1
|
Surendra Panikar S, Shmuel S, Lewis JS, Pereira PMR. PET and Optical Imaging of Caveolin-1 in Gastric Tumors. ACS OMEGA 2023; 8:35884-35892. [PMID: 37810678 PMCID: PMC10552508 DOI: 10.1021/acsomega.3c03614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023]
Abstract
Previous studies have suggested tumoral caveolin-1 (CAV1) as a predictive biomarker for the response to anti-HER2 antibody drug therapies in gastric tumors. In this study, radiolabeled and fluorescently labeled anti-CAV1 antibodies were developed and tested as an immunoPET or optical imaging agent to detect CAV1 in HER2-positive/CAV1-high NCIN87 gastric tumors. The expression of CAV1 receptors in NCIN87 gastric tumors and nontumor murine organs was determined by Western blot. Binding assays were performed to validate the anti-CAV1 antibody specificity for CAV1-expressing NCIN87 cancer cells. Subcutaneous and orthotopic NCIN87 xenografts were used for PET imaging and ex vivo biodistribution of the radioimmunoconjugate. Additional HER2-PET and CAV1-optical imaging was also performed to determine CAV1 in the HER2-positive tumors. 89Zr-labeled anti-CAV1 antibody was able to bind to CAV1-expressing NCIN87 cells with a Bmax value of 2.7 × 103 CAV1 receptors/cell in vitro. ImmunoPET images demonstrated the localization of the antibody in subcutaneous NCIN87 xenografts. In the orthotopic model, CAV1 expression was also observed by optical imaging in the HER2-positive tumors previously imaged with HER2-PET. Ex vivo biodistribution analysis further confirmed these imaging results. The preclinical data from this study demonstrate the potential of using CAV1-PET and optical imaging for detecting gastric tumors.
Collapse
Affiliation(s)
- Sandeep Surendra Panikar
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shayla Shmuel
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Department
of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Patrícia M. R. Pereira
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
2
|
Ren L, Zhou P, Wu H, Liang Y, Xu R, Lu H, Chen Q. Caveolin-1 is a prognostic marker and suppresses the proliferation of breast cancer. Transl Cancer Res 2022; 10:3797-3810. [PMID: 35116679 PMCID: PMC8798413 DOI: 10.21037/tcr-21-1139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022]
Abstract
Background To explore the role of caveolin-1 (Cav-1) in breast cancer (BC). Methods Cav-1 expression data were downloaded from the Tumor Immune Estimation Resource (TIMER) and Gene Expression Omnibus (GEO) databases. We compared the expression of Cav-1 in different tumor tissues and between BC tissues and normal tissues (NTs), as well as the differences between different clinical traits. Kaplan-Meier survival analysis and univariate and multivariate Cox regression analyses were used to determine whether Cav-1 serves as a prognostic factor. The correlations of Cav-1 expression with the immune microenvironment and infiltrating immune cells were also analyzed. Quantitative polymerase chain reaction (qPCR) was used to detect Cav-1 mRNA expression in the MCF-7, SKB-R3, MDB-MB-231, and SUM-159 cell lines. LV-Cav-1-RNAi was transfected into MCF-7 and MDB-MB-231 cells, and the MTT assay was used to detect cell proliferation. Subsequently, MDB-MB-231 cells carrying the Cav-1-RNAi gene were used to determine the effects of Cav-1 knockdown on tumor growth in vivo using a severe combined immunodeficiency (SCID) model. Results Cav-1 was enriched in most solid tumors, and its expression was lower in BC tissues than in NT. Cav-1 expression was shown to be related to patients’ clinical outcomes. Cav-1 was expressed in the MCF-7, SKB-R3, MDB-MB-231, and SUM-159 cell lines. The MTT assay revealed that the proliferative ability of MDB-MB-231 and MCF-7 cells was accelerated. The tumor volume of SCID mice administered with LV-Cav-1-RNAi cells was increased. Conclusions These results suggest that Cav-1 may serve as a suppressor in the development of BC.
Collapse
Affiliation(s)
- Liping Ren
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peijuan Zhou
- Department of Traditional Chinese Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huajia Wu
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Liang
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Xu
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hai Lu
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianjun Chen
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Breast Disease, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Low JY, Laiho M. Caveolae-Associated Molecules, Tumor Stroma, and Cancer Drug Resistance: Current Findings and Future Perspectives. Cancers (Basel) 2022; 14:cancers14030589. [PMID: 35158857 PMCID: PMC8833326 DOI: 10.3390/cancers14030589] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell membranes contain small invaginations called caveolae. They are a specialized lipid domain and orchestrate cellular signaling events, mechanoprotection, and lipid homeostasis. Formation of the caveolae depends on two classes of proteins, the caveolins and cavins, which form large complexes that allow their self-assembly into caveolae. Loss of either of these two proteins leads to distortion of the caveolae structure and disruption of many physiological processes that affect diseases of the muscle, metabolic states governing lipids, and the glucose balance as well as cancers. In cancers, the expression of caveolins and cavins is heterogenous, and they undergo alterations both in the tumors and the surrounding tumor microenvironment stromal cells. Remarkably, their expression and function has been associated with resistance to many cancer drugs. Here, we summarize the current knowledge of the resistance mechanisms and how this knowledge could be applied into the clinic in future. Abstract The discovery of small, “cave-like” invaginations at the plasma membrane, called caveola, has opened up a new and exciting research area in health and diseases revolving around this cellular ultrastructure. Caveolae are rich in cholesterol and orchestrate cellular signaling events. Within caveola, the caveola-associated proteins, caveolins and cavins, are critical components for the formation of these lipid rafts, their dynamics, and cellular pathophysiology. Their alterations underlie human diseases such as lipodystrophy, muscular dystrophy, cardiovascular disease, and diabetes. The expression of caveolins and cavins is modulated in tumors and in tumor stroma, and their alterations are connected with cancer progression and treatment resistance. To date, although substantial breakthroughs in cancer drug development have been made, drug resistance remains a problem leading to treatment failures and challenging translation and bench-to-bedside research. Here, we summarize the current progress in understanding cancer drug resistance in the context of caveola-associated molecules and tumor stroma and discuss how we can potentially design therapeutic avenues to target these molecules in order to overcome treatment resistance.
Collapse
Affiliation(s)
- Jin-Yih Low
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Correspondence: ; Tel.: +1-410-502-9748; Fax: +1-410-502-2821
| | - Marikki Laiho
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA;
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
4
|
Yang C, He B, Dai W, Zhang H, Zheng Y, Wang X, Zhang Q. The role of caveolin-1 in the biofate and efficacy of anti-tumor drugs and their nano-drug delivery systems. Acta Pharm Sin B 2021; 11:961-977. [PMID: 33996409 PMCID: PMC8105775 DOI: 10.1016/j.apsb.2020.11.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
As one of the most important components of caveolae, caveolin-1 is involved in caveolae-mediated endocytosis and transcytosis pathways, and also plays a role in regulating the cell membrane cholesterol homeostasis and mediating signal transduction. In recent years, the relationship between the expression level of caveolin-1 in the tumor microenvironment and the prognostic effect of tumor treatment and drug treatment resistance has also been widely explored. In addition, the interplay between caveolin-1 and nano-drugs is bidirectional. Caveolin-1 could determine the intracellular biofate of specific nano-drugs, preventing from lysosomal degradation, and facilitate them penetrate into deeper site of tumors by transcytosis; while some nanocarriers could also affect caveolin-1 levels in tumor cells, thereby changing certain biophysical function of cells. This article reviews the role of caveolin-1 in tumor prognosis, chemotherapeutic drug resistance, antibody drug sensitivity, and nano-drug delivery, providing a reference for the further application of caveolin-1 in nano-drug delivery systems.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ADC, antibody drug conjugates
- BBB, blood–brain barrier
- Biofate
- CAFs, cancer-associated fibroblasts
- CPT, camptothecin
- CSD, caveolin scaffolding domain
- CTB, cholera toxins B
- Cancer
- Caveolin-1
- Drug resistance
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- ER, endoplasmic reticulum
- ERK, extracellular regulated protein kinases
- FGF2, fibroblast growth factor 2
- GGT, γ-glutamyl transpeptidase
- GPI, glycosylphosphatidylinositol
- HER2, human epidermal growth factor receptor 2
- HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A
- HSA, human serum albumin
- IBC, infiltrating breast cancer
- IR, insulin receptor
- MAPK, mitogen-activated protein kinase
- MDR, multidrug resistance
- MSV, multistage nanovectors
- NPs, nanoparticles
- Nano-drug delivery systems
- PC, prostate cancer
- PDGF, platelet-derived growth factor
- PFS, progression free survival
- ROS, reactive oxygen species
- SCLC, small cell lung cancer
- SV40, simian virus 40
- Transcytosis
- cell SMA, styrene maleic acid
Collapse
|
5
|
Abstract
Resistance of solid tumors to chemo- and radiotherapy remains a major obstacle in anti-cancer treatment. Herein, the membrane protein caveolin-1 (CAV1) came into focus as it is highly expressed in many tumors and high CAV1 levels were correlated with tumor progression, invasion and metastasis, and thus a worse clinical outcome. Increasing evidence further indicates that the heterogeneous tumor microenvironment, also known as the tumor stroma, contributes to therapy resistance resulting in poor clinical outcome. Again, CAV1 seems to play an important role in modulating tumor host interactions by promoting tumor growth, metastasis, therapy resistance and cell survival. However, the mechanisms driving stroma-mediated tumor growth and radiation resistance remain to be clarified. Understanding these interactions and thus, targeting CAV1 may offer a novel strategy for preventing cancer therapy resistance and improving clinical outcomes. In this review, we will summarize the resistance-promoting effects of CAV1 in tumors, and emphasize its role in the tumor-stroma communication as well as the resulting malignant phenotype of epithelial tumors.
Collapse
Affiliation(s)
- Julia Ketteler
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
6
|
Wu CH, Liu FC, Pan CH, Lai MT, Lan SJ, Wu CH, Sheu MJ. Suppression of Cell Growth, Migration and Drug Resistance by Ethanolic Extract of Antrodia cinnamomea in Human Lung Cancer A549 Cells and C57BL/6J Allograft Tumor Model. Int J Mol Sci 2018. [PMID: 29522490 PMCID: PMC5877652 DOI: 10.3390/ijms19030791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The purpose of this study was to investigate the inhibitory activities of ethanolic extracts from Antrodia cinnamomea (EEAC) on lung cancer. Cell proliferation and cell cycle distribution were analyzed using (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) (MTT) assay and flow cytometry, respectively. Wound-healing assay, Western blotting, and a murine tumor model were separately used to examine cell migration, protein expression, and tumor repression. Our results showed that EEAC induced cell cycle arrest at the G0/G1 phase resulting decreased cell viability in A549 cells. Moreover, EEAC up-regulated the growth-suppressing proteins, adenosine 5′-monophosphate-activated protein kinase (AMPK), p21 and p27, but down-regulated the growth-promoting proteins, protein kinase B (Akt), mammalian tarfet of rapamycin (mTOR), extracellular signal-regulating kinase 1/2 (ERK1/2), retinoblastoma protein (Rb), cyclin E, and cyclin D1. EEAC also inhibited A549 cell migration and reduced expression of gelatinases. In addition, our data showed that tumor growth was suppressed after treatment with EEAC in a murine allograft tumor model. Some bioactive compounds from EEAC, such as cordycepin and zhankuic acid A, were demonstrated to reduce the protein expressions of matrix metalloproteinase (MMP)-9 and cyclin D1 in A549 cells. Furthermore, EEAC enhanced chemosensitivity of A549 to paclitaxel by reducing the protein levels of caveolin-1. Our data suggests that EEAC has the potential to be an adjuvant medicine for the treatment of lung cancer.
Collapse
Affiliation(s)
- Chi-Han Wu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Fon-Chang Liu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Chun-Hsu Pan
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ming-Tsung Lai
- Department of Pathology, Taichung Hospital, Ministry of Health and Welfare Taiwan, Taichung 40343, Taiwan.
| | - Shou-Jen Lan
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan.
| | - Chieh-Hsi Wu
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ming-Jyh Sheu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
7
|
Nguyen KCT, Cho KA. Versatile Functions of Caveolin-1 in Aging-related Diseases. Chonnam Med J 2017; 53:28-36. [PMID: 28184336 PMCID: PMC5299127 DOI: 10.4068/cmj.2017.53.1.28] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 12/24/2022] Open
Abstract
Caveolin-1 (Cav-1) is a trans-membrane protein that is a major component of the caveolae structure on the plasma membrane. Cav-1 is involved in the regulation of various cellular processes, including cell growth, differentiation, endocytosis, and in particular it has been implied in cellular senescence. Here we review current knowledge about Cav-1 in cellular signaling and discuss the role of Cav-1 in aging-related diseases.
Collapse
Affiliation(s)
- Kim Cuc Thi Nguyen
- Deparment of Life Science, ThaiNguyen University of Science, TanThinh Ward, ThaiNguyen, VietNam
| | - Kyung A Cho
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
8
|
Zou W, Ma X, Hua W, Chen B, Cai G. Caveolin-1 mediates chemoresistance in cisplatin-resistant ovarian cancer cells by targeting apoptosis through the Notch-1/Akt/NF-κB pathway. Oncol Rep 2016; 34:3256-63. [PMID: 26503358 DOI: 10.3892/or.2015.4320] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/26/2015] [Indexed: 11/06/2022] Open
Abstract
Caveolin-1 (Cav-1), a family of ubiquitously expressed oligomeric structural proteins in many mammalian cells, has been shown to be an effective regulator of tumorigenesis. Recent studies have indicated that Cav-1 can promote resistance to chemotherapy in a variety of tumors. However, the regulation of Cav-1 on chemoresistance in ovarian cancer is still unknown. In the present study, the mRNA and protein expression level was investigated by RT-PCR and western blot analysis, respectively, and the 50% inhibitory concentration (IC50) value was measured by MTT assay. The protein expression level of P-glycoprotein (P-gp), Notch-1, p-Akt and p-NF-κB p65 were detected using western blot analysis and the apoptotic ratio was determined using the Annexin V-FITC/PI detection kit. The results showed that the mRNA and protein expression levels of Cav-1 were significantly higher in SKOV3/DDP and A2780/DDP than in SKOV3 and A2780, respectively. Knockdown of Cav-1 significantly decreased the IC50 value in cisplatin-resistant cells. The protein expression level of P-gp in SKOV3/DDP and A2780/DDP was significant higher than SKOV3 and A2780, respectively, and had no correlation with the Cav-1 siRNA transfection. The apoptotic ratio induced by cisplatin in normal ovarian cancer cells was higher than cisplatin-resistant ovarian cancer cells, and knockdown of Cav-1 could significantly enhance cisplatin induced cell apoptosis. Furthermore, knockdown of Cav-1 was also able to significantly downregulate the protein expression level of Notch-1, p-Akt and p-NF-κB p65 in cisplatin-resistant ovarian cancer cells. Overexpression of Cav-1 upregulated the IC50 value, but under the effect of Notch-1 siRNA or LY294002 or PDTC, the IC50 value was markedly decreased. Our results suggested that Cav-1 can promote the chemoresistance of ovarian cancer by targeting apoptosis through the Notch-1/Akt/NF-κB pathway.
Collapse
|
9
|
Emerging Role of miRNAs in the Drug Resistance of Gastric Cancer. Int J Mol Sci 2016; 17:424. [PMID: 27011182 PMCID: PMC4813275 DOI: 10.3390/ijms17030424] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/11/2016] [Accepted: 03/16/2016] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer is the third leading cause of cancer mortality worldwide. Unfortunately, most gastric cancer cases are diagnosed in an advanced, non-curable stage and with a limited response to chemotherapy. Drug resistance is one of the most important causes of therapy failure in gastric cancer patients. Although the mechanisms of drug resistance have been broadly studied, the regulation of these mechanisms has not been completely understood. Accumulating evidence has recently highlighted the role of microRNAs in the development and maintenance of drug resistance due to their regulatory features in specific genes involved in the chemoresistant phenotype of malignancies, including gastric cancer. This review summarizes the current knowledge about the miRNAs’ characteristics, their regulation of the genes involved in chemoresistance and their potential as targeted therapies for personalized treatment in resistant gastric cancer.
Collapse
|
10
|
Gupta R, Toufaily C, Annabi B. Caveolin and cavin family members: dual roles in cancer. Biochimie 2014; 107 Pt B:188-202. [PMID: 25241255 DOI: 10.1016/j.biochi.2014.09.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 09/04/2014] [Indexed: 12/16/2022]
Abstract
Caveolae are specialized plasma membrane subdomains with distinct lipid and protein compositions, which play an essential role in cell physiology through regulation of trafficking and signaling functions. The structure and functions of caveolae have been shown to require the proteins caveolins. Recently, members of the cavin protein family were found to be required, in concert with caveolins, for the formation and function of caveolae. Caveolins have a paradoxical role in the development of cancer formation. They have been involved in both tumor suppression and oncogenesis, depending on tumor type and progress stage. High expression of caveolins and cavins leads to inhibition of cancer-related pathways, such as growth factor signaling pathways. However, certain cancer cells that express caveolins and cavins have been shown to be more aggressive and metastatic because of their increased potential for anchorage-independent growth. Here, we will survey the functional roles of caveolins and of different cavin family members in cancer regulation.
Collapse
Affiliation(s)
- Reshu Gupta
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMed, Département de Chimie, Université du Québec à Montréal, Québec H3C 3P8, Canada.
| | - Chirine Toufaily
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMed, Département de Chimie, Université du Québec à Montréal, Québec H3C 3P8, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMed, Département de Chimie, Université du Québec à Montréal, Québec H3C 3P8, Canada
| |
Collapse
|
11
|
Zhang Y, Qu X, Li C, Fan Y, Che X, Wang X, Cai Y, Hu X, Liu Y. miR-103/107 modulates multidrug resistance in human gastric carcinoma by downregulating Cav-1. Tumour Biol 2014; 36:2277-85. [PMID: 25407491 DOI: 10.1007/s13277-014-2835-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/10/2014] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of non-protein-coding small RNAs with the capacity to regulate fundamental biological processes essential for cancer initiation and progression. In the present study, we analyzed miRNA expression levels between multidrug-resistant gastric carcinoma cell line SGC7901/ADR and its parent cell line SGC7901 using a miRNA microarray. MiR-103/107 was downregulated compared with parental SGC7901 cells. Overexpression of miR-103/107 sensitized SGC7901/ADR cells to doxorubicin (DOX), as demonstrated by in vitro and in vivo drug sensitivity assay. We further confirmed that miR-103/107 inhibited P-gp function in gastric cancer SGC7901/ADR cells. Finally, we verified that caveolin-1 (Cav-1), a critical component of lipid rafts, was a target of miR-103/107.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, China
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Campbell L, Al-Jayyoussi G, Gutteridge R, Gumbleton N, Griffiths R, Gumbleton S, Smith MW, Griffiths DFR, Gumbleton M. Caveolin-1 in renal cell carcinoma promotes tumour cell invasion, and in co-operation with pERK predicts metastases in patients with clinically confined disease. J Transl Med 2013; 11:255. [PMID: 24119769 PMCID: PMC4015803 DOI: 10.1186/1479-5876-11-255] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/30/2013] [Indexed: 11/11/2022] Open
Abstract
Background Up to 40% of patients initially diagnosed with clinically-confined renal cell carcinoma (RCC) and who undergo curative surgery will nevertheless relapse with metastatic disease (mRCC) associated with poor long term survival. The discovery of novel prognostic/predictive biomarkers and drug targets is needed and in this context the aim of the current study was to investigate a putative caveolin-1/ERK signalling axis in clinically confined RCC, and to examine in a panel of RCC cell lines the effects of caveolin-1 (Cav-1) on pathological processes (invasion and growth) and select signalling pathways. Methods Using immunohistochemistry we assessed the expression of both Cav-1 and phosphorylated-ERK (pERK) in 176 patients with clinically confined RCC, their correlation with histological parameters and their impact upon disease-free survival. Using a panel of RCC cell lines we explored the functional effects of Cav-1 knockdown upon cell growth, cell invasion and VEGF-A secretion, as well Cav-1 regulation by cognate cell signalling pathways. Results We found a significant correlation (P = 0.03) between Cav-1 and pERK in a cohort of patients with clinically confined disease which represented a prognostic biomarker combination (HR = 4.2) that effectively stratified patients into low, intermediate and high risk groups with respect to relapse, even if the patients’ tumours displayed low grade and/or low stage disease. In RCC cell lines Cav-1 knockdown unequivocally reduced cell invasive capacity while also displaying both pro-and anti-proliferative effects; targeted knockdown of Cav-1 also partially suppressed VEGF-A secretion in VHL-negative RCC cells. The actions of Cav-1 in the RCC cell lines appeared independent of both ERK and AKT/mTOR signalling pathways. Conclusion The combined expression of Cav-1 and pERK serves as an independent biomarker signature with potential merit in RCC surveillance strategies able to predict those patients with clinically confined disease who will eventually relapse. In a panel of in-vitro RCC cells Cav-1 promotes cell invasion with variable effects on cell growth and VEGF-A secretion. Cav-1 has potential as a therapeutic target for the prevention and treatment of mRCC.
Collapse
Affiliation(s)
- Lee Campbell
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3XF, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kibria G, Hatakeyama H, Ohga N, Hida K, Harashima H. The effect of liposomal size on the targeted delivery of doxorubicin to Integrin αvβ3-expressing tumor endothelial cells. Biomaterials 2013; 34:5617-27. [PMID: 23623323 DOI: 10.1016/j.biomaterials.2013.03.094] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/29/2013] [Indexed: 11/30/2022]
Abstract
Size of the liposomes (LPs) specially governs its biodistribution. In this study, LPs were developed with controlled sizes, where variation in LP size dictates the ligand-receptor interaction, cellular internalization and its distribution within the tumor microenvironment. The therapeutic efficacies of doxorubicin (DOX)-loaded RGD modified small size (~100 nm in diameter, dnm) and large size (~300 dnm) PEGylated LPs (RGD-PEG-LPs) were compared to that of Doxil (a clinically used DOX-loaded PEG-LP, ~100 dnm) in DOX resistant OSRC-2 (Renal cell carcinoma, RCC) tumor xenografts. Doxil, which accumulated in tumor tissue via the enhanced permeability and retention (EPR) effect, failed to suppress tumor growth. Small size RGD-PEG-LP, that targets the tumor endothelial cells (TECs) and extravasates to tumor cells, failed to provide anti-tumor effect. Large size RGD-PEG-LP preferentially targets the TECs via minimization of the EPR effect, and significantly reduced the tumor growth, which was exerted through its strong anti-angiogenic activity on the tumor vasculature rather than having a direct effect on DOX resistant RCC. The prepared large size RGD-PEG-LP that targets the TECs via interacting with Integrin αvβ3, is a potentially effective and alternate therapeutic strategy for the treatment of DOX resistant tumor cells by utilizing DOX, in cases where Doxil is ineffective.
Collapse
Affiliation(s)
- Golam Kibria
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, Hokkaido 060-0812, Japan
| | | | | | | | | |
Collapse
|
14
|
Quann K, Gonzales DM, Mercier I, Wang C, Sotgia F, Pestell RG, Lisanti MP, Jasmin JF. Caveolin-1 is a negative regulator of tumor growth in glioblastoma and modulates chemosensitivity to temozolomide. Cell Cycle 2013; 12:1510-20. [PMID: 23598719 PMCID: PMC3680531 DOI: 10.4161/cc.24497] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Caveolin-1 (Cav-1) is a critical regulator of tumor progression in a variety of cancers where it has been shown to act as either a tumor suppressor or tumor promoter. In glioblastoma multiforme, it has been previously demonstrated to function as a putative tumor suppressor. Our studies here, using the human glioblastoma-derived cell line U-87MG, further support the role of Cav-1 as a negative regulator of tumor growth. Using a lentiviral transduction approach, we were able to stably overexpress Cav-1 in U-87MG cells. Gene expression microarray analyses demonstrated significant enrichment in gene signatures corresponding to downregulation of MAPK, PI3K/AKT and mTOR signaling, as well as activation of apoptotic pathways in Cav-1-overexpressing U-87MG cells. These same gene signatures were later confirmed at the protein level in vitro. To explore the ability of Cav-1 to regulate tumor growth in vivo, we further show that Cav-1-overexpressing U-87MG cells display reduced tumorigenicity in an ectopic xenograft mouse model, with marked hypoactivation of MAPK and PI3K/mTOR pathways. Finally, we demonstrate that Cav-1 overexpression confers sensitivity to the most commonly used chemotherapy for glioblastoma, temozolomide. In conclusion, Cav-1 negatively regulates key cell growth and survival pathways and may be an effective biomarker for predicting response to chemotherapy in glioblastoma.
Collapse
Affiliation(s)
- Kevin Quann
- Department of Stem Cell Biology & Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Senetta R, Stella G, Pozzi E, Sturli N, Massi D, Cassoni P. Caveolin-1 as a promoter of tumour spreading: when, how, where and why. J Cell Mol Med 2013; 17:325-36. [PMID: 23521716 PMCID: PMC3823014 DOI: 10.1111/jcmm.12030] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/15/2013] [Indexed: 11/29/2022] Open
Abstract
Caveolae are non-clathrin invaginations of the plasma membrane in most cell types; they are involved in signalling functions and molecule trafficking, thus modulating several biological functions, including cell growth, apoptosis and angiogenesis. The major structural protein in caveolae is caveolin-1, which is known to act as a key regulator in cancer onset and progression through its role as a tumour suppressor. Caveolin-1 can also promote cell proliferation, survival and metastasis as well as chemo- and radioresistance. Here, we discuss recent findings and novel concepts that support a role for caveolin-1 in cancer development and its distant spreading. We also address the potential application of caveolin-1 in tumour therapy and diagnosis.
Collapse
Affiliation(s)
- Rebecca Senetta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | | | | | | |
Collapse
|
16
|
Volatile anesthetics protect cancer cells against tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis via caveolins. Anesthesiology 2011; 115:499-508. [PMID: 21862885 DOI: 10.1097/aln.0b013e3182276d42] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Volatile anesthetics have a dual effect on cell survival dependent on caveolin expression. The effect of volatile anesthetics on cancer cell survival and death after anesthetic exposure has not been well investigated. The authors examined the effects of isoflurane exposure on apoptosis and its regulation by caveolin-1 (Cav-1). METHODS The authors exposed human colon cancer cell lines to isoflurane and proapoptotic stimuli and assessed what role Cav-1 plays in cell protection. They evaluated apoptosis using assays for nucleosomal fragmentation, cleaved caspase 3 expression, and caspase activity assays. To test the mechanism, they used pharmacologic inhibitors (i.e., pertussis toxin) and assessed changes in glycolysis. RESULTS Apoptosis as measured by nucleosomal fragmentation was enhanced by isoflurane (1.2% in air) in HT29 (by 64% relative to control, P < 0.001) and decreased in HCT116 (by 23% relative to control, P < 0.001) cells. Knockdown of Cav-1 in HCT116 cells increased the sensitivity to apoptotic stimuli but not with scrambled small interfering RNA (siRNA) treatment (19.7 ± 0.4 vs. 20.0 ± 0.6, P = 0.7786 and 19.7 ± 0.5 vs. 16.3 ± 0.4, P = 0.0012, isoflurane vs. control in Cav-1 small interfering RNA vs. scrambled small interfering RNA treated cells, respectively). The protective effect of isoflurane with various exposure times on apoptosis was enhanced in HT29 cells overexpressing Cav-1 (P < 0.001 by two-way ANOVA). Pertussis toxin effectively blocked the antiapoptotic effect of isoflurane exhibited by Cav-1 in all cell lines. Cav-1 cells had increased glycolysis with isoflurane exposure; however, in the presence of tumor necrosis factor-related apoptosis-inducing ligand, this increase in glycolysis was maintained in HT29-Cav-1 but not control cells. CONCLUSION Brief isoflurane exposure leads to resistance against apoptosis via a Cav-1-dependent mechanism.
Collapse
|
17
|
Pongjit K, Chanvorachote P. Caveolin-1 sensitizes cisplatin-induced lung cancer cell apoptosis via superoxide anion-dependent mechanism. Mol Cell Biochem 2011; 358:365-73. [DOI: 10.1007/s11010-011-0988-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
|