1
|
Chintalaramulu N, Singh DP, Sapkota B, Raman D, Alahari S, Francis J. Caveolin-1: an ambiguous entity in breast cancer. Mol Cancer 2025; 24:109. [PMID: 40197489 PMCID: PMC11974173 DOI: 10.1186/s12943-025-02297-8] [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: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer in women and the second leading cause of death from cancer among women. Metastasis is the major cause of BC-associated mortality. Accumulating evidence implicates Caveolin-1 (Cav-1), a structural protein of plasma membrane caveolae, in BC metastasis. Cav-1 exhibits a dual role, as both a tumor suppressor and promoter depending on the cellular context and BC subtype. This review highlights the role of Cav-1 in modulating glycolytic metabolism, tumor-stromal interactions, apoptosis, and senescence. Additionally, stromal Cav-1's expression is identified as a potential prognostic marker, offering insights into its contrasting roles in tumor suppression and progression. Furthermore, Cav-1's context-dependent effects are explored in BC subtypes including hormone receptor-positive, HER2-positive, and triple-negative BC (TNBC). The review further delves into the role of Cav-1 in regulating the metastatic cascade including extracellular matrix interactions, cell migration and invasion, and premetastatic niche formation. The later sections discuss the therapeutic targeting of Cav-1 by metabolic inhibitors such as betulinic acid and Cav-1 modulating compounds. While Cav-1 may be a potential biomarker and therapeutic target, its heterogeneous expression and context-specific activity necessitates further research to develop precise interventions. Future studies investigating the mechanistic role of Cav-1 in metastasis may pave the way for effective treatment of metastatic BC.
Collapse
Affiliation(s)
- Naveen Chintalaramulu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | | | - Biplov Sapkota
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
2
|
Huang X, Ali A, Yachioui DEI, Le Dévédec SE, Hankemeier T. Lipid dysregulation in triple negative breast cancer: Insights from mass spectrometry-based approaches. Prog Lipid Res 2025; 98:101330. [PMID: 39914749 DOI: 10.1016/j.plipres.2025.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Triple negative breast cancer (TNBC) has the worst prognosis among breast cancers due to its aggressive nature and the absence of targeted treatments. Development of novel anti-cancer drugs for TNBC faces challenges stemming from its heterogeneity and high potential for metastasis. Metabolomics can be a useful technology in finding novel therapeutic targets and probing the heterogeneity of TNBC. Metabolomics has been enabled by advancements in mass spectrometry (MS)-based platforms that facilitated comprehensive profiling of TNBC metabolism. This review provides an overview of metabolomic changes in TNBC with emphasis on lipid alterations, and describes the key MS analytical techniques, providing the necessary background for examining the role of lipids in TNBC development.
Collapse
Affiliation(s)
- Xiaoyue Huang
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Ahmed Ali
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Dounia E I Yachioui
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sylvia E Le Dévédec
- Division of Cell Systems and Drug Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands.
| | - Thomas Hankemeier
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
3
|
Kundrapu DB, Rao PA, Malla RR. Enhanced efficacy of quercetin and taxifolin encapsulated with pH-responsive injectable BSA hydrogel for targeting triple-negative breast cancer cells. Int J Biol Macromol 2025; 287:138477. [PMID: 39667444 DOI: 10.1016/j.ijbiomac.2024.138477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Quercetin (QUE) and Taxifolin (TAX) are natural flavonoids with diverse biological activities, holding promise for cancer treatment. However, their clinical application is limited by their poor solubility and bioavailability. Self-assembled bovine serum albumin (BSA) hydrogels have demonstrated biocompatibility and proteolytic stability, making them suitable platforms for drug delivery. The present study validated the anticancer efficacy of QUE, TAX, and DOX encapsulated in BSA hydrogel (QUE@ BSA hydrogel, TAX@ BSA hydrogel, and DOX@ BSA hydrogel), which exhibited 93.5, 90 and 91.2 %% entrapment efficiency, respectively, and controlled release profiles with 90.8,95.8 and 90.8 % drug release, respectively, at lower pH using MDA-MB 231 and MDA-MB 468 TNBC cell lines. Characterization by SEM, XRD, FT-IR and DLS revealed distinctive features of QUE@ BSA hydrogel, TAX@ BSA hydrogel, and DOX@BSA hydrogels, suggesting potential for targeted drug delivery. Further, investigations showed that separate treatment with QUE@BSA hydrogel, TAX@BSA hydrogel, and DOX@BSA hydrogel disrupted cell membrane integrity, akin to inducing cytotoxicity with IC50 of 12.90, 15.52 and 6.9 μM, respectively, in MDA-MB 231 cells and 16.67, 19.16 and 5.2 μM, respectively, in MDA-MB 468 cells. Moreover, they reduced mammosphere formation and cell migration. Additionally, they induced cell cycle arrest, reduced cell proliferation, and induced apoptosis in TNBC cells. They also induced ROS generation and ER stress, highlighting their potential to suppress TNBC progression. Overall, this study underscores the promise of QUE@ BSA hydrogel and TAX@BSA hydrogel as effective anticancer agents against TNBC cell lines in line with DOX@BSA hydrogel, offering controlled drug release and enhanced therapeutic outcomes.
Collapse
Affiliation(s)
- Durga Bhavani Kundrapu
- Cancer Biology Laboratory, Dept of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Podilapu Atchutha Rao
- Dept of Chemistry, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Rama Rao Malla
- Cancer Biology Laboratory, Dept of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India.
| |
Collapse
|
4
|
Marni R, Malla M, Chakraborty A, Voonna MK, Bhattacharyya PS, Kgk D, Malla RR. Combination of ionizing radiation and 2-thio-6-azauridine induces cell death in radioresistant triple negative breast cancer cells by downregulating CD151 expression. Cancer Chemother Pharmacol 2024; 94:685-706. [PMID: 39167147 DOI: 10.1007/s00280-024-04709-w] [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: 06/12/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) represents the most aggressive subtype of breast cancer and is frequently resistant to therapy, ultimately resulting in treatment failure. Clinical trials have demonstrated the potential of sensitizing radiation therapy (RT)-resistant TNBC through the combination of chemotherapy and RT. This study sought to explore the potential of CD151 as a therapy response marker in the co-treatment strategy involving ionizing radiation (IR) and the repurposed antiviral drug 2-Thio-6-azauridine (TAU) for sensitizing RT-resistant TNBC (TNBC/RR). METHODS The investigation encompassed a variety of assessments, including viability using MTT and LDH assays, cell proliferation through BrdU incorporation and clonogenic assays, cell cycle analysis via flow cytometry, cell migration using wound scratch and Boyden chamber invasion assays, DNA damage assessment through γH2AX analysis, apoptosis evaluation through acridine-orange and ethidium bromide double staining assays, as well as caspase 3 activity measurement using a colorimetric assay. CD151 expression was examined through ELISA, flow cytometry and RT-qPCR. RESULTS The results showed a significant reduction in TNBC/RR cell viability following co-treatment. Moreover, the co-treatment reduced cell migration, induced apoptosis, downregulated CD151 expression, and increased caspase 3 activity in TNBC/RR cells. Additionally, CD151 was predicted to serve as a therapy response marker for co-treatment with TAU and IR. CONCLUSION These findings suggest the potential of combination treatment with IR and TAU as a promising strategy to overcome RT resistance in TNBC. Furthermore, CD151 emerges as a valuable therapy response marker for chemoradiotherapy.
Collapse
Affiliation(s)
- Rakshmitha Marni
- Cancer Biology Laboratory, Department of Life Sciences, GITAM (Deemed to Be University), GITAM School of Science, Visakhapatnam, 530045, A.P, India
| | - Manas Malla
- Department of Computer Science and Engineering, GITAM (Deemed to Be University), GITAM School of Technology, Visakhapatnam, 530045, A.P, India
| | | | - Murali Krishna Voonna
- Mahatma Gandhi Cancer Hospital & Research Institute, Visakhapatnam-, 530017, A.P, India
| | | | - Deepak Kgk
- Mahatma Gandhi Cancer Hospital & Research Institute, Visakhapatnam-, 530017, A.P, India
| | - Rama Rao Malla
- Cancer Biology Laboratory, Department of Life Sciences, GITAM (Deemed to Be University), GITAM School of Science, Visakhapatnam, 530045, A.P, India.
| |
Collapse
|
5
|
Tosi G, Paoli A, Zuccolotto G, Turco E, Simonato M, Tosoni D, Tucci F, Lugato P, Giomo M, Elvassore N, Rosato A, Cogo P, Pece S, Santoro MM. Cancer cell stiffening via CoQ 10 and UBIAD1 regulates ECM signaling and ferroptosis in breast cancer. Nat Commun 2024; 15:8214. [PMID: 39294175 PMCID: PMC11410950 DOI: 10.1038/s41467-024-52523-y] [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: 04/19/2023] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
CoQ10 (Coenzyme Q10) is an essential fat-soluble metabolite that plays a key role in cellular metabolism. A less-known function of CoQ10 is whether it may act as a plasma membrane-stabilizing agent and whether this property can affect cancer development and progression. Here, we show that CoQ10 and its biosynthetic enzyme UBIAD1 play a critical role in plasmamembrane mechanical properties that are of interest for breast cancer (BC) progression and treatment. CoQ10 and UBIAD1 increase membrane fluidity leading to increased cell stiffness in BC. Furthermore, CoQ10 and UBIAD1 states impair ECM (extracellular matrix)-mediated oncogenic signaling and reduce ferroptosis resistance in BC settings. Analyses on human patients and mouse models reveal that UBIAD1 loss is associated with BC development and progression and UBIAD1 expression in BC limits CTCs (circulating tumor cells) survival and lung metastasis formation. Overall, this study reveals that CoQ10 and UBIAD1 can be further investigated to develop therapeutic interventions to treat BC patients with poor prognosis.
Collapse
Affiliation(s)
- Giovanni Tosi
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Alessandro Paoli
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Turco
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Manuela Simonato
- Pediatric Research Institute "Città della Speranza", Padova, Italy
| | | | | | - Pietro Lugato
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy
| | - Monica Giomo
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Paola Cogo
- Pediatric Research Institute "Città della Speranza", Padova, Italy
- Division of Pediatrics, Department of Medicine, Udine University, Udine, Italy
| | - Salvatore Pece
- IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milano, Italy
| | - Massimo M Santoro
- Laboratory of Angiogenesis and Cancer Metabolism, Department of Biology, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
6
|
Gu L, Pillay RP, Aronson R, Kaur M. Cholesteryl ester transfer protein knock-down in conjunction with a cholesterol-depleting agent decreases tamoxifen resistance in breast cancer cells. IUBMB Life 2024; 76:712-730. [PMID: 38733508 DOI: 10.1002/iub.2823] [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: 02/05/2024] [Accepted: 03/25/2024] [Indexed: 05/13/2024]
Abstract
The cholesterogenic phenotype, encompassing de novo biosynthesis and accumulation of cholesterol, aids cancer cell proliferation and survival. Previously, the role of cholesteryl ester (CE) transfer protein (CETP) has been implicated in breast cancer aggressiveness, but the molecular basis of this observation is not clearly understood, which this study aims to elucidate. CETP knock-down resulted in a >50% decrease in cell proliferation in both 'estrogen receptor-positive' (ER+; Michigan Cancer Foundation-7 (MCF7) breast cancer cells) and 'triple-negative' breast cancer (TNBC; MDA-MB-231) cell lines. Intriguingly, the abrogation of CETP together with the combination treatment of tamoxifen (5 μM) and acetyl plumbagin (a cholesterol-depleting agent) (5 μM) resulted in twofold to threefold increase in apoptosis in both cell lines. CETP knockdown also showed decreased intracellular CE levels, lipid raft and lipid droplets in both cell lines. In addition, RT2 Profiler PCR array (Qiagen, Germany)-based gene expression analysis revealed an overall downregulation of genes associated in cholesterol biosynthesis, lipid signalling and drug resistance in MCF7 cells post-CETP knock-down. On the contrary, resistance in MDA-MB-231 cells was reduced through increased expression in cholesterol efflux genes and the expression of targetable surface receptors by endocrine therapy. The pilot xenograft mice study substantiated CETP's role as a cancer survival gene as knock-down of CETP stunted the growth of TNBC tumour by 86%. The principal findings of this study potentiate CETP as a driver in breast cancer growth and aggressiveness and thus targeting CETP could limit drug resistance via the reduction in cholesterol accumulation in breast cancer cells, thereby reducing cancer aggressiveness.
Collapse
Affiliation(s)
- Liang Gu
- Department of School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| | - Ruvesh Pascal Pillay
- Department of School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| | - Ruth Aronson
- Department of School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| | - Mandeep Kaur
- Department of School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
7
|
Xu Z, Li J, Fang S, Lian M, Zhang C, Lu J, Sheng K. Cinobufagin disrupts the stability of lipid rafts by inhibiting the expression of caveolin-1 to promote non-small cell lung cancer cell apoptosis. Arch Med Sci 2024; 20:887-908. [PMID: 39050162 PMCID: PMC11264083 DOI: 10.5114/aoms/174578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/27/2023] [Indexed: 07/27/2024] Open
Abstract
Introduction The study was designed to explore how cinobufagin (CB) regulates the development of non-small cell lung cancer (NSCLC) cells through lipid rafts. Material and methods The effects of CB at gradient concentrations (0, 0.5, 1 and 2 µM) on NSCLC cell viability, apoptosis, reactive oxygen species (ROS) level, phosphorylation of Akt, and apoptosis- and lipid raft-related protein expression were assessed by MTT assay, flow cytometry and Western blot. Cholesterol and sphingomyelin were labeled with BODIPY to evaluate the effect of CB (2 µM) on them. Sucrose density gradient centrifugation was used to extract lipid rafts. The effect of CB on the expression and distribution of caveolin-1 was determined by immunofluorescence, quantitative reverse transcription polymerase chain reaction and Western blot. After overexpression of caveolin-1, the above experiments were performed again to observe whether the regulatory effect of CB was reversed. Results CB inhibited NSCLC cell viability while promoting apoptosis and ROS level. CB redistributed the lipid content on the membrane surface and reduced the content of caveolin-1 in the cell membrane. In addition, CB repressed the activation of AKT. However, caveolin-1 overexpression reversed the effects of CB on apoptosis, AKT activation and lipid raft. Conclusions CB regulates the activity of Akt in lipid rafts by inhibiting caveolin-1 expression to promote NSCLC cell apoptosis.
Collapse
Affiliation(s)
- Zhongqing Xu
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinwei Li
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuyu Fang
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Lian
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changxiao Zhang
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahuan Lu
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Sheng
- Department of Gerontology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Paba C, Dorigo V, Senigagliesi B, Tormena N, Parisse P, Voitchovsky K, Casalis L. Lipid bilayer fluidity and degree of order regulates small EVs adsorption on model cell membrane. J Colloid Interface Sci 2023; 652:1937-1943. [PMID: 37690301 DOI: 10.1016/j.jcis.2023.08.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/09/2023] [Accepted: 08/19/2023] [Indexed: 09/12/2023]
Abstract
Small extracellular vesicles (sEVs) are known to play an important role in the communication between distant cells and to deliver biological information throughout the body. To date, many studies have focused on the role of sEVs characteristics such as cell origin, surface composition, and molecular cargo on the resulting uptake by the recipient cell. Yet, a full understanding of the sEV fusion process with recipient cells and in particular the role of cell membrane physical properties on the uptake are still lacking. Here we explore this problem using sEVs from a cellular model of triple-negative breast cancer fusing to a range of synthetic planar lipid bilayers both with and without cholesterol, and designed to mimic the formation of 'raft'-like nanodomains in cell membranes. Using time-resolved Atomic Force Microscopy we were able to track the sEVs interaction with the different model membranes, showing the process to be strongly dependent on the local membrane fluidity. The strongest interaction and fusion is observed over the less fluid regions, with sEVs even able to disrupt ordered domains at sufficiently high cholesterol concentration. Our findings suggest the biophysical characteristics of recipient cell membranes to be crucial for sEVs uptake regulation.
Collapse
Affiliation(s)
- Carolina Paba
- Department of Physics, University of Trieste, 34127 Trieste, Italy
| | | | | | - Nicolò Tormena
- Department of Physics, University of Durham, Durham DH1 3LE, United Kingdom
| | - Pietro Parisse
- Elettra Sincrotrone Trieste, 34149 Basovizza TS, Italy; IOM-CNR, 34149 Basovizza TS, Italy.
| | - Kislon Voitchovsky
- Department of Physics, University of Durham, Durham DH1 3LE, United Kingdom.
| | | |
Collapse
|
9
|
Li D, Park Y, Hemati H, Liu X. Cell aggregation activates small GTPase Rac1 and induces CD44 cleavage by maintaining lipid raft integrity. J Biol Chem 2023; 299:105377. [PMID: 37866630 PMCID: PMC10692920 DOI: 10.1016/j.jbc.2023.105377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/13/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Lipid rafts are highly ordered membrane domains that are enriched in cholesterol and glycosphingolipids and serve as major platforms for signal transduction. Cell detachment from the extracellular matrix (ECM) triggers lipid raft disruption and anoikis, which is a barrier for cancer cells to metastasize. Compared to single circulating tumor cells (CTCs), our recent studies have demonstrated that CD44-mediatd cell aggregation enhances the stemness, survival and metastatic ability of aggregated cells. Here, we investigated whether and how lipid rafts are involved in CD44-mediated cell aggregation. We found that cell detachment, which mimics the condition when tumor cells detach from the ECM to metastasize, induced lipid raft disruption in single cells, but lipid raft integrity was maintained in aggregated cells. We further found that lipid raft integrity in aggregated cells was required for Rac1 activation to prevent anoikis. In addition, CD44 and γ-secretase coexisted at lipid rafts in aggregated cells, which promoted CD44 cleavage and generated CD44 intracellular domain (CD44 ICD) to enhance stemness of aggregated cells. Consequently, lipid raft disruption inhibited Rac1 activation, CD44 ICD generation, and metastasis. Our findings reveal two new pathways regulated by CD44-mediated cell aggregation via maintaining lipid raft integrity. These findings also suggest that targeting cell aggregation-mediated pathways could be a novel therapeutic strategy to prevent CTC cluster-initiated metastasis.
Collapse
Affiliation(s)
- Dong Li
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Younhee Park
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Hami Hemati
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Xia Liu
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA.
| |
Collapse
|
10
|
Bhowmick S, Biswas T, Ahmed M, Roy D, Mondal S. Caveolin-1 and lipids: Association and their dualism in oncogenic regulation. Biochim Biophys Acta Rev Cancer 2023; 1878:189002. [PMID: 37848094 DOI: 10.1016/j.bbcan.2023.189002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023]
Abstract
Caveolin-1 (Cav-1) is a structural protein of caveolae that functions as a molecular organizer for different cellular functions including endocytosis and cellular signaling. Cancer cells take advantage of the physical position of Cav-1, as it can communicate with extracellular matrix, help to organize growth factor receptors, redistribute cholesterol and glycosphingolipids, and finally transduce signals within the cells for oncogenesis. Recent studies emphasize the exceeding involvement of Cav-1 with different lipid bodies and in altering the metabolism, especially lipid metabolism. However, the association of Cav-1 with different lipid bodies like lipid rafts, lipid droplets, cholesterols, sphingolipids, and fatty acids is remarkably dynamic. The lipid-Cav-1 alliance plays a dual role in carcinogenesis. Both cancer progression and regression are modified and affected by the type of lipid molecule's association with Cav-1. Accordingly, this Cav-1-lipid cooperation exemplifies a cancer-type-specific treatment strategy for a better prognosis of the disease. In this review, we first present Cav-1 as an oncogenic molecule and its communication via lipid raft. We discussed the involvement of Cav-1 with lipid droplets, Cholesterol, sphingolipids, gangliosides, and ceramides. Further, we describe the Cav-1-mediated altered Fatty acid metabolism in cancer and the strategic therapeutic approaches toward Cav-1 targeting.
Collapse
Affiliation(s)
- Sramana Bhowmick
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Tannishtha Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Mehnaz Ahmed
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Debarshi Roy
- Department of Biological Sciences, Alcorn State University, Lorman, MS 39096, USA
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
11
|
Kuburich NA, Sabapathy T, Demestichas BR, Maddela JJ, den Hollander P, Mani SA. Proactive and reactive roles of TGF-β in cancer. Semin Cancer Biol 2023; 95:120-139. [PMID: 37572731 PMCID: PMC10530624 DOI: 10.1016/j.semcancer.2023.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/14/2023]
Abstract
Cancer cells adapt to varying stress conditions to survive through plasticity. Stem cells exhibit a high degree of plasticity, allowing them to generate more stem cells or differentiate them into specialized cell types to contribute to tissue development, growth, and repair. Cancer cells can also exhibit plasticity and acquire properties that enhance their survival. TGF-β is an unrivaled growth factor exploited by cancer cells to gain plasticity. TGF-β-mediated signaling enables carcinoma cells to alter their epithelial and mesenchymal properties through epithelial-mesenchymal plasticity (EMP). However, TGF-β is a multifunctional cytokine; thus, the signaling by TGF-β can be detrimental or beneficial to cancer cells depending on the cellular context. Those cells that overcome the anti-tumor effect of TGF-β can induce epithelial-mesenchymal transition (EMT) to gain EMP benefits. EMP allows cancer cells to alter their cell properties and the tumor immune microenvironment (TIME), facilitating their survival. Due to the significant roles of TGF-β and EMP in carcinoma progression, it is essential to understand how TGF-β enables EMP and how cancer cells exploit this plasticity. This understanding will guide the development of effective TGF-β-targeting therapies that eliminate cancer cell plasticity.
Collapse
Affiliation(s)
- Nick A Kuburich
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Thiru Sabapathy
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Breanna R Demestichas
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Joanna Joyce Maddela
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Petra den Hollander
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Sendurai A Mani
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
12
|
Kciuk M, Alam M, Ali N, Rashid S, Głowacka P, Sundaraj R, Celik I, Yahya EB, Dubey A, Zerroug E, Kontek R. Epigallocatechin-3-Gallate Therapeutic Potential in Cancer: Mechanism of Action and Clinical Implications. Molecules 2023; 28:5246. [PMID: 37446908 PMCID: PMC10343677 DOI: 10.3390/molecules28135246] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Cellular signaling pathways involved in the maintenance of the equilibrium between cell proliferation and apoptosis have emerged as rational targets that can be exploited in the prevention and treatment of cancer. Epigallocatechin-3-gallate (EGCG) is the most abundant phenolic compound found in green tea. It has been shown to regulate multiple crucial cellular signaling pathways, including those mediated by EGFR, JAK-STAT, MAPKs, NF-κB, PI3K-AKT-mTOR, and others. Deregulation of the abovementioned pathways is involved in the pathophysiology of cancer. It has been demonstrated that EGCG may exert anti-proliferative, anti-inflammatory, and apoptosis-inducing effects or induce epigenetic changes. Furthermore, preclinical and clinical studies suggest that EGCG may be used in the treatment of numerous disorders, including cancer. This review aims to summarize the existing knowledge regarding the biological properties of EGCG, especially in the context of cancer treatment and prophylaxis.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (M.K.); (R.K.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Pola Głowacka
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 90-001 Lodz, Poland;
- Doctoral School of Medical University of Lodz, Hallera 1 Square, 90-700 Lodz, Poland
| | - Rajamanikandan Sundaraj
- Department of Biochemistry, Centre for Drug Discovery, Karpagam Academy of Higher Education, Coimbatore 641021, India;
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38280, Turkey;
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia;
| | - Amit Dubey
- Computational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida 201310, India;
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Chennai 600077, India
| | - Enfale Zerroug
- LMCE Laboratory, Group of Computational and Pharmaceutical Chemistry, University of Biskra, Biskra 07000, Algeria;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (M.K.); (R.K.)
| |
Collapse
|
13
|
Alboni S, Secco V, Papotti B, Vilella A, Adorni MP, Zimetti F, Schaeffer L, Tascedda F, Zoli M, Leblanc P, Villa E. Hydroxypropyl-β-Cyclodextrin Depletes Membrane Cholesterol and Inhibits SARS-CoV-2 Entry into HEK293T-ACE hi Cells. Pathogens 2023; 12:pathogens12050647. [PMID: 37242317 DOI: 10.3390/pathogens12050647] [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: 03/14/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Vaccination has drastically decreased mortality due to coronavirus disease 19 (COVID-19), but not the rate of acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Alternative strategies such as inhibition of virus entry by interference with angiotensin-I-converting enzyme 2 (ACE2) receptors could be warranted. Cyclodextrins (CDs) are cyclic oligosaccharides that are able to deplete cholesterol from membrane lipid rafts, causing ACE2 receptors to relocate to areas devoid of lipid rafts. To explore the possibility of reducing SARS-CoV-2 entry, we tested hydroxypropyl-β-cyclodextrin (HPβCD) in a HEK293T-ACE2hi cell line stably overexpressing human ACE2 and Spike-pseudotyped SARS-CoV-2 lentiviral particles. We showed that HPβCD is not toxic to the cells at concentrations up to 5 mM, and that this concentration had no significant effect on cell cycle parameters in any experimental condition tested. Exposure of HEK293T-ACEhi cells to concentrations of HPβCD starting from 2.5 mM to 10 mM showed a concentration-dependent reduction of approximately 50% of the membrane cholesterol content. In addition, incubation of HEK293T-ACEhi cells with HIV-S-CoV-2 pseudotyped particles in the presence of increasing concentrations of HPβCD (from 0.1 to 10 mM) displayed a concentration-dependent effect on SARS-CoV-2 entry efficiency. Significant effects were detected at concentrations at least one order of magnitude lower than the lowest concentration showing toxic effects. These data indicate that HPβCD is a candidate for use as a SARS-CoV-2 prophylactic agent.
Collapse
Affiliation(s)
- Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Centre for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Valentina Secco
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Antonietta Vilella
- Centre for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Laurent Schaeffer
- Institut NeuroMyoGène INMG-PGNM Pathophysiologie & Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, 69008 Lyon, France
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Centre for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Consorzio Interuniversitario Biotecnologie (CIB), 34148 Trieste, Italy
| | - Michele Zoli
- Centre for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Pascal Leblanc
- Institut NeuroMyoGène INMG-PGNM Pathophysiologie & Génétique du Neurone et du Muscle, UMR5261, Inserm U1315, 69008 Lyon, France
| | - Erica Villa
- CHIMOMO Department, University of Modena and Reggio Emilia, and Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy
| |
Collapse
|
14
|
Li D, Park Y, Hemati H, Liu X. Cell aggregation prevents anoikis and induces CD44 cleavage by maintaining lipid raft integrity to promote triple negative breast cancer metastasis. RESEARCH SQUARE 2023:rs.3.rs-2535728. [PMID: 36824757 PMCID: PMC9949249 DOI: 10.21203/rs.3.rs-2535728/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, and metastasis is the major cause of cancer morbidity and mortality. Therefore, it is urgent to discover novel therapeutic targets and develop effective treatments for this lethal disease. Circulating tumor cells (CTCs) are considered "seeds of metastasis". Compared to single CTCs, our previous studies have demonstrated that CD44 homophilic interaction mediates CTC aggregation to enhance the stemness, survival and metastatic ability of aggregated cells. Importantly, the presence of CD44+ CTC clusters correlates with a poor prognosis in breast cancer patients. Here, we further investigated the underlying mechanism of how CD44-mediated cell aggregation promotes TNBC metastasis. We found that cell detachment, which mimics the condition when tumor cells detach from the extracellular matrix (ECM) to metastasize, induces lipid raft disruption in single cells, but lipid rafts integrity is maintained in aggregated cells. We further found that lipid rafts integrity in aggregated cells is required for Rac1 activation to prevent anoikis. In addition, CD44 and γ-secretase coexisted at lipid rafts in aggregated cells, which promotes CD44 cleavage and generates CD44 intracellular domain (CD44 ICD) to enhance stemness. Consequently, lipid rafts disruption inhibited Rac1 activation, CD44 ICD generation and metastasis. These data reveal a new mechanism of cell aggregation-mediated TNBC metastasis via maintaining lipid raft integrity after cell detachment. The finding provides a potential therapeutic strategy to prevent CTC cluster-initiated metastasis by disrupting lipid raft integrity and its-mediated downstream pathways.
Collapse
|
15
|
Zhang L, Wang Y, Chen P, Wang D, Sun T, Zhang Z, Wang R, Kang X, Fang Y, Lu H, Cai J, Ren M, Dong SS, Zhang K. A mechanistic study on the cellular uptake, intracellular trafficking, and antisense gene regulation of bottlebrush polymer-conjugated oligonucleotides. RSC Chem Biol 2023; 4:138-145. [PMID: 36794022 PMCID: PMC9906284 DOI: 10.1039/d2cb00149g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
We have developed a non-cationic transfection vector in the form of bottlebrush polymer-antisense oligonucleotide (ASO) conjugates. Termed pacDNA (polymer-assisted compaction of DNA), these agents show improved biopharmaceutical characteristics and antisense potency in vivo while suppressing non-antisense side effects. Nonetheless, there still is a lack of the mechanistic understanding of the cellular uptake, subcellular trafficking, and gene knockdown with pacDNA. Here, we show that the pacDNA enters human non-small cell lung cancer cells (NCI-H358) predominantly by scavenger receptor-mediated endocytosis and macropinocytosis and trafficks via the endolysosomal pathway within the cell. The pacDNA significantly reduces a target gene expression (KRAS) in the protein level but not in the mRNA level, despite that the transfection of certain free ASOs causes ribonuclease H1 (RNase H)-dependent degradation of KRAS mRNA. In addition, the antisense activity of pacDNA is independent of ASO chemical modification, suggesting that the pacDNA always functions as a steric blocker.
Collapse
Affiliation(s)
- Lei Zhang
- Chemicobiology and Functional Materials Institute, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology Nanjing 210094 P. R. China
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Yuyan Wang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Peiru Chen
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Dali Wang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Tingyu Sun
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Zheyu Zhang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Ruimeng Wang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Xi Kang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Yang Fang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Hao Lu
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Jiansong Cai
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Mengqi Ren
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Sijia S Dong
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
| | - Ke Zhang
- Department of Chemistry and Chemical Biology, Northeastern University Boston Massachusetts 02115 USA
- Departments of Chemical Engineering and Bioengineering, Northeastern University Boston Massachusetts 02115 USA
| |
Collapse
|
16
|
Luan H, Bielecki TA, Mohapatra BC, Islam N, Mushtaq I, Bhat AM, Mirza S, Chakraborty S, Raza M, Storck MD, Toss MS, Meza JL, Thoreson WB, Coulter DW, Rakha EA, Band V, Band H. EHD2 overexpression promotes tumorigenesis and metastasis in triple-negative breast cancer by regulating store-operated calcium entry. eLife 2023; 12:81288. [PMID: 36625722 PMCID: PMC9988264 DOI: 10.7554/elife.81288] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 01/10/2023] [Indexed: 01/11/2023] Open
Abstract
With nearly all cancer deaths a result of metastasis, elucidating novel pro-metastatic cellular adaptations could provide new therapeutic targets. Here, we show that overexpression of the EPS15-Homology Domain-containing 2 (EHD2) protein in a large subset of breast cancers (BCs), especially the triple-negative (TNBC) and HER2+ subtypes, correlates with shorter patient survival. The mRNAs for EHD2 and Caveolin-1/2, structural components of caveolae, show co-overexpression across breast tumors, predicting shorter survival in basal-like BC. EHD2 shRNA knockdown and CRISPR-Cas9 knockout with mouse Ehd2 rescue, in TNBC cell line models demonstrate a major positive role of EHD2 in promoting tumorigenesis and metastasis. Mechanistically, we link these roles of EHD2 to store-operated calcium entry (SOCE), with EHD2-dependent stabilization of plasma membrane caveolae ensuring high cell surface expression of the SOCE-linked calcium channel Orai1. The novel EHD2-SOCE oncogenic axis represents a potential therapeutic target in EHD2- and CAV1/2-overexpressing BC.
Collapse
Affiliation(s)
- Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
| | - Bhopal C Mohapatra
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Namista Islam
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
- Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| | - Aaqib M Bhat
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| | - Sameer Mirza
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| | - Sukanya Chakraborty
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| | - Mohsin Raza
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
| | - Michael S Toss
- Department of Histopathology, Nottingham University Hospital NHS Trust, City Hospital CampusNottinghamUnited Kingdom
| | - Jane L Meza
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
- Department of Biostatistics, College of Public Health, University of Nebraska Medical CenterOmahaUnited States
| | - Wallace B Thoreson
- Stanley M. Truhlsen Eye Institute, University of Nebraska Medical CenterOmahaUnited States
| | - Donald W Coulter
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
- Department of Pediatrics, University of Nebraska Medical CenterOmahaUnited States
| | - Emad A Rakha
- Department of Histopathology, Nottingham University Hospital NHS Trust, City Hospital CampusNottinghamUnited Kingdom
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical CenterOmahaUnited States
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmahaUnited States
- Department of Pathology & Microbiology, College of Medicine, University of Nebraska Medical CenterOmahaUnited States
| |
Collapse
|
17
|
Sha YL, Liu Y, Yang JX, Wang YY, Gong BC, Jin Y, Qu TY, Xia FT, Han L, Zhao Q. B3GALT4 remodels the tumor microenvironment through GD2-mediated lipid raft formation and the c-met/AKT/mTOR/IRF-1 axis in neuroblastoma. J Exp Clin Cancer Res 2022; 41:314. [PMID: 36284313 PMCID: PMC9594894 DOI: 10.1186/s13046-022-02523-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Beta-1,3-galactosyltransferase-4 (B3GALT4) plays a critical regulatory role in tumor biology. However, the role of B3GALT4 in modulating the tumor microenvironment (TME) of neuroblastoma (NB) remains unknown. METHODS Public datasets and clinical NB samples were collected to evaluate the expression and clinical significance of GD2 and B3GALT4 in NB patients. CCK-8, colony formation, and transwell assays and experiments in tumor-bearing mouse models were conducted to investigate the function of B3GALT4. Flow cytometry, ELISA, immunohistochemistry, immunofluorescence, western blotting, and chemotaxis assays were conducted to ascertain the immunomodulatory mechanism of B3GALT4. The combined therapeutic effect of the lipid raft inhibitor MβCD and anti-GD2 mAb was validated in a murine model of NB. RESULTS GD2 was overexpressed in NB tissues and high expression of GD2 was associated with poor prognosis in NB patients. B3GALT4 was downregulated in NB tissues, and low expression of B3GALT4 indicated poor prognosis in NB patients. Silencing B3GALT4 significantly enhanced tumor progression both in vitro and in vivo. Meanwhile, the overexpression of B3GALT4 increased the recruitment of CD8+ T lymphocytes via the chemokines CXCL9 and CXCL10. Additionally, B3GALT4 regulated NB-cell GD2 expression and lipid raft formation. Mechanistically, B3GALT4 regulated the expression of CXCL9 and CXCL10 via the c-Met signaling in the lipid rafts and the downstream AKT/mTOR/IRF-1 pathway. The lipid raft inhibitor, MβCD, attenuated B3GALT4 deficiency-induced tumor progression and immune evasion. Last, MβCD combined with anti-GD2 mAb treatment significantly enhanced the antitumor effect and the infiltration of CD8+ T cells. CONCLUSIONS Upregulation of B3GALT4 promotes the secretion of CXCL9 and CXCL10 to recruit CD8+ T lymphocytes via the GD2-mediated lipid rafts and the c-Met/AKT/mTOR/IRF-1 pathway. Moreover, lipid raft inhibitors may enhance the efficacy of anti-GD2 immunotherapy for NB.
Collapse
Affiliation(s)
- Yong-Liang Sha
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jia-Xing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yang-Yang Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bao-Cheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tong-Yuan Qu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Fan-Tong Xia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Han
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
18
|
Marni R, Kundrapu DB, Chakraborti A, Malla R. Insight into drug sensitizing effect of diallyl disulfide and diallyl trisulfide from Allium sativum L. on paclitaxel-resistant triple-negative breast cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115452. [PMID: 35690339 DOI: 10.1016/j.jep.2022.115452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ayurvedic practitioners and herbal healers in India and China have extensively used garlic (Allium sativum L.) to treat cancers. Diallyl disulfide (DADS) and diallyl trisulfide (DATS) are major volatile organosulfur phytochemical constituents found in garlic. AIM OF THE STUDY To find new insight into the drug sensitizing effect of DADS and DATS on paclitaxel (PTX)-resistant triple-negative breast cancer cells (TNBC/PR). MATERIALS AND METHODS This study estimates the non-toxic concentration of DADS and DATS against normal healthy breast epithelial cell line (MCF-12A) by using a trypan blue viability assay. Also, it evaluates the effect of DADS and DATS on the sensitization of established stable TNBC/PR cell clones (MDA-MB 231 PR and MDA-MB 468 PR) by MTT, BrdU incorporation, intracellular ROS, cell cycle, and apoptosis assays. RESULTS The results show that DADS and DATS are non-cytotoxicity against MCF-12A cells. Nevertheless, DADS and DATS have shown significantly high cytotoxicity against MDA-MB 231 PR and MDA-MB 468 PR cells. They also inhibited PTX-resistant cell proliferation by blocking the cell cycle. Further, they induced apoptosis by activation of caspase 3 and 9. N-acetyl cysteine pre-treatment inhibited DADS and DATS-induced intracellular ROS release. In silico study shows that DADS and DATS interact with a large extracellular loop (LEL) of CD151 with a binding energy of -4.0 kcal/mol and transmembrane domain (TM) with a binding affinity of 11.7 and 13.6 kcal/mol, respectively. They also inhibited the surface expression of CD151 in TNBC/PR cells. CONCLUSION This study implies that DADS and DATS could be considered for sensitizing drug-resistant breast cancers.
Collapse
Affiliation(s)
- Rakshmitha Marni
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, A.P, India.
| | - Durga Bhavani Kundrapu
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, A.P, India.
| | | | - RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, A.P, India.
| |
Collapse
|
19
|
Centonze G, Natalini D, Piccolantonio A, Salemme V, Morellato A, Arina P, Riganti C, Defilippi P. Cholesterol and Its Derivatives: Multifaceted Players in Breast Cancer Progression. Front Oncol 2022; 12:906670. [PMID: 35719918 PMCID: PMC9204587 DOI: 10.3389/fonc.2022.906670] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol is an essential lipid primarily synthesized in the liver through the mevalonate pathway. Besides being a precursor of steroid hormones, bile acid, and vitamin D, it is an essential structural component of cell membranes, is enriched in membrane lipid rafts, and plays a key role in intracellular signal transduction. The lipid homeostasis is finely regulated end appears to be impaired in several types of tumors, including breast cancer. In this review, we will analyse the multifaceted roles of cholesterol and its derivatives in breast cancer progression. As an example of the bivalent role of cholesterol in the cell membrane of cancer cells, on the one hand, it reduces membrane fluidity, which has been associated with a more aggressive tumor phenotype in terms of cell motility and migration, leading to metastasis formation. On the other hand, it makes the membrane less permeable to small water-soluble molecules that would otherwise freely cross, resulting in a loss of chemotherapeutics permeability. Regarding cholesterol derivatives, a lower vitamin D is associated with an increased risk of breast cancer, while steroid hormones, coupled with the overexpression of their receptors, play a crucial role in breast cancer progression. Despite the role of cholesterol and derivatives molecules in breast cancer development is still controversial, the use of cholesterol targeting drugs like statins and zoledronic acid appears as a challenging promising tool for breast cancer treatment.
Collapse
Affiliation(s)
- Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy
| | - Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy
| | - Pietro Arina
- University College London (UCL), Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Chiara Riganti
- Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy.,Department of Oncology, University of Torino, Torino, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Interdepartmental Center of Research in Molecular Biotechnology, University of Torino, Torino, Italy
| |
Collapse
|
20
|
Microfluidic-Based Cationic Cholesterol Lipid siRNA Delivery Nanosystem: Highly Efficient In Vitro Gene Silencing and the Intracellular Behavior. Int J Mol Sci 2022; 23:ijms23073999. [PMID: 35409359 PMCID: PMC8999516 DOI: 10.3390/ijms23073999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/04/2022] Open
Abstract
Safe and efficient delivery of small interfering RNA (siRNA) is essential to gene therapy towards intervention of genetic diseases. Herein, we developed a novel cationic cholesterol lipid derivative (CEL) in which cholesterol hydrophobic skeleton was connected to L-lysine cationic headgroup via a hexanediol linker as the non-viral siRNA delivery carrier. Well-organized CEL/siRNA nanocomplexes (100-200 nm) were prepared by microfluidic-assisted assembly of CEL and siRNA at various N/P ratios. The CEL and CEL/siRNA nanocomplexes have lower cytotoxicity compared with bPEI25k. Delightfully, we disclosed that, in Hela-Luc and H1299-Luc cell lines, the micro-fluidic-based CEL/siRNA nanocomplexes exhibited high siRNA transfection efficiency under both serum-free condition (74-98%) and low-serum circumstances (80-87%), higher than that of lipofectamine 2000. These nanocomplexes also showed high cellular uptake through the caveolae/lipid-raft mediated endocytosis pathway, which may greatly contribute to transfection efficiency. Moreover, the time-dependent (0-12 h) dynamic intracellular imaging demonstrated the efficient delivery to cytoplasm after lysosomal co-localization. The results indicated that the microfluidic-based CEL/siRNA nanosystems possessed good stability, low cytotoxicity, high siRNA delivery efficiency, rapid cellular uptake and caveolae/lipid raft-dependent internalization. Additionally, this study provides a simple approach for preparing and applying a "helper lipid-free" cationic lipid siRNA delivery system as potential nanotherapeutics towards gene silencing treatment of (tumor) diseases.
Collapse
|
21
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles: Pleiotropic Impacts on Breast Cancer Occurrence, Development, and Therapy. Int J Mol Sci 2022; 23:ijms23062927. [PMID: 35328347 PMCID: PMC8954385 DOI: 10.3390/ijms23062927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 01/27/2023] Open
Abstract
Breast cancer (BC) is one of the most devastating cancers, with high morbidity and mortality, among the female population worldwide. In BC, mesenchymal stem cells (MSCs), as pluripotent stromal stem cells, play a significant role in TME formation and tumor progression. Recently, an increasing number of studies have demonstrated that extracellular vesicles (EVs) are essential for the crosstalk between MSCs and BC cells. MSC-derived EVs (MSC-EVs) can deliver a diversity of molecules, including lipids, proteins, and nucleic acids, etc., to target cells, and produce corresponding effects. Studies have demonstrated that MSC-EVs exert both inhibitory and promotive effects in different situations and different stages of BC. Meanwhile, MSC-EVs provide novel therapeutic options for BC, such as EVs as carriers for drug delivery. Therefore, in this review, we summarize the role of MSC-EVs in BC progression and application in clinical treatment, in the hope of providing a basis for further research.
Collapse
|
22
|
Behzadi M, Amininasab M, Eghtedardoost M, Bagheri M. Turn-folded magainin lipopeptide analog induces cytoplasmic vacuoles in MDA-MB-231 cells through G2-phase arrest. Biochem Biophys Res Commun 2021; 583:199-205. [PMID: 34752987 DOI: 10.1016/j.bbrc.2021.10.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/30/2021] [Indexed: 02/04/2023]
Abstract
Selective induced non-canonical programmed deaths in the lipid raft type 1-enriched MDA-MB-231 is a promising treatment approach. Cationic amphiphilic peptides conjugated to relatively long fatty acyl chains that tend to self-aggregate are prone to upregulate necroptotic and paraptotic signaling. We investigated the toxic effects of an N-terminally palmitoylated magainin derivate (P1MK5E) in the MDA-MB-231 cells in relation to its structure at molecular level. The modeling showed that the palmitoylation reinforces a turn-like structural motif in the lipopeptide which is likely required for its activity. P1MK5E triggered intracellular generation of reactive oxygen species (ROS), G2-phase arrest, mitochondrial membrane potential (ΔΨmt) disturbance and presumable flopping of phosphatidylserine (PtdSer) to the cancer cell membrane outer surface in a comparable manner to doxorubicin (DOX) that induces apoptotic signaling. Despite forming extensive congregates of different sizes at the cell surface, P1MK5E had little impacts on the MDA-MB-231 membrane integrity. The cell death upon exposure to the lipopeptide was, however, caspase 3 independent and characterized by cytoplasmic vacuolation and no distinct nuclear fragmentation that is to be privileged in the treatment of apoptotic resistance pathways in triple-negative breast cancers (TNBCs).
Collapse
Affiliation(s)
- Malihe Behzadi
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335, Tehran, Iran
| | - Mehriar Amininasab
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, 16 Azar Street, 14174-66191, Tehran, Iran
| | - Marzieh Eghtedardoost
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-111, Tehran, Tehran, Iran
| | - Mojtaba Bagheri
- Peptide Chemistry Laboratory, Institute of Biochemistry and Biophysics, University of Tehran, 16 Azar Street, 14176-14335, Tehran, Iran.
| |
Collapse
|
23
|
Scully T, Ettela A, LeRoith D, Gallagher EJ. Obesity, Type 2 Diabetes, and Cancer Risk. Front Oncol 2021; 10:615375. [PMID: 33604295 PMCID: PMC7884814 DOI: 10.3389/fonc.2020.615375] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and type 2 diabetes have both been associated with increased cancer risk and are becoming increasingly prevalent. Metabolic abnormalities such as insulin resistance and dyslipidemia are associated with both obesity and type 2 diabetes and have been implicated in the obesity-cancer relationship. Multiple mechanisms have been proposed to link obesity and diabetes with cancer progression, including an increase in insulin/IGF-1 signaling, lipid and glucose uptake and metabolism, alterations in the profile of cytokines, chemokines, and adipokines, as well as changes in the adipose tissue directly adjacent to the cancer sites. This review aims to summarize and provide an update on the epidemiological and mechanistic evidence linking obesity and type 2 diabetes with cancer, focusing on the roles of insulin, lipids, and adipose tissue.
Collapse
Affiliation(s)
- Tiffany Scully
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Abora Ettela
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Emily Jane Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
24
|
Greenlee JD, Subramanian T, Liu K, King MR. Rafting Down the Metastatic Cascade: The Role of Lipid Rafts in Cancer Metastasis, Cell Death, and Clinical Outcomes. Cancer Res 2021; 81:5-17. [PMID: 32999001 PMCID: PMC7952000 DOI: 10.1158/0008-5472.can-20-2199] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/01/2020] [Accepted: 09/21/2020] [Indexed: 11/16/2022]
Abstract
Lipid rafts are tightly packed, cholesterol- and sphingolipid-enriched microdomains within the plasma membrane that play important roles in many pathophysiologic processes. Rafts have been strongly implicated as master regulators of signal transduction in cancer, where raft compartmentalization can promote transmembrane receptor oligomerization, shield proteins from enzymatic degradation, and act as scaffolds to enhance intracellular signaling cascades. Cancer cells have been found to exploit these mechanisms to initiate oncogenic signaling and promote tumor progression. This review highlights the roles of lipid rafts within the metastatic cascade, specifically within tumor angiogenesis, cell adhesion, migration, epithelial-to-mesenchymal transition, and transendothelial migration. In addition, the interplay between lipid rafts and different modes of cancer cell death, including necrosis, apoptosis, and anoikis, will be described. The clinical role of lipid raft-specific proteins, caveolin and flotillin, in assessing patient prognosis and evaluating metastatic potential of various cancers will be presented. Collectively, elucidation of the complex roles of lipid rafts and raft components within the metastatic cascade may be instrumental for therapeutic discovery to curb prometastatic processes.
Collapse
Affiliation(s)
- Joshua D Greenlee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Tejas Subramanian
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Kevin Liu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
25
|
Soteriou C, Kalli AC, Connell SD, Tyler AII, Thorne JL. Advances in understanding and in multi-disciplinary methodology used to assess lipid regulation of signalling cascades from the cancer cell plasma membrane. Prog Lipid Res 2020; 81:101080. [PMID: 33359620 DOI: 10.1016/j.plipres.2020.101080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/31/2022]
Abstract
The lipid bilayer is a functional component of cells, forming a stable platform for the initiation of key biological processes, including cell signalling. There are distinct changes in the lipid composition of cell membranes during oncogenic transformation resulting in aberrant activation and inactivation of signalling transduction pathways. Studying the role of the cell membrane in cell signalling is challenging, since techniques are often limited to by timescale, resolution, sensitivity, and averaging. To overcome these limitations, combining 'computational', 'wet-lab' and 'semi-dry' approaches offers the best opportunity to resolving complex biological processes involved in membrane organisation. In this review, we highlight analytical tools that have been applied for the study of cell signalling initiation from the cancer cell membranes through computational microscopy, biological assays, and membrane biophysics. The cancer therapeutic potential of extracellular membrane-modulating agents, such as cholesterol-reducing agents is also discussed, as is the need for future collaborative inter-disciplinary research for studying the role of the cell membrane and its components in cancer therapy.
Collapse
Affiliation(s)
- C Soteriou
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK; Leeds Institute of Cardiovascular and Metabolic Medicine and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK; Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | - A C Kalli
- Leeds Institute of Cardiovascular and Metabolic Medicine and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - S D Connell
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | - A I I Tyler
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK
| | - J L Thorne
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK.
| |
Collapse
|
26
|
Bobin-Dubigeon C, Bard JM, Luu TH, Le Vacon F, Nazih H. Basolateral Secretion from Caco-2 Cells Pretreated with Fecal Waters from Breast Cancer Patients Affects MCF7 Cell Viability. Nutrients 2020; 13:nu13010031. [PMID: 33374116 PMCID: PMC7824055 DOI: 10.3390/nu13010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 11/26/2022] Open
Abstract
We hypothesized that the role of microbiota in breast cancer relates to its influence on gut lipid metabolism. This was tested in an in vitro model combining MCF-7 and Caco-2 cells. A total of 32 women newly diagnosed for breast cancer before any treatment and 28 healthy women provided their stools. Bacterial DNA was amplified by qPCR targeting 16s rRNA specific to Bacteroidetes and Firmicutes phyla, Lactobacillales sp., Clostridium cluster IV, Faecalibacterium prausnitzii, Clostridium cluster XIVa, Roseburia intestinalis, Blautia sp., Lactonifactor longoviformis, Bifidobacterium sp., Coriobacteriaceae, Eggertella lenta, Escherichia, and Shigella. Fecal waters (FW) were quantified for short chain fatty acids (SCFA). Caco-2 cells grown on filter inserts were incubated apically with 10% FW for 24 h, and LXR, apolipoproteins AIV, and E gene expression were estimated by real time (RT) qPCR. Then, MCF-7 cells were incubated with the whole basolateral medium for 24 h, and their viability was estimated by 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide (MTT) test. Regression models were used to determine the correlation between MCF-7 viability and bacteria relative abundance, Caco-2 cells lipid metabolism gene expression and stool composition, as well as microbiota composition and short chain fatty acids. Logistic regression models established disease odds ratios (OR) for MCF-7 viability and Caco-2 gene expression. The OR of MCF-7 viability was 1.05 (1.01–1.10) (OR (5th–95th), p = 0.04), while that of apo AIV gene expression was 0.63 (0.39–1.01), p = 0.055). Viability correlated with % Bifidobacterium sp. (21.18 ± 7.66, p = 0.008) and valerate (−2.849 ± 1.048, p = 0.009) (β ± s.d.). This study suggests that microbiota interacts with intestine cell lipid metabolism. Since these metabolites can reach breast cells by systemic circulation, we hypothesized that they may influence cancer disease.
Collapse
Affiliation(s)
- Christine Bobin-Dubigeon
- EA 2160—IUML FR3473 CNRS, Université de Nantes, 44035 Nantes, France; (C.B.-D.); (T.-H.L.); (H.N.)
- Institut de Cancérologie de l’Ouest, 44805 Saint-Herblain, France
| | - Jean-Marie Bard
- EA 2160—IUML FR3473 CNRS, Université de Nantes, 44035 Nantes, France; (C.B.-D.); (T.-H.L.); (H.N.)
- Institut de Cancérologie de l’Ouest, 44805 Saint-Herblain, France
- Correspondence:
| | - Trang-Huyen Luu
- EA 2160—IUML FR3473 CNRS, Université de Nantes, 44035 Nantes, France; (C.B.-D.); (T.-H.L.); (H.N.)
| | | | - Hassan Nazih
- EA 2160—IUML FR3473 CNRS, Université de Nantes, 44035 Nantes, France; (C.B.-D.); (T.-H.L.); (H.N.)
| |
Collapse
|
27
|
Chhuon C, Zhang SY, Jung V, Lewandowski D, Lipecka J, Pawlak A, Sahali D, Ollero M, Guerrera IC. A sensitive S-Trap-based approach to the analysis of T cell lipid raft proteome. J Lipid Res 2020; 61:1512-1523. [PMID: 32769147 PMCID: PMC7604723 DOI: 10.1194/jlr.d120000672] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The analysis of T cell lipid raft proteome is challenging due to the highly dynamic nature of rafts and the hydrophobic character of raft-resident proteins. We explored an innovative strategy for bottom-up lipid raftomics based on suspension-trapping (S-Trap) sample preparation. Mouse T cells were prepared from splenocytes by negative immunoselection, and rafts were isolated by a detergent-free method and OptiPrep gradient ultracentrifugation. Microdomains enriched in flotillin-1, LAT, and cholesterol were subjected to proteomic analysis through an optimized protocol based on S-Trap and high pH fractionation, followed by nano-LC-MS/MS. Using this method, we identified 2,680 proteins in the raft-rich fraction and established a database of 894 T cell raft proteins. We then performed a differential analysis on the raft-rich fraction from nonstimulated versus anti-CD3/CD28 T cell receptor (TCR)-stimulated T cells. Our results revealed 42 proteins present in one condition and absent in the other. For the first time, we performed a proteomic analysis on rafts from ex vivo T cells obtained from individual mice, before and after TCR activation. This work demonstrates that the proposed method utilizing an S-Trap-based approach for sample preparation increases the specificity and sensitivity of lipid raftomics.
Collapse
Affiliation(s)
- Cerina Chhuon
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
| | - Shao-Yu Zhang
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
| | - Vincent Jung
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
| | - Daniel Lewandowski
- CEA/DRF/IBFJ/iRCM/LRTS, Fontenay-aux-Roses Cedex, France
- CEA/DRF/IBFJ/iRCM/LRTS, Fontenay-aux-Roses Cedex, France
- CEA/DRF/IBFJ/iRCM/LRTS, Fontenay-aux-Roses Cedex, France
- Université Paris-Sud, Paris, France
| | - Joanna Lipecka
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
| | - André Pawlak
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
| | - Dil Sahali
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
- AP-HP (Assistance Publique des Hôpitaux de Paris), Department of Nephrology and Renal Transplantation, Groupe Hospitalier Henri-Mondor, Créteil, France
- Université Paris Est Créteil, Créteil, France
| | - Mario Ollero
- Institut Mondor de Recherche Biomédicale, INSERM, U955, Créteil, France
- Université Paris Est Créteil, Créteil, France
| | - Ida Chiara Guerrera
- Proteomic Platform Necker, Structure Fédérative de Recherche SFR Necker US24, Paris, France
| |
Collapse
|
28
|
Wu J, Guo L, Qiu X, Ren Y, Li F, Cui W, Song S. Genkwadaphnin inhibits growth and invasion in hepatocellular carcinoma by blocking DHCR24-mediated cholesterol biosynthesis and lipid rafts formation. Br J Cancer 2020; 123:1673-1685. [PMID: 32958824 PMCID: PMC7686505 DOI: 10.1038/s41416-020-01085-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The liver is the central organ for cholesterol homoeostasis, and its dysfunction might cause liver pathological alterations including hepatocellular carcinomas (HCCs). 3β-hydroxysteroid-Δ24 reductase (DHCR24), a crucial enzyme of cholesterol biosynthetic pathway, is involved in lipid rafts formation. Genkwadaphnin (GD) is a daphnane diterpene isolated from the flower buds of Daphne genkwa Siebold et Zuccarini (Thymelaeaceae). METHODS We evaluated in vitro and in vivo effect of GD using HCC cells and BALB/c nude mice. Microarray assays were used to identify the differential genes by GD. DHCR24 expression and activity, cholesterol level, lipid rafts structure and the role of DHCR24 in human HCC specimens were tested by various molecular biology techniques. RESULTS High expression of DHCR24 in human HCC specimens was correlated with poor clinical outcome. Interfering DHCR24 altered growth and migration of HCC cells. GD inhibited growth and metastasis of HCC cells both in vivo and in vitro. GD suppressed DHCR24 expression and activity, as well as DHCR24-mediated cholesterol biosynthesis and lipid rafts formation, then further inhibited HCC cell invasion and migration. CONCLUSIONS Our data suggest that DHCR24-mediated cholesterol metabolism might be an effective therapeutic strategy in HCC, and natural product GD might be a promising agent for HCC therapy.
Collapse
Affiliation(s)
- Jie Wu
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Ling Guo
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Xiaoran Qiu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Yong Ren
- Department of Pathology, Central Theater Command General Hospital PLA, Wuhan, Hubei, 430070, People's Republic of China
| | - Feifei Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Wei Cui
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
| | - Shaojiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
29
|
Wang R, Zhu W, Peng J, Li K, Li C. Lipid rafts as potential mechanistic targets underlying the pleiotropic actions of polyphenols. Crit Rev Food Sci Nutr 2020; 62:311-324. [PMID: 32951435 DOI: 10.1080/10408398.2020.1815171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Polyphenols have attracted a lot of global attention due to their diverse biological actions against cancer, obesity, and cardiovascular diseases. Although extensive research has been carried out to elucidate the mechanisms of pleiotropic actions of polyphenols, this remains unclear. Lipid rafts are distinct nanodomains enriched in cholesterol and sphingolipids, present in the inner and outer leaflets of cell membranes, forming functional platforms for the regulation of cellular processes and diseases. Recent studies focusing on the interaction between polyphenols and cellular lipid rafts shed new light on the pleiotropic actions of polyphenols. Polyphenols are postulated to interact with lipid rafts in two ways: first, they interfere with the structural integrity of lipid rafts, by disrupting their structure and clustering of the ordered domains; second, they modulate the downstream signaling pathways mediated by lipid rafts, by binding to receptor proteins associated with lipid rafts, such as the 67 kDa laminin receptor (67LR), epidermal growth factor receptor (EGFR), and others. This study aims to elaborate the mechanism of interaction between polyphenols and lipid rafts, and describe pleiotropic preventive effects of polyphenols.
Collapse
Affiliation(s)
- Ruifeng Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wei Zhu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jinming Peng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Kaikai Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Chunmei Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Environment Correlative Food Science, Huazhong Agricultural University, Ministry of Education, Wuhan, China
| |
Collapse
|
30
|
Akella M, Malla R. Molecular modeling and in vitro study on pyrocatechol as potential pharmacophore of CD151 inhibitor. J Mol Graph Model 2020; 100:107681. [PMID: 32738620 DOI: 10.1016/j.jmgm.2020.107681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/25/2020] [Accepted: 06/24/2020] [Indexed: 11/24/2022]
Abstract
CD151 has been recognized as a prognostic marker, the therapeutic target of breast cancers, but less explored for small molecule inhibitors due to lack of a validated model. The 3-D structure of CD151 large extracellular loop (LEL) was modeled using the LOMETS server and validated by the Ramachandran plot. The validated structure was employed for molecular docking and structure-based pharmacophore analysis. Druglikeness was evaluated by the ADMET description protocol. Antiproliferative activity was evaluated by MTT, BrdU incorporation, flow cytometry, and cell death ELISAPLUS assay. This study predicted the best model for CD151-LEL with 94.1% residues in favored regions and Z score -2.79 kcal/mol using the threading method. The web-based receptor cavity method identified one functional target site, which was suitable for the binding of aromatic and heterocyclic compounds. Molecular docking study identified pyrocatechol (PCL) and 5-fluorouracil (FU) as potential leads of CD151-LEL. The pharmacophore model identified interaction points of modeled CD151-LEL with PCL and FU. Also, the analysis of ADMET properties revealed the drug-likeness of PCL and FU. The viability of MDA-MB 231 cells was significantly reduced with PCL and FU but less affected MCF-12A, normal healthy breast epithelial cell line. With 50% toxic concentration, both PCL and FU significantly inhibited 82.46 and 87.12% proliferation, respectively, of MDA-MB 231 cells by altering morphology and inducing G1 cell cycle arrest and apoptosis. In addition, PCL and FU inhibited the CD151 expression by 4.5-and 4.8-folds, respectively. This study suggests the further assessment of pyrocatechol as a potential lead of CD151 in breast cancer at the molecular level.
Collapse
Affiliation(s)
- Manasa Akella
- Cancer Biology Lab, Dept. of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to Be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - RamaRao Malla
- Cancer Biology Lab, Dept. of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to Be University), Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
31
|
DiPasquale M, Nguyen MHL, Rickeard BW, Cesca N, Tannous C, Castillo SR, Katsaras J, Kelley EG, Heberle FA, Marquardt D. The antioxidant vitamin E as a membrane raft modulator: Tocopherols do not abolish lipid domains. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183189. [PMID: 31954106 PMCID: PMC10443432 DOI: 10.1016/j.bbamem.2020.183189] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 01/06/2023]
Abstract
The antioxidant vitamin E is a commonly used vitamin supplement. Although the multi-billion dollar vitamin and nutritional supplement industry encourages the use of vitamin E, there is very little evidence supporting its actual health benefits. Moreover, vitamin E is now marketed as a lipid raft destabilizing anti-cancer agent, in addition to its antioxidant behaviour. Here, we studied the influence of vitamin E and some of its vitamers on membrane raft stability using phase separating unilamellar lipid vesicles in conjunction with small-angle scattering techniques and fluorescence microscopy. We find that lipid phase behaviour remains unperturbed well beyond physiological concentrations of vitamin E (up to a mole fraction of 0.10). Our results are consistent with a proposed line active role of vitamin E at the domain boundary. We discuss the implications of these findings as they pertain to lipid raft modification in native membranes, and propose a new hypothesis for the antioxidant mechanism of vitamin E.
Collapse
Affiliation(s)
- Mitchell DiPasquale
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada
| | - Michael H L Nguyen
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada
| | - Brett W Rickeard
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada
| | - Nicole Cesca
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada
| | - Christopher Tannous
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada
| | - Stuart R Castillo
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada
| | - John Katsaras
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA; Joint Institute for Neutron Sciences, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA; Department of Physics and Astronomy, University of Tennessee, Knoxville, TN 37996, USA
| | - Elizabeth G Kelley
- NIST Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | | | - Drew Marquardt
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario,Canada; Department of Physics, University of Windsor, Windsor, Ontario, Canada.
| |
Collapse
|
32
|
Wu Y, Zhao Y, He X, He Z, Wang T, Wan L, Chen L, Yan N. Hydroxypropyl‑β‑cyclodextrin attenuates the epithelial‑to‑mesenchymal transition via endoplasmic reticulum stress in MDA‑MB‑231 breast cancer cells. Mol Med Rep 2019; 21:249-257. [PMID: 31746388 PMCID: PMC6896369 DOI: 10.3892/mmr.2019.10802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/02/2019] [Indexed: 01/04/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) has been reported to serve vital roles in regulating the progress of cancer metastasis. In addition, lipid rafts enriched in sphingolipids and cholesterol serve important roles in physiological and biochemical processes as a signaling platform. The present study explored the effects of hydroxypropyl-β-cyclodextrin (HP-β-CD), a cholesterol-depleting agent of lipid rafts, on the transforming growth factor (TGF)-β/Smad signaling pathway and endoplasmic reticulum (ER) stress in mediating EMT in MDA-MB-231 breast cancer cells. HP-β-CD treatment inhibited TGF-β1-induced EMT, based on increased expression of E-cadherin and decreased expression of vimentin. HP-β-CD reduced the expression of the TGF receptor TβRI and blocked the phosphorylation of Smad2. In addition, HP-β-CD increased the expression of ER stress-related proteins (binding immunoglobulin protein and activating transcription factor 6), but TGF-β1 could reverse these changes. Sodium 4-phenylbutyrate, an inhibitor of ER stress, suppressed these effects of HP-β-CD on EMT and TGF-β/Smad signaling pathway inhibition in breast cancer cells. Thus, HP-β-CD can block the TGF-β/Smad signaling pathway via diminishing the expression of TβRI which helps to activate ER stress and attenuate EMT in MDA-MB-231 cells, highlighting a potential target of lipid rafts for breast cancer treatment.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yiyang Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xuanhong He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiqiang He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tian Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Linxi Wan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lai Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Nianlong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
33
|
Shailender G, Patanla K, Malla RR. ShRNA-mediated matrix metalloproteinase-2 gene silencing protects normal cells and sensitizes cancer cells against ionizing-radiation induced damage. J Cell Biochem 2019; 121:1332-1352. [PMID: 31489968 DOI: 10.1002/jcb.29369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/20/2019] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Ionizing radiation (IR) affects healthy tissues during the treatment of cancer radiation therapy and other nuclear and radiological accidents. Some natural compounds showed nonspecific radioprotective activity with severe side effects. The present study is aimed to develop potent and specific radioprotective short hairpin RNA (shRNA), which selectively protects normal cells from IR by specifically targeting matrix metalloproteinases (MMP-2). RESULTS IR reduced the viability of human normal dermal fibroblasts (HDFs) in a dose-response manner. It enhanced the expression of MMP-2 at 10 Gy. Plasmid MMP-2shRNA (pMMP-2) reduced the IR (10 Gy) induced cytotoxicity analyzed by lactate dehydrogenase (LDH) assay, normalized IR induced cellular and morphological changes with enhanced the clonogenicity in 48 hours at 2 µg/mL. It reduced the ROS generation, released HDFs from G2 /M arrest and rescued from apoptosis analyzed by DCFDA dye, cell cycle analysis by PI stain and annexin V assay, respectively. pMMP-2 also modulates the expression of EGFR and reduced IR induced expression of DNA damage response protein, ATM and increased the expression of repair proteins, KU70/KU80, and RAD51. In addition, decreased the expression of cell cycle regulatory proteins cyclin-dependent kinases (CDK1) and Cyclin B as well as proapoptotic proteins BAX, caspase-3, and Cytochrome-C and increased the expression of survival protein, Bcl-2. In contrary pMMP-2 decreased the LDH activity, survival fraction and blocked G2 /M phase of cell cycle and increased apoptosis in MCF-7 cells. In addition, decreased the expression of EGFR, proapoptotic BAX and DNA repair proteins ATM, KU70/80 and RAD51, increased expression of cyclinB as well as CDK1. CONCLUSION Results conclude that pMMP-2 protected HDFs from IR and sensitized the MCF-7 cells. Therefore, pMMP-2 can be employed for better treatment of radiation accidents and during the treatment of radiotherapy.
Collapse
Affiliation(s)
- Gugalavath Shailender
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Vishakhapatnam, India
| | - Kiranmayi Patanla
- Department of Biotechnology, GIS, GITAM (Deemed to be University), Vishakhapatnam, India
| | - Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM (Deemed to be University), Vishakhapatnam, India
| |
Collapse
|
34
|
Zhao Z, Dong Q, Liu X, Wei L, Liu L, Li Y, Wang X. Dynamic transcriptome profiling in DNA damage-induced cellular senescence and transient cell-cycle arrest. Genomics 2019; 112:1309-1317. [PMID: 31376528 DOI: 10.1016/j.ygeno.2019.07.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 04/14/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022]
Abstract
Cellular senescence is an irreversible cell cycle arrest process associated with aging and senescence-related diseases. DNA damage is an extensive feature of cellular senescence and aging. Different levels of DNA damage could lead to cellular senescence or transient cell-cycle arrest, but the genetic regulatory mechanisms determining cell fate are still not clear. In this work, high-resolution time course analysis of gene expression in DNA damage-induced cellular senescence and transient cell-cycle arrest was used to explore the transcriptomic differences between different cell fates after DNA damage response and to investigate the key regulatory factors affecting senescent cell fates. Pathways such as the cell cycle, DNA repair and cholesterol metabolism showed characteristic differential response. A number of key transcription factors were predicted to regulating cell cycle and DNA repair. Our study provides genome-wide insights into the molecular-level mechanisms of senescent cell fate decisions after DNA damage response.
Collapse
Affiliation(s)
- Zhen Zhao
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Qiongye Dong
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xuehui Liu
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Lei Wei
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Liyang Liu
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yanda Li
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xiaowo Wang
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and System Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
35
|
Qian XL, Pan YH, Huang QY, Shi YB, Huang QY, Hu ZZ, Xiong LX. Caveolin-1: a multifaceted driver of breast cancer progression and its application in clinical treatment. Onco Targets Ther 2019; 12:1539-1552. [PMID: 30881011 PMCID: PMC6398418 DOI: 10.2147/ott.s191317] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human breast cancer is one of the most frequent cancer diseases and causes of death among female population worldwide. It appears at a high incidence and has a high malignancy, mortality, recurrence rate and poor prognosis. Caveolin-1 (Cav1) is the main component of caveolae and participates in various biological events. More and more experimental studies have shown that Cav1 plays a critical role in the progression of breast cancer including cell proliferation, apoptosis, autophagy, invasion, migration and breast cancer metastasis. Besides, Cav1 has been found to be involved in chemotherapeutics and radiotherapy resistance, which are still the principal problems encountered in clinical breast cancer treatment. In addition, stromal Cav1 may be a potential indicator for breast cancer patients' prognosis. In the current review, we cover the state-of-the-art study, development and progress on Cav1 and breast cancer, altogether describing the role of Cav1 in breast cancer progression and application in clinical treatment, in the hope of providing a basis for further research and promoting CAV1 gene as a potential target to diagnose and treat aggressive breast cancers.
Collapse
Affiliation(s)
- Xian-Ling Qian
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yi-Hang Pan
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qi-Yuan Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yu-Bo Shi
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
| | - Qing-Yun Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
| | - Zhen-Zhen Hu
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China, ;
| | - Li-Xia Xiong
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China, ;
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China, ;
| |
Collapse
|
36
|
Mentoor I, Engelbrecht AM, Nell T. Fatty acids: Adiposity and breast cancer chemotherapy, a bad synergy? Prostaglandins Leukot Essent Fatty Acids 2019; 140:18-33. [PMID: 30553399 DOI: 10.1016/j.plefa.2018.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Globally, breast cancer continues to be a major concern in women's health. Lifestyle related risk factors, specifically excess adipose tissue (adiposity) has reached epidemic proportions and has been identified as a major risk factor in the development of breast cancer. Dysfunctional adipose tissue has evoked research focusing on its association with metabolic-related conditions, breast cancer risk and progression. Adipose dysfunction in coordination with immune cells and inflammation, are responsible for accelerated cell growth and survival of cancer cells. Recently, evidence also implicates adiposity as a potential risk factor for chemotherapy resistance. Chemotherapeutic agents have been shown to negatively impact adipose tissue. Since adipose tissue is a major storage site for fatty acids, it is not unlikely that these negative effects may disrupt adipose tissue homeostasis. It is therefore argued that fatty acid composition may be altered due to the chemotherapeutic pharmacokinetics, which in turn could have severe health related outcomes. The underlying molecular mechanisms elucidating the effects of fatty acid composition in adiposity-linked drug resistance are still unclear and under explored. This review focuses on the potential role of adiposity in breast cancer and specifically emphasizes the role of fatty acids in cancer progression and treatment resistance.
Collapse
Affiliation(s)
- Ilze Mentoor
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - A-M Engelbrecht
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - Theo Nell
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa.
| |
Collapse
|
37
|
Zakany F, Pap P, Papp F, Kovacs T, Nagy P, Peter M, Szente L, Panyi G, Varga Z. Determining the target of membrane sterols on voltage-gated potassium channels. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:312-325. [PMID: 30553843 DOI: 10.1016/j.bbalip.2018.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/30/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022]
Abstract
Cholesterol, an essential lipid component of cellular plasma membranes, regulates fluidity, mechanical integrity, raft structure and may specifically interact with membrane proteins. Numerous effects on ion channels by cholesterol, including changes in current amplitude, voltage dependence and gating kinetics, have been reported. We have previously described such changes in the voltage-gated potassium channel Kv1.3 of lymphocytes by cholesterol and its analog 7-dehydrocholesterol (7DHC). In voltage-gated channels membrane depolarization induces movement of the voltage sensor domains (VSD), which is transmitted by a coupling mechanism to the pore domain (PD) to open the channel. Here, we investigated whether cholesterol effects were mediated by the VSD to the pore or the PD was the direct target. Specificity was tested by comparing Kv1.3 and Kv10.1 channels having different VSD-PD coupling mechanisms. Current recordings were performed with two-electrode voltage-clamp fluorometry, where movement of the VSDs was monitored by attaching fluorophores to external cysteine residues introduced in the channel sequence. Loading the membrane with cholesterol or 7DHC using methyl-β-cyclodextrin induced changes in the steady-state and kinetic parameters of the ionic currents while leaving fluorescence parameters mostly unaffected in both channels. Non-stationary noise analysis revealed that reduction of single channel conductance rather than that of open probability caused the observed current decrease. Furthermore, confocal laser scanning and stimulated emission depletion microscopy demonstrated significant changes in the distribution of these ion channels in response to sterol loading. Our results indicate that sterol-induced effects on ion channel gating directly target the pore and do not act via the VSD.
Collapse
Affiliation(s)
- Florina Zakany
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary
| | - Pal Pap
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary; MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary
| | - Ferenc Papp
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary; MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary
| | - Tamas Kovacs
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary
| | - Peter Nagy
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary
| | - Maria Peter
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Temesvari Krt. 62, Szeged H-6726, Hungary
| | - Lajos Szente
- CycloLab Cyclodextrin R & D Laboratory Ltd., Illatos u. 7, Budapest H-1097, Hungary
| | - Gyorgy Panyi
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary; MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary
| | - Zoltan Varga
- Division of Biophysics, Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary; MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Egyetem ter 1, Debrecen H-4032, Hungary.
| |
Collapse
|
38
|
Morin EE, Li XA, Schwendeman A. HDL in Endocrine Carcinomas: Biomarker, Drug Carrier, and Potential Therapeutic. Front Endocrinol (Lausanne) 2018; 9:715. [PMID: 30555417 PMCID: PMC6283888 DOI: 10.3389/fendo.2018.00715] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022] Open
Abstract
High-density lipoprotein (HDL) have long been studied for their protective role against cardiovascular diseases, however recently relationship between HDL and cancer came into focus. Several epidemiological studies have shown an inverse correlation between HDL-cholesterol (HDL-C) and cancer risk, and some have even implied that HDL-C can be used as a predictive measure for survival prognosis in for specific sub-population of certain types of cancer. HDL itself is an endogenous nanoparticle capable of removing excess cholesterol from the periphery and returning it to the liver for excretion. One of the main receptors for HDL, scavenger receptor type B-I (SR-BI), is highly upregulated in endocrine cancers, notably due to the high demand for cholesterol by cancer cells. Thus, the potential to exploit administration of cholesterol-free reconstituted or synthetic HDL (sHDL) to deplete cholesterol in endocrine cancer cell and stunt their growth of use chemotherapeutic drug loaded sHDL to target payload delivery to cancer cell has become increasingly attractive. This review focuses on the role of HDL and HDL-C in cancer and application of sHDLs as endocrine cancer therapeutics.
Collapse
Affiliation(s)
- Emily E. Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Xiang-An Li
- Department of Physiology, Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
39
|
Gupta VK, Sharma NS, Kesh K, Dauer P, Nomura A, Giri B, Dudeja V, Banerjee S, Bhattacharya S, Saluja A, Banerjee S. Metastasis and chemoresistance in CD133 expressing pancreatic cancer cells are dependent on their lipid raft integrity. Cancer Lett 2018; 439:101-112. [PMID: 30290209 DOI: 10.1016/j.canlet.2018.09.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/25/2018] [Accepted: 09/21/2018] [Indexed: 02/01/2023]
Abstract
Metabolic rewiring is an integral part of tumor growth. Among metabolic pathways, the Mevalonic-Acid-Pathway (MVAP) plays a key role in maintaining membrane architecture through cholesterol synthesis, thereby affecting invasiveness. In the current study, we show for the first time that CD133Hi pancreatic tumor initiating cells (TIC) have increased expression of MVAP enzymes, cholesterol-content and Caveolin expression. Further, we show that CD133 in these cells is localized in the lipid-rafts (characterized by Cav-1-cholesterol association). Disruption of lipid-rafts by either depleting Cav-1 or by inhibiting MVAP by lovastatin decreased metastatic-potential and chemoresistance in CD133Hi cells while not affecting the CD133lo cells. Additionally, disruption of lipid-raft results in deregulation of FAK-signaling, decreasing invasiveness in pancreatic-TICs. Furthermore, this also inhibits ABC-transporter activity resulting in sensitizing TICs to standard chemotherapeutic agents. Repurposing existing drugs for new clinical applications is one of the safest and least resource intensive approaches to improve therapeutic options. In this context, our study is extremely timely as it shows that targeting lipid-rafts with statins can sensitize the normally resistant pancreatic TICHi-cells to standard chemotherapy and decrease metastasis, thereby defining a novel strategy for targeting the TICHi-PDAC.
Collapse
Affiliation(s)
| | - Nikita S Sharma
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Kousik Kesh
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Patricia Dauer
- Department of Surgery, University of Miami, Miami, FL, 33136, USA; Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Alice Nomura
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Bhuwan Giri
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Vikas Dudeja
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Santanu Banerjee
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | | | - Ashok Saluja
- Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Sulagna Banerjee
- Department of Surgery, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
40
|
Wittkowski KM, Dadurian C, Seybold MP, Kim HS, Hoshino A, Lyden D. Complex polymorphisms in endocytosis genes suggest alpha-cyclodextrin as a treatment for breast cancer. PLoS One 2018; 13:e0199012. [PMID: 29965997 PMCID: PMC6028090 DOI: 10.1371/journal.pone.0199012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Abstract
Most breast cancer deaths are caused by metastasis and treatment options beyond radiation and cytotoxic drugs, which have severe side effects, and hormonal treatments, which are or become ineffective for many patients, are urgently needed. This study reanalyzed existing data from three genome-wide association studies (GWAS) using a novel computational biostatistics approach (muGWAS), which had been validated in studies of 600-2000 subjects in epilepsy and autism. MuGWAS jointly analyzes several neighboring single nucleotide polymorphisms while incorporating knowledge about genetics of heritable diseases into the statistical method and about GWAS into the rules for determining adaptive genome-wide significance. Results from three independent GWAS of 1000-2000 subjects each, which were made available under the National Institute of Health's "Up For A Challenge" (U4C) project, not only confirmed cell-cycle control and receptor/AKT signaling, but, for the first time in breast cancer GWAS, also consistently identified many genes involved in endo-/exocytosis (EEC), most of which had already been observed in functional and expression studies of breast cancer. In particular, the findings include genes that translocate (ATP8A1, ATP8B1, ANO4, ABCA1) and metabolize (AGPAT3, AGPAT4, DGKQ, LPPR1) phospholipids entering the phosphatidylinositol cycle, which controls EEC. These novel findings suggest scavenging phospholipids as a novel intervention to control local spread of cancer, packaging of exosomes (which prepare distant microenvironment for organ-specific metastases), and endocytosis of β1 integrins (which are required for spread of metastatic phenotype and mesenchymal migration of tumor cells). Beta-cyclodextrins (βCD) have already been shown to be effective in in vitro and animal studies of breast cancer, but exhibits cholesterol-related ototoxicity. The smaller alpha-cyclodextrins (αCD) also scavenges phospholipids, but cannot fit cholesterol. An in-vitro study presented here confirms hydroxypropyl (HP)-αCD to be twice as effective as HPβCD against migration of human cells of both receptor negative and estrogen-receptor positive breast cancer. If the previous successful animal studies with βCDs are replicated with the safer and more effective αCDs, clinical trials of adjuvant treatment with αCDs are warranted. Ultimately, all breast cancer are expected to benefit from treatment with HPαCD, but women with triple-negative breast cancer (TNBC) will benefit most, because they have fewer treatment options and their cancer advances more aggressively.
Collapse
Affiliation(s)
- Knut M. Wittkowski
- Center for Clinical and Translational Science, The Rockefeller University, New York, New York, United States of America
| | - Christina Dadurian
- Center for Clinical and Translational Science, The Rockefeller University, New York, New York, United States of America
| | - Martin P. Seybold
- Institut für Formale Methoden der Informatik, Universität Stuttgart, Stuttgart, Germany
| | - Han Sang Kim
- Department of Pediatrics, and Cell and Developmental Biology Weill Medical College of Cornell University, New York, New York, United States of America
| | - Ayuko Hoshino
- Department of Pediatrics, and Cell and Developmental Biology Weill Medical College of Cornell University, New York, New York, United States of America
| | - David Lyden
- Department of Pediatrics, and Cell and Developmental Biology Weill Medical College of Cornell University, New York, New York, United States of America
| |
Collapse
|
41
|
Kumar M, Irungbam K, Kataria M. Depletion of membrane cholesterol compromised caspase-8 imparts in autophagy induction and inhibition of cell migration in cancer cells. Cancer Cell Int 2018; 18:23. [PMID: 29467593 PMCID: PMC5819249 DOI: 10.1186/s12935-018-0520-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/12/2018] [Indexed: 01/08/2023] Open
Abstract
Background Cholesterol in lipid raft plays crucial role on cancer cell survival during metastasis of cancer cells. Cancer cells are reported to enrich cholesterol in lipid raft which make them more susceptible to cell death after cholesterol depletion than normal cells. Methyl-β-cyclodextrin (MβCD), an amphipathic polysaccharide known to deplete the membrane cholesterol, induces cell death selectively in cancer cells. Present work was designed to identify the major form of programmed cell death in membrane cholesterol depleted cancer cells (MDA-MB 231 and 4T1) and its impact on migration efficiency of cancer cells. Methods Membrane cholesterol alteration and morphological changes in 4T1 and MDA-MB 231 cancer cells by MβCD were measured by fluorescent microscopy. Cell death and cell proliferation were observed by PI, AO/EB and MTT assay respectively. Programme cell death was confirmed by flow cytometer. Caspase activation was assessed by MTT and PI after treatments with Z-VAD [OME]-FMK, mitomycin c and cycloheximide. Necroptosis, autophagy, pyroptosis and paraptosis were examined by cell proliferation assay and flow cytometry. Relative quantitation of mRNA of caspase-8, necroptosis and autophagy genes were performed. Migration efficiency of cancer cells were determined by wound healing assay. Results We found caspase independent cell death in cholesterol depleted MDA-MB 231 cells which was reduced by (3-MA) an autophagy inhibitor. Membrane cholesterol depletion neither induces necroptosis, paraptosis nor pyroptosis in MDA-MB 231 cells. Subsequent activation of caspase-8 after co-incubation of mitomycin c and cycloheximide separately, restored the cell viability in cholesterol depleted MDA-MB 231 cells. Down regulation of caspase-8 mRNA in cholesterol depleted cancer cells ensures that caspase-8 indirectly promotes the induction of autophagy. In another experiment we have demonstrated that membrane cholesterol depletion reduces the migration efficiency in cancer cells. Conclusion Together our experimental data suggests that membrane cholesterol is the crucial for the recruitment and activation of caspase-8 as well as its non-apoptotic functions in cancer cells. Enriched cholesterol in lipid raft of cancer cells may be regulating the cross talk between caspase-8 and autophagy machineries to promote their survival and migration. Therefore it can be explored to understand and address the issues of chemotherapeutic and drugs resistance.
Collapse
Affiliation(s)
- Mukesh Kumar
- Indian Veterinary Research Institute, Bareilly, India
| | | | - Meena Kataria
- Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
42
|
Chatterjee AD, Roy D, Guevara P, Pal R, Naryan M, Roychowdhury S, Das S. Arachidonic Acid Induces the Migration of MDA-MB-231 Cells by Activating Raft-associated Leukotriene B4 Receptors. CLINICAL CANCER DRUGS 2018; 5:28-41. [PMID: 30443489 PMCID: PMC6233886 DOI: 10.2174/2212697x05666180418145601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The migration of tumor cells is critical in spreading cancers through the lymphatic nodes and circulatory systems. Although arachidonic acid (AA) and its soluble metabolites have been shown to induce the migration of breast and colon cancer cells, the mechanism by which it induces such migration has not been fully understood. OBJECTIVE The effect of AA on migratory responses of the MDA-MB-231 cell line (a triple-negative breast cancer cell) was examined and compared with MCF-7 (estrogen-receptor positive) breast cancer cells to elucidate the mechanism of AA-induced migration. METHODS Migrations of breast cancer cells were examined with the help of wound-healing assays. AA-induced eicosanoid synthesis was monitored by RP-HPLC. Cellular localizations of lipoxygenase and lipid rafts were assessed by immunoblot and confocal microscopy. RESULTS AA treatment stimulated the synthesis of leukotriene B4 (LTB4) and HETE-8, but lowered the levels of prostaglandin E2 (PGE2), prostaglandin D2 (PGD2), and HETE-5 in MDA-MB-231 cells. Further analysis indicated that AA increased the expression of 5-lipoxygenase (5-LOX) in this cell line and inhibiting its expression by small molecule inhibitors lowered the production of LTB4 and reduced migration. In contrast, MCF-7 cells did not show any appreciable changes in eicosanoid synthesis, 5-LOX expression, or cellular migration. CONCLUSION Our results suggest that AA treatment activates the BLT1 receptor (present in membrane microdomains) and stimulates the synthesis of LTB4 production, which is likely to be associated with the migration of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Atasi De Chatterjee
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Debarshi Roy
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Priscilla Guevara
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Rituraj Pal
- Department of Chemistry, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Mahesh Naryan
- Department of Chemistry, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Sukla Roychowdhury
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| | - Siddhartha Das
- Department of Biological Sciences, Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
- The Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968-0519, USA
| |
Collapse
|
43
|
Rac1-stimulated macropinocytosis enhances Gβγ activation of PI3Kβ. Biochem J 2017; 474:3903-3914. [PMID: 29046393 DOI: 10.1042/bcj20170279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/05/2017] [Accepted: 10/10/2017] [Indexed: 12/30/2022]
Abstract
Phosphoinositide 3-kinases (PI 3-kinases) are regulated by a diverse range of upstream activators, including receptor tyrosine kinases (RTKs), G-protein-coupled receptors (GPCRs), and small GTPases from the Ras, Rho and Rab families. For the Class IA PI 3-kinase PI3Kβ, two mechanisms for GPCR-mediated regulation have been described: direct binding of Gβγ subunits to the C2-helical domain linker of p110β, and Dock180/Elmo1-mediated activation of Rac1, which binds to the Ras-Binding Domain of p110β. We now show that the integration of these dual pathways is unexpectedly complex. In breast cancer cells, expression of constitutively activated Rac1 (CA-Rac1) along with either GPCR stimulation or expression of Gβγ led to an additive PI3Kβ-dependent activation of Akt. Whereas CA-Rac1-mediated activation of Akt was blocked in cells expressing a mutated PI3Kβ that cannot bind Gβγ, Gβγ and GPCR-mediated activation of Akt was preserved when Rac1 binding to PI3Kβ was blocked. Surprisingly, PI3Kβ-dependent CA-Rac1 signaling to Akt was still seen in cells expressing a mutant p110β that cannot bind Rac1. Instead of directly binding to PI3Kβ, CA-Rac1 acts by enhancing Gβγ coupling to PI3Kβ, as CA-Rac1-mediated Akt activation was blocked by inhibitors of Gβγ. Cells expressing CA-Rac1 exhibited a robust induction of macropinocytosis, and inhibitors of macropinocytosis blocked the activation of Akt by CA-Rac1 or lysophosphatidic acid. Our data suggest that Rac1 can potentiate the activation of PI3Kβ by GPCRs through an indirect mechanism, by driving the formation of macropinosomes that serve as signaling platforms for Gβγ coupling to PI3Kβ.
Collapse
|
44
|
Elevated tumor LDLR expression accelerates LDL cholesterol-mediated breast cancer growth in mouse models of hyperlipidemia. Oncogene 2017; 36:6462-6471. [PMID: 28759039 PMCID: PMC5690879 DOI: 10.1038/onc.2017.247] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Obesity is associated with an increase in cancer-specific mortality in women with breast cancer. Elevated cholesterol, particularly low-density lipoprotein cholesterol (LDL-C) is frequently seen in obese women. Here, we aimed to determine the importance of elevated circulating LDL, and LDL receptor (LDLR) expression in tumor cells, on the growth of breast cancer using mouse models of hyperlipidemia. We describe two novel immunodeficient mouse models of hyperlipidemia (Rag1−/−/LDLR−/− and Rag1−/−/ApoE (apolipoprotein E)−/− mice), in addition to established immunocompetent LDLR−/− and ApoE−/− mice. The mice were used to study the effects of elevated LDL-C in human triple negative (MDA-MB-231) and mouse Her2/Neu overexpressing (MCNeuA) breast cancers. Tumors derived from MCNeuA and MDA-MB-231 cells had high LDLR expression and formed larger tumors in mice with high circulating LDL-C concentrations than in mice with lower LDL-C. Silencing the LDLR in the tumor cells led to decreased growth of Her2Neu overexpressing tumors in LDLR−/− and ApoE−/− mice, with increased Caspase 3 cleavage. Additionally, in vitro, silencing the LDLR led to decreased cell survival in serum-starved conditions, associated with Caspase 3 cleavage. Examining publically available human datasets, we found that high LDLR expression in human breast cancers was associated with decreased recurrence-free survival, particularly in patients treated with systemic therapies. Overall, our results highlight the importance of the LDLR in the growth of triple negative and HER2 overexpressing breast cancers in the setting of elevated circulating LDL-C, which may be important contributing factors to the increased recurrence and mortality in obese women with breast cancer.
Collapse
|