Gauthier
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
| | - David Cluet
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
| | - Julien Falk
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, CGphiMC UMR5534, 69622 Villeurbanne Cedex, France.
| | - Valérie Castellani
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, CGphiMC UMR5534, 69622 Villeurbanne Cedex, France.
| | - Chonggang Yuan
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- East China Normal University, School of Life Sciences, Laboratory of Molecular and Cellular Neurophysiology, Shanghai 200062, China.
| | - Brian B Rudkin
- East China Normal University, Key Laboratory of Brain Functional Genomics of the Ministry of Education of PR China, Joint Laboratory of Neuropathogenesis, ECNU, ENS Lyon, CNRS, Shanghai 200062, China.
- Univ. Lyon, Ecole normale supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS, Differentiation & Cell Cycle Group, Laboratoire de Biologie Moléculaire de la Cellule, UMR5239, 69007 Lyon, France.
- Univ. Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France.
| |
Collapse
|
40
|
Ma YD, Thiyagarajan V, Tsai MJ, Lue SI, Chia YC, Shyue SK, Weng CF. Pyrogallol abates VSMC migration via modulation of Caveolin-1, matrix metalloproteinase and intima hyperplasia in carotid ligation mouse. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 48:63-75. [PMID: 27768988 DOI: 10.1016/j.etap.2016.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 06/06/2023]
Abstract
Migration of vascular smooth muscle cells (VSMCs) contributes to intimal hyperplasia and other vascular diseases. Caveolin-1 (Cav-1) has been recognized as a proliferative inhibitor of VSMCs and is likely to be an important regulator of VSMC migration. The underlying mechanism of pyrogallol on the VSMC migration is not fully understood. This study attempted to dissect the role of Cav-1 and matrix metalloproteinase (MMP) in VSMC migration and to investigate the effect of pyrogallol on VSMC mobility during carotid artery ligation mice. The mRNA expression of MMP-3 and MMP-13 was down-regulated in cultured VSMC prepared from Cav-1-deficient (Cav-1 KO) mice whereas MMP-14 expression was up-regulated. Pyrogallol effectively inhibited the migration of Cav-1 KO VSMC by promoting the expression of tissue inhibitors of metalloproteinase (TIMP)-2. Pyrogallol also inhibited the migration of Cav-1 wild type (WT) VSMC, however, by increasing TIMP-1 expression and repressing MMP-2 activity. In a parallel in vivo study, intra-peritoneal (ip) of pyrogallol to carotid artery ligated mice significantly suppressed intima formation in mice carotid artery. Furthermore, the proMMP-9 activity in pyrogallol-treated mice serum significantly increased from Day 0 to Day 2 and decreased from Day 2 to Day 7 in a time-dependent manner. In addition, WT mice treated with pyrogallol had significantly reduced neointima formation, whereas no differences were observed in Cav-1 knock out (KO) mice. These results suggest that pyrogallol not only inhibited VSMC migration but also effectively diminishes the severity of neointima hyperplasia, implying that pyrogallol possesses potential anti-atherogenic effects for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Yu-Dong Ma
- Department of Life Science and the Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
| | - Varadharajan Thiyagarajan
- Department of Life Science and the Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
| | - May-Jywan Tsai
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 11221, Taiwan
| | - Sheng-I Lue
- Department of Life Science and the Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
| | - Yi-Chen Chia
- Department of Food Science & Technology, Tajen University, Ping Tung Hsien, Taiwan
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ching-Feng Weng
- Department of Life Science and the Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| |
Collapse
|
41
|
Nwosu ZC, Ebert MP, Dooley S, Meyer C. Caveolin-1 in the regulation of cell metabolism: a cancer perspective. Mol Cancer 2016; 15:71. [PMID: 27852311 PMCID: PMC5112640 DOI: 10.1186/s12943-016-0558-7] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/03/2016] [Indexed: 12/16/2022] Open
Abstract
Caveolin-1 (CAV1) is an oncogenic membrane protein associated with endocytosis, extracellular matrix organisation, cholesterol distribution, cell migration and signaling. Recent studies reveal that CAV1 is involved in metabolic alterations – a critical strategy adopted by cancer cells to their survival advantage. Consequently, research findings suggest that CAV1, which is altered in several cancer types, influences tumour development or progression by controlling metabolism. Understanding the molecular interplay between CAV1 and metabolism could help uncover druggable metabolic targets or pathways of clinical relevance in cancer therapy. Here we review from a cancer perspective, the findings that CAV1 modulates cell metabolism with a focus on glycolysis, mitochondrial bioenergetics, glutaminolysis, fatty acid metabolism, and autophagy.
Collapse
Affiliation(s)
- Zeribe Chike Nwosu
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Matthias Philip Ebert
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Christoph Meyer
- Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany. .,Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany.
| |
Collapse
|
42
|
Wang Z, Wang N, Liu P, Peng F, Tang H, Chen Q, Xu R, Dai Y, Lin Y, Xie X, Peng C, Situ H. Caveolin-1, a stress-related oncotarget, in drug resistance. Oncotarget 2016; 6:37135-50. [PMID: 26431273 PMCID: PMC4741920 DOI: 10.18632/oncotarget.5789] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/08/2015] [Indexed: 12/28/2022] Open
Abstract
Caveolin-1 (Cav-1) is both a tumor suppressor and an oncoprotein. Cav-1 overexpression was frequently confirmed in advanced cancer stages and positively associated with ABC transporters, cancer stem cell populations, aerobic glycolysis activity and autophagy. Cav-1 was tied to various stresses including radiotherapy, fluid shear and oxidative stresses and ultraviolet exposure, and interacted with stress signals such as AMP-activated protein kinase. Finally, a Cav-1 fluctuation model during cancer development is provided and Cav-1 is suggested to be a stress signal and cytoprotective. Loss of Cav-1 may increase susceptibility to oncogenic events. However, research to explore the underlying molecular network between Cav-1 and stress signals is warranted.
Collapse
Affiliation(s)
- Zhiyu Wang
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Neng Wang
- Department of Breast Oncology, Sun Yat-sen Univeristy Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Pengxi Liu
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fu Peng
- Pharmacy College, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Guangzhou, China
| | - Hailin Tang
- Department of Breast Oncology, Sun Yat-sen Univeristy Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Qianjun Chen
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Xu
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Dai
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Lin
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-sen Univeristy Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Cheng Peng
- Pharmacy College, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Guangzhou, China
| | - Honglin Situ
- Department of Mammary Disease, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
43
|
Hsu EC, Kulp SK, Huang HL, Tu HJ, Salunke SB, Sullivan NJ, Sun D, Wicha MS, Shapiro CL, Chen CS. Function of Integrin-Linked Kinase in Modulating the Stemness of IL-6-Abundant Breast Cancer Cells by Regulating γ-Secretase-Mediated Notch1 Activation in Caveolae. Neoplasia 2016; 17:497-508. [PMID: 26152358 PMCID: PMC4719004 DOI: 10.1016/j.neo.2015.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/19/2015] [Accepted: 06/02/2015] [Indexed: 01/02/2023] Open
Abstract
Interleukin-6 (IL-6) and Notch signaling are important regulators of breast cancer stem cells (CSCs), which drive the malignant phenotype through self-renewal, differentiation, and development of therapeutic resistance. We investigated the role of integrin-linked kinase (ILK) in regulating IL-6–driven Notch1 activation and the ability to target breast CSCs through ILK inhibition. Ectopic expression/short hairpin RNA-mediated knockdown of ILK, pharmacological inhibition of ILK with the small molecule T315, Western blot analysis, immunofluorescence, and luciferase reporter assays were used to evaluate the regulation of IL-6–driven Notch1 activation by ILK in IL-6–producing triple-negative breast cancer cell lines (MDA-MB-231, SUM-159) and in MCF-7 and MCF-7IL-6 cells. The effects of ILK on γ-secretase complex assembly and cellular localization were determined by immunofluorescence, Western blots of membrane fractions, and immunoprecipitation. In vivo effects of T315-induced ILK inhibition on CSCs in SUM-159 xenograft models were assessed by mammosphere assays, flow cytometry, and tumorigenicity assays. Results show that the genetic knockdown or pharmacological inhibition of ILK suppressed Notch1 activation and the abundance of the γ-secretase components presenilin-1, nicastrin, and presenilin enhancer 2 at the posttranscriptional level via inhibition of caveolin-1-dependent membrane assembly of the γ-secretase complex. Accordingly, knockdown of ILK inhibited breast CSC-like properties in vitro and the breast CSC subpopulation in vivo in xenograft tumor models. Based on these findings, we propose a novel function of ILK in regulating γ-secretase–mediated Notch1 activation, which suggests the targeting of ILK as a therapeutic approach to suppress IL-6–induced breast CSCs.
Collapse
Affiliation(s)
- En-Chi Hsu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Samuel K Kulp
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Han-Li Huang
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Huang-Ju Tu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Santosh B Salunke
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Nicholas J Sullivan
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| | - Charles L Shapiro
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA; Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
44
|
Shukla A, Cutucache CE, Sutton GL, Pitner MA, Rai K, Rai S, Opavsky R, Swanson PC, Joshi SS. Absence of caveolin-1 leads to delayed development of chronic lymphocytic leukemia in Eμ-TCL1 mouse model. Exp Hematol 2015; 44:30-7.e1. [PMID: 26435347 DOI: 10.1016/j.exphem.2015.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 09/16/2015] [Accepted: 09/18/2015] [Indexed: 11/19/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the United States. The tissue microenvironment, specifically the lymph nodes, influences the biological and clinical behavior of CLL cells. Gene expression profiling of CLL cells from peripheral blood, bone marrow, and lymph nodes revealed Cav-1 as one of the genes that might be involved in the pathogenesis of CLL. We have previously reported that the knockdown of Cav-1 in primary CLL cells exhibits a significant decrease in cell migration and immune synapse formation. However, the precise role of Cav-1 in CLL initiation and progression in vivo is not known. Therefore, we decreased the expression of Cav-1 in vivo by breeding Eμ-TCL1 with cav-1 knockout mice. We observed a significant decrease in the number of CLL cells and rate of proliferation of CLL cells in spleen, liver, and bone marrow from Eμ-TCL1-Cav1(-/+) and Eμ-TCL1-Cav1(-/-) mice as compared with Eμ-TCL1 mice. In addition, there was a significant increase in survival of Eμ-TCL1-Cav1(-/+) and Eμ-TCL1-Cav1(-/-) compared with Eμ-TCL1 mice. Mechanistically, we observed a decrease in MAPK-Erk signaling measured by p-Erk levels in Eμ-TCL1-Cav1(-/+) mice when compared with Eμ-TCL1-Cav(wt/wt). Together these results indicate that decreased Cav-1 in Eμ-TCL1 mice significantly delays the onset of CLL and decreases leukemic progression by inhibiting MAPK-Erk signaling, suggesting a role for Cav-1 in the proliferation and progression of CLL.
Collapse
Affiliation(s)
- Ashima Shukla
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | | | - Garrett L Sutton
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Michael A Pitner
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Karan Rai
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Siddharth Rai
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Rene Opavsky
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE
| | - Patrick C Swanson
- Department of Medical Microbiology and Immunology, Creighton University Medical Center, Omaha, NE
| | - Shantaram S Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE.
| |
Collapse
|
45
|
Liu K, He Q, Liao G, Han J. Identification of critical genes and gene interaction networks that mediate osteosarcoma metastasis to the lungs. Exp Ther Med 2015; 10:1796-1806. [PMID: 26640552 PMCID: PMC4665845 DOI: 10.3892/etm.2015.2767] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 09/01/2015] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma (OS) is the most commonly diagnosed bone tumor in young adults under the age of 20. Metastasis is considered an important factor underlying cancer-associated morbidity and mortality, and, as a result, the survival rate of patients with metastatic OS is low. In spite of this, the mechanisms underlying metastasis in OS are currently not well understood. The present study compared gene expression levels between five non-metastatic and four metastatic OS tumor samples, using an Affymetrix microarray. A total of 282 genes were differentially expressed in the metastatic samples, as compared with the non-metastatic samples. Of these differentially expressed genes (DEGs), 212 were upregulated and 70 were downregulated. The following DEGs were associated with metastasis: Homeobox only protein; lysosomal-associated membrane protein-3; chemokine (C-C motif) ligand-18; carcinoembryonic antigen-related cell adhesion molecule-6; keratin-19; prostaglandin-endoperoxide synthase-2; clusterin; and nucleoside diphosphate kinase-1. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway analyses were conducted, which identified 529 biological processes (P<0.01) and 10 KEGG pathways (P<0.05) that were significantly over-represented in the metastatic samples, as compared with the non-metastatic samples. Interaction networks for the DEGs were constructed using the corresponding GO terms and KEGG pathways, and these identified numerous genes that may contribute to OS metastasis. Among the enriched biological processes, four DEGs were consistently over-represented: Jun proto-oncogene, caveolin-1, nuclear factor-κB-inhibitor-α and integrin alpha-4; thus suggesting that they may have key roles in OS metastasis, and may be considered potential therapeutic targets in the treatment of patients with OS.
Collapse
Affiliation(s)
- Kegui Liu
- Department of Osteoarticular Surgery, Yantai Shan Hospital, Yantai, Shandong 264000, P.R. China
| | - Qunhui He
- Department of Anesthesiology, Yuhuang Ding Hospital, Yantai, Shandong 264000, P.R. China
| | - Guangjun Liao
- Department of Orthopedic Surgery, Yantai Shan Hospital, Yantai, Shandong 264000, P.R. China
| | - Jian Han
- Department of Orthopedic Surgery, Yantai Shan Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
46
|
Conde-Perez A, Gros G, Longvert C, Pedersen M, Petit V, Aktary Z, Viros A, Gesbert F, Delmas V, Rambow F, Bastian BC, Campbell AD, Colombo S, Puig I, Bellacosa A, Sansom O, Marais R, Van Kempen LCLT, Larue L. A caveolin-dependent and PI3K/AKT-independent role of PTEN in β-catenin transcriptional activity. Nat Commun 2015; 6:8093. [PMID: 26307673 PMCID: PMC4560817 DOI: 10.1038/ncomms9093] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/16/2015] [Indexed: 12/22/2022] Open
Abstract
Loss of the tumour suppressor PTEN is frequent in human melanoma, results in MAPK activation, suppresses senescence and mediates metastatic behaviour. How PTEN loss mediates these effects is unknown. Here we show that loss of PTEN in epithelial and melanocytic cell lines induces the nuclear localization and transcriptional activation of β-catenin independent of the PI3K-AKT-GSK3β axis. The absence of PTEN leads to caveolin-1 (CAV1)-dependent β-catenin transcriptional modulation in vitro, cooperates with NRAS(Q61K) to initiate melanomagenesis in vivo and induces efficient metastasis formation associated with E-cadherin internalization. The CAV1-β-catenin axis is mediated by a feedback loop in which β-catenin represses transcription of miR-199a-5p and miR-203, which suppress the levels of CAV1 mRNA in melanoma cells. These data reveal a mechanism by which loss of PTEN increases CAV1-mediated dissociation of β-catenin from membranous E-cadherin, which may promote senescence bypass and metastasis.
Collapse
Affiliation(s)
- Alejandro Conde-Perez
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Gwendoline Gros
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Christine Longvert
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Valérie Petit
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Zackie Aktary
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Amaya Viros
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Franck Gesbert
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Véronique Delmas
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Florian Rambow
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Boris C Bastian
- Departments of Dermatology and Pathology and UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143, USA
| | | | - Sophie Colombo
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | - Isabel Puig
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| | | | - Owen Sansom
- The Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Leon C L T Van Kempen
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen 6500 HB, The Netherlands
- Jewish General Hospital, Lady Davis Institute for Medical Research, Montreal, Quebec QC H3T 1E2, Canada
- Department of Pathology, McGill University, Montreal, Quebec QC H3T 1E2, Canada
| | - Lionel Larue
- Normal and Pathological Development of Melanocytes, Institut Curie, Orsay 91405, France
- CNRS, UMR3347 Bat. 110, Orsay Cedex 91405, France
- INSERM U1021, Orsay Cedex 91405, France
- Equipe labellisée-Ligue Nationale contre le Cancer, Orsay Cedex 91405, France
| |
Collapse
|
47
|
Lesser-Known Molecules in Ovarian Carcinogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:321740. [PMID: 26339605 PMCID: PMC4538335 DOI: 10.1155/2015/321740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 06/14/2015] [Accepted: 07/07/2015] [Indexed: 12/23/2022]
Abstract
Currently, the deciphering of the signaling pathways brings about new advances in the understanding of the pathogenic mechanism of ovarian carcinogenesis, which is based on the interaction of several molecules with different biochemical structure that, consequently, intervene in cell metabolism, through their role as regulators in proliferation, differentiation, and cell death. Given that the ensemble of biomarkers in OC includes more than 50 molecules the interest of the researchers focuses on the possible validation of each one's potential as prognosis markers and/or therapeutic targets. Within this framework, this review presents three protein molecules: ALCAM, c-FLIP, and caveolin, motivated by the perspectives provided through the current limited knowledge on their role in ovarian carcinogenesis and on their potential as prognosis factors. Their structural stability, once altered, triggers the initiation of the sequences characteristic for ovarian carcinogenesis, through their role as modulators for several signaling pathways, contributing to the disruption of cellular junctions, disturbance of pro-/antiapoptotic equilibrium, and alteration of transmission of the signals specific for the molecular pathways. For each molecule, the text is built as follows: (i) general remarks, (ii) structural details, and (iii) particularities in expression, from different tumors to landmarks in ovarian carcinoma.
Collapse
|
48
|
Knopf JD, Tholen S, Koczorowska MM, De Wever O, Biniossek ML, Schilling O. The stromal cell-surface protease fibroblast activation protein-α localizes to lipid rafts and is recruited to invadopodia. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [PMID: 26209915 DOI: 10.1016/j.bbamcr.2015.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibroblast activation protein alpha (FAPα) is a cell surface protease expressed by cancer-associated fibroblasts in the microenvironment of most solid tumors. As there is increasing evidence for proteases having non-catalytic functions, we determined the FAPα interactome in cancer-associated fibroblasts using the quantitative immunoprecipitation combined with knockdown (QUICK) method. Complex formation with adenosin deaminase, erlin-2, stomatin, prohibitin, Thy-1 membrane glycoprotein, and caveolin-1 was further validated by immunoblotting. Co-immunoprecipitation (co-IP) of the known stoichiometric FAPα binding partner dipeptidyl-peptidase IV (DPPIV) corroborated the proteomic strategy. Reverse co-IPs validated the FAPα interaction with caveolin-1, erlin-2, and stomatin while co-IP upon RNA-interference mediated knock-down of DPPIV excluded adenosin deaminase as a direct FAPα interaction partner. Many newly identified FAPα interaction partners localize to lipid rafts, including caveolin-1, a widely-used marker for lipid raft localization. We hypothesized that this indicates a recruitment of FAPα to lipid raft structures. In density gradient centrifugation, FAPα co-fractionates with caveolin-1. Immunofluorescence optical sectioning microscopy of FAPα and lipid raft markers further corroborates recruitment of FAPα to lipid rafts and invadopodia. FAPα is therefore an integral component of stromal lipid rafts in solid tumors. In essence, we provide one of the first interactome analyses of a cell surface protease and translate these results into novel biological aspects of a marker protein for cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Julia D Knopf
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany; Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Stefan Tholen
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Maria M Koczorowska
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Ghent University Hospital, 1P7, De Pintelaan 185, 9000 Gent, Belgium
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, University of Freiburg, D-79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
49
|
Abstract
Rhabdomyosarcoma is the most common soft tissue sarcoma of childhood and adolescence. Despite advances in therapy, patients with histological variant of rhabdomyosarcoma known as alveolar rhabdomyosarcoma (ARMS) have a 5-year survival of less than 30%. Caveolin-1 (CAV1), encoding the structural component of cellular caveolae, is a suggested tumor suppressor gene involved in cell signaling. In the present study we report that compared to other forms of rhabdomyosarcoma (RMS) CAV1 expression is either undetectable or very low in ARMS cell lines and tumor samples. DNA methylation analysis of the promoter region and azacytidine-induced re-expression suggest the involvement of epigenetic mechanisms in the silencing of CAV1. Reintroduction of CAV1 in three of these cell lines impairs their clonogenic capacity and promotes features of muscular differentiation. In vitro, CAV1-expressing cells show high expression of Caveolin-3 (CAV3), a muscular differentiation marker. Blockade of MAPK signaling is also observed. In vivo, CAV1-expressing xenografts show growth delay, features of muscular differentiation and increased cell death. In summary, our results suggest that CAV1 could function as a potent tumor suppressor in ARMS tumors. Inhibition of CAV1 function therefore, could contribute to aberrant cell proliferation, leading to ARMS development.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Much effort has been devoted to determining how cellular and noncellular components of the tumoral niche initiate and promote cancer development. Cancer cells perceive biochemical signals from components of the extracellular matrix (ECM) and sense physical features, such as matrix stiffness and cell confinement. The past decade has seen a better understanding of the biophysics and mechanobiology associated with cancer cells. Indeed, loss of mechanisms controlling the production, the degradation, and the remodeling of ECM contributes to tumor growth or cell dissemination by affecting cell contractility in response to ECM stiffness and by stimulating mechanical dependence of growth factor activation. RESULTS Cell plasticity allows adaptative strategies for cancer cells to survive or eventually escape from tumoral environment through modification of the microenvironment-cell interface, internal tension increase, and nuclear deformation partly leading to intratumoral heterogeneity. However, although alteration of the biomechanical properties of the ECM are sufficient to promote cell migration and invasion in cancer cells, this microenvironment can also provide a hospitable niche for tumor dormancy and resistance to cancer therapy. CONCLUSION The review will focus on how physicochemical properties of ECM might promote tumor growth or cell dissemination or on the contrary maintain quiescent state of cancer cells. It is crucial to clarify the molecular basis of mechanotransduction in the development and progression of tumors to identify new potential biomarkers and anticancer therapeutic targets.
Collapse
|