1
|
Wang Z, Pang S, Liu X, Dong Z, Tian Y, Ashrafizadeh M, Rabiee N, Ertas YN, Mao Y. Chitosan- and hyaluronic acid-based nanoarchitectures in phototherapy: Combination cancer chemotherapy, immunotherapy and gene therapy. Int J Biol Macromol 2024; 273:132579. [PMID: 38795895 DOI: 10.1016/j.ijbiomac.2024.132579] [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: 02/01/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Cancer phototherapy has been introduced as a new potential modality for tumor suppression. However, the efficacy of phototherapy has been limited due to a lack of targeted delivery of photosensitizers. Therefore, the application of biocompatible and multifunctional nanoparticles in phototherapy is appreciated. Chitosan (CS) as a cationic polymer and hyaluronic acid (HA) as a CD44-targeting agent are two widely utilized polymers in nanoparticle synthesis and functionalization. The current review focuses on the application of HA and CS nanostructures in cancer phototherapy. These nanocarriers can be used in phototherapy to induce hyperthermia and singlet oxygen generation for tumor ablation. CS and HA can be used for the synthesis of nanostructures, or they can functionalize other kinds of nanostructures used for phototherapy, such as gold nanorods. The HA and CS nanostructures can combine chemotherapy or immunotherapy with phototherapy to augment tumor suppression. Moreover, the CS nanostructures can be functionalized with HA for specific cancer phototherapy. The CS and HA nanostructures promote the cellular uptake of genes and photosensitizers to facilitate gene therapy and phototherapy. Such nanostructures specifically stimulate phototherapy at the tumor site, with particle toxic impacts on normal cells. Moreover, CS and HA nanostructures demonstrate high biocompatibility for further clinical applications.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng 252000, Shandong, PR China
| | - Shuo Pang
- Department of Urinary Surgery, Jinan Third People's Hospital, Jinan, Shandong 250101, PR China
| | - Xiaoli Liu
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zi Dong
- Department of Gastroenterology, Lincang People's Hospital, Lincang, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, United States
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077 India
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Türkiye; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Türkiye; UNAM-National Nanotechnology Research Center, Bilkent University, Ankara 06800, Türkiye.
| | - Ying Mao
- Department of Oncology, Suining Central Hospital, Suining City, Sichuan, China.
| |
Collapse
|
2
|
OUP accepted manuscript. Glycobiology 2022; 32:743-750. [DOI: 10.1093/glycob/cwac028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 01/10/2023] Open
|
3
|
Díaz M, Pibuel M, Paglilla N, Poodts D, Álvarez E, Papademetrio DL, Hajos SE, Lompardía SL. 4-Methylumbelliferone induces antitumor effects independently of hyaluronan synthesis inhibition in human acute leukemia cell lines. Life Sci 2021; 287:120065. [PMID: 34678263 DOI: 10.1016/j.lfs.2021.120065] [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: 07/20/2021] [Revised: 09/28/2021] [Accepted: 10/15/2021] [Indexed: 11/24/2022]
Abstract
AIMS Despite continuous improvement in the treatment of acute leukemia, new therapies are still needed to overcome resistance and reduce adverse effects. The aim of this work was to study the tumor-suppressive effects of 4-methylumbelliferone (4MU) in human acute leukemia cell lines. In addition, we aimed to address the extent of these effects in relation to the inhibition of hyaluronic acid (HA) synthesis. MAIN METHODS HA levels were measured by an ELISA-like assay. Human acute leukemia cell lines were treated with 4MU, HA or their combination. Cell proliferation was assessed by the [3H]-Tdr uptake assay, metabolic activity by the XTT assay and cell death was determined by DAPI, AO/EB and AnnexinV-PE/7-AAD staining. Senescence induction was evaluated by SA-β-Gal and C12FDG staining. Total and surface RHAMM expression levels were assessed by flow cytometry and fluorescence microscopy. KEY FINDINGS 4MU reduced metabolic activity and inhibited cell proliferation in all leukemia cells, and these effects were explained by the induction of senescence or cell death depending on the cell line evaluated. Exogenous HA failed to prevent most of the tumor-suppressive effects observed. Results from this work suggest that the tumor-suppressive effects exerted by 4MU would be explained by HA-synthesis-independent mechanisms. SIGNIFICANCE These findings broaden the knowledge of 4MU as a potential treatment in acute leukemia. We report for the first time the existence of tumor-suppressive effects of 4MU on human acute leukemia cell lines that are independent of its role as HA-synthesis inhibitor.
Collapse
Affiliation(s)
- Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Matías Pibuel
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Nadia Paglilla
- Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daniela Poodts
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Elida Álvarez
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daniela L Papademetrio
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvia E Hajos
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Silvina L Lompardía
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Microbiología, Inmunología y Biotecnología, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
4
|
Garantziotis S. Modulation of hyaluronan signaling as a therapeutic target in human disease. Pharmacol Ther 2021; 232:107993. [PMID: 34587477 DOI: 10.1016/j.pharmthera.2021.107993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
The extracellular matrix is an active participant, modulator and mediator of the cell, tissue, organ and organismal response to injury. Recent research has highlighted the role of hyaluronan, an abundant glycosaminoglycan constituent of the extracellular matrix, in many fundamental biological processes underpinning homeostasis and disease development. From this basis, emerging studies have demonstrated the therapeutic potential of strategies which target hyaluronan synthesis, biology and signaling, with significant promise as therapeutics for a variety of inflammatory and immune diseases. This review summarizes the state of the art in this field and discusses challenges and opportunities in what could emerge as a new class of therapeutic agents, that we term "matrix biologics".
Collapse
Affiliation(s)
- Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
5
|
Jadhao M, Tsai EM, Yang HC, Chen YF, Liang SS, Wang TN, Teng YN, Huang HW, Wang LF, Chiu CC. The Long-Term DEHP Exposure Confers Multidrug Resistance of Triple-Negative Breast Cancer Cells through ABC Transporters and Intracellular ROS. Antioxidants (Basel) 2021; 10:949. [PMID: 34208283 PMCID: PMC8230873 DOI: 10.3390/antiox10060949] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022] Open
Abstract
The characteristics of phthalates had been thought to be similar to endocrine disruptors, which increases cancer risk. The role of phthalates in acquired drug resistance remains unclear. In this study, we investigated the effect of di-(2-ethylhexyl) phthalate (DEHP) on acquired drug resistance in breast cancer. MCF7 and MDA-MB-231 breast cancer cells were exposed to long-term physiological concentration of DEHP for more than three months. Long-exposure DEHP permanently attenuated the anti-proliferative effect of doxorubicin with estrogen receptor-independent activity even after withdrawal of DEHP. Long term DEHP exposure significantly reduced ROS (O2-) level in MDA-MB-231 cells while increased in MCF7 cells. ATP-binding cassette (ABC) transporters possess a widely recognized mechanism of drug resistance and are considered a target for drug therapy. Upregulation of ABC family proteins, ABCB-1 and ABCC-1 observed in DEHP-exposed clones compared to doxorubicin-resistant (DoxR) and parental MDA-MB-231 cells. A viability assay showed enhanced multidrug resistance in DEHP-exposed clones against Dox, topotecan, and irinotecan. Inhibition of ABC transporters with tariquidar, enhanced drug cytotoxicity through increased drug accumulation reversing acquired multidrug resistance in MDA-MB-231 breast cancer cells. Tariquidar enhanced Dox cytotoxicity by increasing intracellular ROS production leading to caspase-3 mediated apoptosis. Activation of PI3K/Akt signaling enhanced proliferation and growth of DEHP-exposed MDA-MB-231 cells. Overall, long-term DEHP exposure resulted in acquired multidrug resistance by upregulating ABCB-1 and ABCC1; apart from proliferation PI3K/Akt may be responsible for acquired drug resistance through ABC transporter upregulation. Targeting ABCB1 and ABCC1 with tariquidar may be a promising strategy for reversing the acquired multidrug resistance of triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Mahendra Jadhao
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; or
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ho-Chun Yang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Shin Liang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Hurng-Wern Huang
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Li-Fang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; or
| | - Chien-Chih Chiu
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
6
|
Tavianatou AG, Piperigkou Z, Koutsakis C, Barbera C, Beninatto R, Franchi M, Karamanos NK. The action of hyaluronan in functional properties, morphology and expression of matrix effectors in mammary cancer cells depends on its molecular size. FEBS J 2021; 288:4291-4310. [PMID: 33512780 DOI: 10.1111/febs.15734] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/24/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
Breast cancer constitutes a heterogeneous disease. The expression profiles of estrogen receptors (ERs), as well as the expression patterns of extracellular matrix (ECM) macromolecules, determine its development and progression. Hyaluronan (HA) is an ECM molecule that regulates breast cancer cells' properties in a molecular size-dependent way. Previous studies have shown that 200-kDa HA fragments modulate the functional properties, morphology, and expression of several matrix mediators of the highly metastatic ERα- /ERβ+ MDA-MB-231 cells. In order to evaluate the effects of HA fragments (< 10, 30 and 200-kDa) in ERβ-suppressed breast cancer cells, the shERβ MDA-MB-231 cells were used. These cells are less aggressive when compared with MDA-MB-231 cells. To this end, the functional properties, the morphology, and the expression of the molecules associated with breast cancer cells metastatic potential were studied. Notably, both cell proliferation and invasion were significantly reduced after treatment with 200-kDa HA. Moreover, as assessed by scanning electron microscopy, 200-kDa HA affected cellular morphology, and as assessed by qPCR, upregulated the epithelial marker Ε-cadherin. The expression profiles of ECM mediators, such as HAS2, CD44, and MMP7, were also altered. On the other hand, cellular migration and the expression levels of syndecan-4 (SDC-4) were not significantly affected in contrast to our observations regarding MDA-MB-231 cells. These novel data demonstrate that the molecular size of the HA determines its effects on ERβ-suppressed breast cancer cells and that 200-kDa HA exhibits antiproliferative effects on these cells. A deeper understanding of this mechanism may contribute to the development of therapeutic strategies against breast cancer.
Collapse
Affiliation(s)
- Anastasia-Gerasimoula Tavianatou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | | | | | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Italy
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
7
|
Abstract
Multidrug resistance (MDR) is a vital issue in cancer treatment. Drug resistance can be developed through a variety of mechanisms, including increased drug efflux, activation of detoxifying systems and DNA repair mechanisms, and escape of drug-induced apoptosis. Identifying the exact mechanism related in a particular case is a difficult task. Proteomics is the large-scale study of proteins, particularly their expression, structures and functions. In recent years, comparative proteomic methods have been performed to analyze MDR mechanisms in drug-selected model cancer cell lines. In this paper, we review the recent developments and progresses by comparative proteomic approaches to identify potential MDR mechanisms in drug-selected model cancer cell lines, which may help understand and design chemical sensitizers.
Collapse
|
8
|
Ooki T, Hatakeyama M. Hyaluronan Degradation Promotes Cancer via Hippo-YAP Signaling: An Intervention Point for Cancer Therapy. Bioessays 2020; 42:e2000005. [PMID: 32449813 DOI: 10.1002/bies.202000005] [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: 01/15/2020] [Revised: 03/16/2020] [Indexed: 12/14/2022]
Abstract
High-molecular-weight hyaluronan acts as a ligand of the tumor-suppressive Hippo signal, whereas degradation of hyaluronan from a high-molecular-weight form to a low-molecular-weight forms by hyaluronidase 2 inhibits Hippo signal activation and thereby activates the pro-oncogenic transcriptional coactivator yes-associated protein (YAP), which creates a cancer-predisposing microenvironment and drives neoplastic transformation of cells through both cell-autonomous and non-cell-autonomous mechanisms. In fact, accumulation of low-molecular-weight hyaluronan in tissue stroma is observed in many types of cancers. Since inhibition of YAP activity suppresses tumor growth in vivo, pharmacological intervention of the Hippo-YAP signal is an attractive approach for future drug development. In this review, pharmacological intervention of excessive hyaluronan degradation as a novel approach for inhibition of the Hippo-YAP signal is also discussed. Development of hyaluronidase inhibitors may provide novel therapeutic strategies for human malignant tumors.
Collapse
Affiliation(s)
- Takuya Ooki
- Division of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| | - Masanori Hatakeyama
- Division of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
9
|
Pęcak A, Skalniak Ł, Pels K, Książek M, Madej M, Krzemień D, Malicki S, Władyka B, Dubin A, Holak TA, Dubin G. Anti-CD44 DNA Aptamers Selectively Target Cancer Cells. Nucleic Acid Ther 2020; 30:289-298. [PMID: 32379519 DOI: 10.1089/nat.2019.0833] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD44 is a type I transmembrane glycoprotein interacting with a number of extracellular components, including hyaluronic acid (HA). CD44-HA axis is involved in a variety of processes, including adhesion, migration, differentiation, trafficking, and others. CD44 is overexpressed in several cancers where binding of HA induces signal transduction leading to activation of antiapoptotic proteins and factors linked to drug resistance. As such, CD44 has been implicated in cancer growth, progression, and metastasis. It has been convincingly demonstrated that blocking CD44-HA interaction decreases cancer cell survival and metastasis. In this study, using in vitro selection, we have developed DNA aptamers recognizing a HA-binding domain of CD44 with high affinity and specificity. The aptamers bind to CD44 with nanomolar affinities and efficiently inhibit the growth of leukemic cancer cells characterized by high expression of CD44. The selectivity is demonstrated by an irrelevant effect on cells characterized by low CD44 levels. The obtained aptamers broaden the existing landscape of potential approaches to the development of antitumor strategies based on inhibition of the CD44 axis.
Collapse
Affiliation(s)
- Aleksandra Pęcak
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Łukasz Skalniak
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Katarzyna Pels
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mirosław Książek
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mariusz Madej
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Dobrosława Krzemień
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.,Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Stanisław Malicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Benedykt Władyka
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Adam Dubin
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Tad A Holak
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
10
|
Roedig H, Damiescu R, Zeng-Brouwers J, Kutija I, Trebicka J, Wygrecka M, Schaefer L. Danger matrix molecules orchestrate CD14/CD44 signaling in cancer development. Semin Cancer Biol 2020; 62:31-47. [PMID: 31412297 DOI: 10.1016/j.semcancer.2019.07.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023]
Abstract
The tumor matrix together with inflammation and autophagy are crucial regulators of cancer development. Embedded in the tumor stroma are numerous proteoglycans which, in their soluble form, act as danger-associated molecular patterns (DAMPs). By interacting with innate immune receptors, the Toll-like receptors (TLRs), DAMPs autonomously trigger aseptic inflammation and can regulate autophagy. Biglycan, a known danger proteoglycan, can regulate the cross-talk between inflammation and autophagy by evoking a switch between pro-inflammatory CD14 and pro-autophagic CD44 co-receptors for TLRs. Thus, these novel mechanistic insights provide some explanation for the plethora of reports indicating that the same matrix-derived DAMP acts either as a promoter or suppressor of tumor growth. In this review we will summarize and critically discuss the role of the matrix-derived DAMPs biglycan, hyaluronan, and versican in regulating the TLR-, CD14- and CD44-signaling dialogue between inflammation and autophagy with particular emphasis on cancer development.
Collapse
Affiliation(s)
- Heiko Roedig
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Roxana Damiescu
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Iva Kutija
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Liliana Schaefer
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
11
|
Abstract
The extracellular matrix is part of the microenvironment and its functions are associated with the physical and chemical properties of the tissue. Among the extracellular components, the glycosaminoglycan hyaluronan is a key component, defining both the physical and biochemical characteristics of the healthy matrices. The hyaluronan metabolism is strictly regulated in physiological conditions, but in the tumoral tissues, its expression, size and binding proteins interaction are dysregulated. Hyaluronan from the tumor microenvironment promotes tumor cell proliferation, invasion, immune evasion, stemness alterations as well as drug resistance. This chapter describes data regarding novel concepts of hyaluronan functions in the tumor. Additionally, we discuss potential clinical applications of targeting HA metabolism in cancer therapy.
Collapse
|
12
|
Landras A, Reger de Moura C, Jouenne F, Lebbe C, Menashi S, Mourah S. CD147 Is a Promising Target of Tumor Progression and a Prognostic Biomarker. Cancers (Basel) 2019; 11:cancers11111803. [PMID: 31744072 PMCID: PMC6896083 DOI: 10.3390/cancers11111803] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022] Open
Abstract
Microenvironment plays a crucial role in tumor development and progression. Cancer cells modulate the tumor microenvironment, which also contribute to resistance to therapy. Identifying biomarkers involved in tumorigenesis and cancer progression represents a great challenge for cancer diagnosis and therapeutic strategy development. CD147 is a glycoprotein involved in the regulation of the tumor microenvironment and cancer progression by several mechanisms—in particular, by the control of glycolysis and also by its well-known ability to induce proteinases leading to matrix degradation, tumor cell invasion, metastasis and angiogenesis. Accumulating evidence has demonstrated the role of CD147 expression in tumor progression and prognosis, suggesting it as a relevant tumor biomarker for cancer diagnosis and prognosis, as well as validating its potential as a promising therapeutic target in cancers.
Collapse
Affiliation(s)
- Alexandra Landras
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
| | - Coralie Reger de Moura
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Fanelie Jouenne
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Celeste Lebbe
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Dermatology Department and Centre d’Investigation Clinique (CIC), Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Suzanne Menashi
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Samia Mourah
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
- Correspondence: ; Tel.: +33-1-42-49-48-85
| |
Collapse
|
13
|
Lokman NA, Price ZK, Hawkins EK, Macpherson AM, Oehler MK, Ricciardelli C. 4-Methylumbelliferone Inhibits Cancer Stem Cell Activation and Overcomes Chemoresistance in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081187. [PMID: 31443261 PMCID: PMC6721459 DOI: 10.3390/cancers11081187] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/07/2019] [Accepted: 08/11/2019] [Indexed: 01/01/2023] Open
Abstract
We have recently shown that the extracellular matrix molecule hyaluronan (HA) plays a role in the development of ovarian cancer chemoresistance. This present study determined if HA production is increased in chemotherapy-resistant ovarian cancers and if the HA inhibitor 4-methylubelliferone (4-MU) can overcome chemoresistance to the chemotherapeutic drug carboplatin (CBP) and inhibit spheroid formation and the expression of cancer stem cell (CSC) markers. We additionally assessed whether 4-MU could inhibit in vivo invasion of chemoresistant primary ovarian cancer cells in the chicken embryo chorioallantoic membrane (CAM) assay. The expression of the HA synthases HAS2 and HAS3 was significantly increased in chemoresistant compared to chemosensitive primary ovarian cancer cells isolated from patient ascites. 4-MU significantly inhibited HA production, cell survival, and spheroid formation of chemoresistant serous ovarian cancer cells. In combination with CBP, 4-MU treatment significantly decreased ovarian cancer cell survival and increased apoptosis of chemoresistant primary cells compared to CBP alone. 4-MU significantly reduced spheroid formation, expression of CSC markers ALDH1A1 and ABCG2 in primary cell spheroid cultures, and ALDH1 immunostaining in patient-derived tissue explant assays following treatment with CBP. Furthermore, 4-MU was very effective at inhibiting in vivo invasion of chemoresistant primary cells in CAM assays. Inhibition of HA is therefore a promising new strategy to overcome chemoresistance and to improve ovarian cancer survival.
Collapse
Affiliation(s)
- Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Zoe K Price
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Emily K Hawkins
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Anne M Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA 5005, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
14
|
Tavianatou AG, Piperigkou Z, Barbera C, Beninatto R, Masola V, Caon I, Onisto M, Franchi M, Galesso D, Karamanos NK. Molecular size-dependent specificity of hyaluronan on functional properties, morphology and matrix composition of mammary cancer cells. Matrix Biol Plus 2019; 3:100008. [PMID: 33543007 PMCID: PMC7852304 DOI: 10.1016/j.mbplus.2019.100008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022] Open
Abstract
High levels of hyaluronan (ΗΑ), a major extracellular matrix (ECM) glycosaminoglycan, have been correlated with poor clinical outcome in several malignancies, including breast cancer. The high and low molecular weight HΑ forms exert diverse biological functions. Depending on their molecular size, ΗΑ forms either promote or attenuate signaling cascades that regulate cancer progression. In order to evaluate the effects of different ΗΑ forms on breast cancer cells' behavior, ΗΑ fragments of defined molecular size were synthesized. Breast cancer cells of different estrogen receptor (ER) status - the low metastatic, ERα-positive MCF-7 epithelial cells and the highly aggressive, ERβ-positive MDA-MB-231 mesenchymal cells - were evaluated following treatment with HA fragments. Scanning electron microscopy revealed that HA fragments critically affect the morphology of breast cancer cells in a molecular-size dependent mode. Moreover, the ΗΑ fragments affect cell functional properties, the expression of major ECM mediators and epithelial-to-mesenchymal transition (ΕΜΤ) markers. Notably, treatment with 200 kDa ΗΑ increased the expression levels of the epithelial marker Ε-cadherin and reduced the expression levels of HA synthase 2 and mesenchymal markers, like fibronectin and snail2/slug. These novel data suggest that the effects of HA in breast cancer cells depend on the molecular size and the ER status. An in-depth understanding on the mechanistic basis of these effects may contribute on the development of novel therapeutic strategies for the pharmacological targeting of aggressive breast cancer.
Collapse
Key Words
- BTH, bovine testes hyaluronidase
- Breast cancer
- CD44
- ECM, extracellular matrix
- EMT, epithelial-to-mesenchymal transition
- ER, estrogen receptor
- Epithelial-to-mesenchymal transition
- Estrogen receptors
- HA, hyaluronan or hyaluronic acid
- HAS, hyaluronan synthase
- HMW HA, high molecular weight hyaluronan
- HYAL, hyaluronidase
- Hyaluronan
- LMW HA, low molecular weight hyaluronan
- MET, mesenchymal-to-epithelial transition
- MMPs, matrix metalloproteinases
- SDC, syndecan
- SEM, scanning electron microscopy
- Scanning electron microscopy
- TIMPs, tissue inhibitors of metalloproteinases
- o-HA, hyaluronan oligomers
- s-HA, sulfated hyaluronan
- tPA, tissue plasminogen activator
- uPA, urokinase plasminogen activator
Collapse
Affiliation(s)
- Anastasia-Gerasimoula Tavianatou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece
| | - Carlo Barbera
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme, (PD), Italy
| | - Riccardo Beninatto
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme, (PD), Italy
| | - Valentina Masola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Italy
| | - Devis Galesso
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme, (PD), Italy
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece
| |
Collapse
|
15
|
Tavianatou AG, Caon I, Franchi M, Piperigkou Z, Galesso D, Karamanos NK. Hyaluronan: molecular size-dependent signaling and biological functions in inflammation and cancer. FEBS J 2019; 286:2883-2908. [PMID: 30724463 DOI: 10.1111/febs.14777] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 02/04/2019] [Indexed: 12/15/2022]
Abstract
Hyaluronan (HA) is a linear nonsulfated glycosaminoglycan of the extracellular matrix that plays a pivotal role in a variety of biological processes. High-molecular weight HA exhibits different biological properties than oligomers and low-molecular weight HA. Depending on their molecular size, HA fragments can influence cellular behavior in a different mode of action. This phenomenon is attributed to the different manner of interaction with the HA receptors, especially CD44 and RHAMM. Both receptors can trigger signaling cascades that regulate cell functional properties, such as proliferation migration, angiogenesis, and wound healing. HA fragments are able to enhance or attenuate the HA receptor-mediated signaling pathways, as they compete with the endogenous HA for binding to the receptors. The modulation of these pathways could be crucial for the development of pathological conditions, such as inflammation and cancer. The primary goal of this review is to critically present the importance of HA molecular size on cellular signaling, functional cell properties, and morphology in normal and pathological conditions, including inflammation and cancer. A deeper understanding of these mechanisms could contribute to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anastasia G Tavianatou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Italy
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH) /Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | | | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH) /Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
16
|
Price ZK, Lokman NA, Ricciardelli C. Differing Roles of Hyaluronan Molecular Weight on Cancer Cell Behavior and Chemotherapy Resistance. Cancers (Basel) 2018; 10:E482. [PMID: 30513961 PMCID: PMC6316154 DOI: 10.3390/cancers10120482] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Hyaluronan (HA), a glycosaminoglycan located in the extracellular matrix, is important in embryo development, inflammation, wound healing and cancer. There is an extensive body of research demonstrating the role of HA in all stages of cancer, from initiation to relapse and therapy resistance. HA interacts with multiple cell surface receptors, including CD44, receptor for hyaluronan mediated motility (RHAMM) and intracellular signaling pathways, including receptor tyrosine kinase pathways, to promote the survival and proliferation of cancer cells. Additionally, HA promotes the formation of cancer stem cell (CSC) populations, which are hypothesized to be responsible for the initiation of tumors and therapy resistance. Recent studies have identified that the molecular weight of HA plays differing roles on both normal and cancer cell behavior. This review explores the role of HA in cancer progression and therapy resistance and how its molecular weight is important in regulating CSC populations, epithelial to mesenchymal transition (EMT), ATP binding cassette (ABC) transporter expression and receptor tyrosine kinase pathways.
Collapse
Affiliation(s)
- Zoe K Price
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, South Australia 5000, Australia.
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, South Australia 5000, Australia.
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, South Australia 5000, Australia.
| |
Collapse
|
17
|
Vitale DL, Spinelli FM, Del Dago D, Icardi A, Demarchi G, Caon I, García M, Bolontrade MF, Passi A, Cristina C, Alaniz L. Co-treatment of tumor cells with hyaluronan plus doxorubicin affects endothelial cell behavior independently of VEGF expression. Oncotarget 2018; 9:36585-36602. [PMID: 30564299 PMCID: PMC6290962 DOI: 10.18632/oncotarget.26379] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 11/07/2018] [Indexed: 12/24/2022] Open
Abstract
Hyaluronan, the main glycosaminoglycan of extracellular matrices, is concentrated in tissues with high cell proliferation and migration rates. In cancer, hyaluronan expression is altered and it becomes fragmented into low-molecular-weight forms, affecting mechanisms associated with cell proliferation, invasion, angiogenesis and multidrug resistance. Here, we analyzed the effect of low-molecular-weight hyaluronan on the response of T lymphoma, osteosarcoma, and mammary adenocarcinoma cell lines to the antineoplastic drug doxorubicin, and whether co-treatment with hyaluronan and doxorubicin modified the behavior of endothelial cells. Our aim was to associate the hyaluronan-doxorubicin response with angiogenic alterations in these tumors. After hyaluronan and doxorubicin co-treatment, hyaluronan altered drug accumulation and modulated the expression of ATP-binding cassette transporters in T-cell lymphoma cells. In contrast, no changes in drug accumulation were observed in cells from solid tumors, indicating that hyaluronan might not affect drug efflux. However, when we evaluated the effect on angiogenic mechanisms, the supernatant from tumor cells treated with doxorubicin exhibited a pro-angiogenic effect on endothelial cells. Hyaluronan-doxorubicin co-treatment increased migration and vessel formation in endothelial cells. This effect was independent of vascular endothelial growth factor but related to fibroblast growth factor-2 expression. Besides, we observed a pro-angiogenic effect on endothelial cells during hyaluronan and doxorubicin co-treatment in the in vivo murine model of T-cell lymphoma. Our results demonstrate for the first time that hyaluronan is a potential modulator of doxorubicin response by mechanisms that involve not only drug efflux but also angiogenic processes, providing an adverse tumor stroma during chemotherapy.
Collapse
Affiliation(s)
- Daiana L Vitale
- Laboratorio de Microambiente Tumoral-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| | - Fiorella M Spinelli
- Laboratorio de Microambiente Tumoral-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| | - Daiana Del Dago
- Laboratorio de Microambiente Tumoral-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| | - Antonella Icardi
- Laboratorio de Microambiente Tumoral-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| | - Gianina Demarchi
- Laboratorio de Fisiopatología de la Hipófisis-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| | - Ilaria Caon
- Dipartimento di Medicina e Chirurgia, Universitá degli Studio dell'Insubria, Varese, Italia
| | - Mariana García
- Laboratorio de Terapia Génica, IIMT-CONICET, Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Marcela F Bolontrade
- Laboratorio de Células Madre-Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Alberto Passi
- Dipartimento di Medicina e Chirurgia, Universitá degli Studio dell'Insubria, Varese, Italia
| | - Carolina Cristina
- Laboratorio de Fisiopatología de la Hipófisis-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| | - Laura Alaniz
- Laboratorio de Microambiente Tumoral-Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA, UNNOBA-CONICET), Junín, Buenos Aires, Argentina
| |
Collapse
|
18
|
Bohaumilitzky L, Huber AK, Stork EM, Wengert S, Woelfl F, Boehm H. A Trickster in Disguise: Hyaluronan's Ambivalent Roles in the Matrix. Front Oncol 2017; 7:242. [PMID: 29062810 PMCID: PMC5640889 DOI: 10.3389/fonc.2017.00242] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/22/2017] [Indexed: 02/04/2023] Open
Abstract
Hyaluronan (HA) is a simple but diverse glycosaminoglycan. It plays a major role in aging, cellular senescence, cancer, and tissue homeostasis. In which way HA affects the surrounding tissues greatly depends on the molecular weight of HA. Whereas high molecular weight HA is associated with homeostasis and protective effects, HA fragments tend to be linked to the pathologic state. Furthermore, the interaction of HA with its binding partners, the hyaladherins, such as CD44, is essential for sustaining tissue integrity and is likewise related to cancer. The naked mole rat, a rodent species, possesses a special form of very high molecular weight (vHMW) HA, which is associated with the extraordinary cancer resistance and longevity of those animals. This review addresses HA and its diverse facets: from HA synthesis to degradation, from oligomeric HA to vHMW-HA and from its beneficial properties to the involvement in pathologies. We further discuss the functions of HA in the naked mole rat and compare them to human conditions. Though intensively researched, this simple polymer bears some secrets that may hold the key for a better understanding of cellular processes and the development of diseases, such as cancer.
Collapse
Affiliation(s)
- Lena Bohaumilitzky
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Ann-Kathrin Huber
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Eva Maria Stork
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Simon Wengert
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Franziska Woelfl
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Heike Boehm
- CSF Biomaterials, Max Planck Institute for Medical Research, Heidelberg, Germany.,Department of Biophysical Chemistry, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Yang X, Ding Y, Xiao M, Liu X, Ruan J, Xue P. Anti-tumor compound RY10-4 suppresses multidrug resistance in MCF-7/ADR cells by inhibiting PI3K/Akt/NF-κB signaling. Chem Biol Interact 2017; 278:22-31. [PMID: 28987325 DOI: 10.1016/j.cbi.2017.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 11/20/2022]
Abstract
RY10-4, an anti-tumor agent, exerts cytotoxicity to various human cancer cell lines. However, few studies reported the effect of combined application of RY10-4 and chemotherapeutic drugs against cancer cells with multidrug resistance (MDR). In this study, P-glycoprotein (P-gp), which is reported to mediate MDR to anti-cancer drugs, was proved to be overexpressed in the adriamycin (ADR)-resistant human breast cancer cells, namely MCF-7/ADR cells. Furthermore, RY10-4 application resulted in a downregulation of P-gp in MCF-7/ADR cells, thus leading to higher chemosensitivity to ADR. Our study further demonstrated that the MDR phenomenon was under the control of the PI3K/Akt/NF-κB pathway, which was suppressed by RY10-4, leading to MDR reversal effects in MCF-7/ADR cells. In vivo, MCF-7/ADR cells were effectively suppressed by the combined ADR/RY10-4 treatment compared with the ADR-alone treatment. Taken together, these results demonstrated that RY10-4 reverses the MDR phenotype in MCF-7/ADR cells by suppressing the PI3K/Akt/NF-κB pathway.
Collapse
Affiliation(s)
- Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yufeng Ding
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Miao Xiao
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Liu
- Department of Anesthesiology, The People's Hospital of Hanchuan, Renmin Hospital of Wuhan University, 432300 Hubei Province, China
| | - Jinlan Ruan
- Key Laboratory of Natural Medicinal Chemistry and Resources Evaluation of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pingping Xue
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
20
|
Lu Y, Lin D, Li W, Yang X. Non-digestible stachyose promotes bioavailability of genistein through inhibiting intestinal degradation and first-pass metabolism of genistein in mice. Food Nutr Res 2017; 61:1369343. [PMID: 28970781 PMCID: PMC5613906 DOI: 10.1080/16546628.2017.1369343] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/15/2017] [Indexed: 12/16/2022] Open
Abstract
This study was designed to explore the molecular mechanism of stachyose in enhancing the gastrointestinal stability and absorption of soybean genistein in mice. Male Kunming mice in each group (n = 8) were administered by intragastric gavage with saline, stachyose (250 mg/kg·bw), genistein (100 mg/kg·bw), and stachyose (50, 250, and 500 mg/kg·bw) together with genistein (100 mg/kg·bw) for 4 consecutive weeks, respectively, and then their urine, feces, blood, gut, and liver were collected. UPLC-qTOF/MS analysis showed that levels of genistein and its metabolites (dihydrogenistein, genistein 7-sulfate sodium salt, genistein 4'-β-D-glucuronide, and genistein 7-β-D-glucuronide) in serum and urine were increased with an increase in stachyose dosages in mice. Furthermore, the feces level of genistein aglycone was also elevated by co-treatment of stachyose with genistein. However, the feces concentration of dihydrogenistein, a characteristic metabolite of genistein by gut microorganism, was decreased by stachyose administration in a dose-dependent manner. Additionally, the simultaneous administration with stachyose and genistein in mice could decrease intestinal SULT, UGT, P-gp, and MRP1 expression, relative to the treatment with individual stachyose or genistein. These results demonstrate that stachyose-mediated inhibition against the intestinal degradation of genistein and expression of phase II enzymes and efflux transporters can largely contribute to the elevated bioavailability of soybean genistein.
Collapse
Affiliation(s)
- Yalong Lu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Dehui Lin
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Wenfeng Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China.,Key Laboratory of Chongqing Municipality for Protection and Utility of Unique Plant Resources in the Wulingshan Region, Life Science and Technology Institute, Yangtze Normal University, Chongqing, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
21
|
Li W, Lu Y, Huang D, Han X, Yang X. Effects of stachyose on absorption and transportation of tea catechins in mice: possible role of Phase II metabolic enzymes and efflux transporters inhibition by stachyose. Food Nutr Res 2016; 60:32783. [PMID: 27782875 PMCID: PMC5081032 DOI: 10.3402/fnr.v60.32783] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 08/30/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022] Open
Abstract
Background Nutritional and absorption-promoting properties of stachyose combined with tea catechins (TC) have been revealed. However, the mechanism involved in non-digestible oligosaccharides-mediated enhancement of flavonoid absorption has largely remained elusive. Methods This study was designed to investigate the molecular mechanism of stachyose in enhancing absorption and transportation of TC in mice. Mice were orally pre-treated with stachyose (50, 250, and 500 mg/kg·bw) for 0–8 weeks, and 1 h before sacrifice, mice were treated with TC (250 mg/kg·bw). Results Gas chromatography-mass spectrometry analysis showed that serum concentrations of epicatechin, epigallocatechin, epicatechin gallate, and epigallocatechin gallate were dose- and time-dependently elevated with stachyose pre-treatment in mice. Furthermore, pre-treatment with stachyose in mice reduced intestinal sulfotransferase and uridine diphosphate-glucuronosyltransferase levels by 3.3–43.2% and 23.9–30.4%, relative to control mice, respectively. Moreover, intestinal P-glycoprotein and multidrug resistance-associated protein-1 contents were decreased in mice by pre-administration of stachyose in dose- and time-dependent manner. Conclusions This is the first time to demonstrate that suppression of Phase II metabolic enzymes and efflux transporters of TC in the intestine can play a major role in increasing absorption of TC by stachyose feeding.
Collapse
Affiliation(s)
- Wenfeng Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Yalong Lu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Di Huang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Xiao Han
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China;
| |
Collapse
|
22
|
Yuan Q, Zhan L, Zhang LL, Wang Q, Liu J, Jiang ZY, Hu XM, Yuan XC. Stanniocalcin 2 induces oxaliplatin resistance in colorectal cancer cells by upregulating P-glycoprotein. Can J Physiol Pharmacol 2016; 94:929-35. [PMID: 27245421 DOI: 10.1139/cjpp-2015-0530] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multidrug resistance (MDR) limits the anticancer effects of chemotherapy in patients with metastatic colorectal cancer (CRC). Oxaliplatin is a common component of combinational therapeutic regimens administered to patients with metastatic CRC; however, it is also used as a constituent of adjuvant therapy for patients at a risk of recurrent disease. In the present study, we investigated the role of stanniocalcin 2 (STC2) in chemoresistance. STC2 knockdown sensitized chemoresistant CRC cells to oxaliplatin. Moreover, the expression of exogenous STC2 in chemonaïve CRC cells induced oxaliplatin resistance. We confirmed that STC2 upregulated P-glycoprotein (P-gp) expression in CRC cells. Furthermore, shRNA against phosphoinositide 3-kinase (PI3K) or Akt inhibited the action of STC2 on P-gp upregulation and MDR in CRC. To our knowledge, this is the first report to demonstrate the induction of oxaliplatin resistance in CRC cells in response to STC2 stimulation of P-gp via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Qiong Yuan
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China.,c Drug Research Base of Cardiovascular and Cerebral Vascular, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Lin Zhan
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Li-Li Zhang
- b Department of Histology and Embryology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qiang Wang
- d Department of Immunology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Juan Liu
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhen-Yu Jiang
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xia-Min Hu
- a Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China.,c Drug Research Base of Cardiovascular and Cerebral Vascular, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xin-Chu Yuan
- b Department of Histology and Embryology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
23
|
Dosio F, Arpicco S, Stella B, Fattal E. Hyaluronic acid for anticancer drug and nucleic acid delivery. Adv Drug Deliv Rev 2016; 97:204-36. [PMID: 26592477 DOI: 10.1016/j.addr.2015.11.011] [Citation(s) in RCA: 403] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/06/2023]
Abstract
Hyaluronic acid (HA) is widely used in anticancer drug delivery, since it is biocompatible, biodegradable, non-toxic, and non-immunogenic; moreover, HA receptors are overexpressed on many tumor cells. Exploiting this ligand-receptor interaction, the use of HA is now a rapidly-growing platform for targeting CD44-overexpressing cells, to improve anticancer therapies. The rationale underlying approaches, chemical strategies, and recent advances in the use of HA to design drug carriers for delivering anticancer agents, are reviewed. Comprehensive descriptions are given of HA-based drug conjugates, particulate carriers (micelles, liposomes, nanoparticles, microparticles), inorganic nanostructures, and hydrogels, with particular emphasis on reports of preclinical/clinical results.
Collapse
|
24
|
Yu S, Cai X, Wu C, Wu L, Wang Y, Liu Y, Yu Z, Qin S, Ma F, Thiery JP, Chen L. Adhesion glycoprotein CD44 functions as an upstream regulator of a network connecting ERK, AKT and Hippo-YAP pathways in cancer progression. Oncotarget 2015; 6:2951-65. [PMID: 25605020 PMCID: PMC4413630 DOI: 10.18632/oncotarget.3095] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/24/2014] [Indexed: 12/14/2022] Open
Abstract
Targeted therapies are considered to be the future of cancer treatment. However, the mechanism through which intracellular signaling pathways coordinate to modulate oncogenesis remains to be elucidated. In this study, we describe a novel crosstalk among ERK, AKT and Hippo-YAP pathways, with CD44 as an upstream regulator. High cell density leads to activation of ERK and AKT but inactivation of YAP in cancer cells. CD44 modulates cell proliferation and cell cycle but not apoptosis. The expression and activity of cell cycle genes were cooperatively regulated by ERK, AKT and Hippo-YAP signaling pathways through CD44-mediated mechanisms. In addition, CD44 depletion abrogates cancer stem cell properties of tumor initiating cells. Taken together, we described a paradigm where CD44 functions as an upstream regulator sensing the extracellular environment to modulate ERK, AKT and Hippo-YAP pathways which cooperatively control downstream gene expression to modulate cell contact inhibition of proliferation, cell cycle progression and maintenance of tumor initiating cells. Our current study provides valuable information to design targeted therapeutic strategies in cancers.
Collapse
Affiliation(s)
- Shiyi Yu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| | - Xiuxiu Cai
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| | - Chenxi Wu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| | - Lele Wu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| | - Yuzhi Wang
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| | - Yan Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| | - Zhenghong Yu
- Department of Medical Oncology, Jinling Hospital, Nanjing, P.R. China
| | - Sheng Qin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Jean Paul Thiery
- Cancer Science Institute, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Liming Chen
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Institute of Life Science, Southeast University, Nanjing, P.R. China
| |
Collapse
|
25
|
Lompardía SL, Díaz M, Papademetrio DL, Mascaró M, Pibuel M, Álvarez E, Hajos SE. Hyaluronan oligomers sensitize chronic myeloid leukemia cell lines to the effect of Imatinib. Glycobiology 2015; 26:343-52. [DOI: 10.1093/glycob/cwv107] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
|
26
|
Schmaus A, Bauer J, Sleeman JP. Sugars in the microenvironment: the sticky problem of HA turnover in tumors. Cancer Metastasis Rev 2015; 33:1059-79. [PMID: 25324146 DOI: 10.1007/s10555-014-9532-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The properties and behavior of tumor cells are closely regulated by their microenvironment. Accordingly, stromal cells and extracellular matrix components can have a pronounced effect on cancer initiation, growth, and progression. The linear glycosaminoglycan hyaluronan (HA) is a major component of the extracellular matrix. Altered synthesis and degradation of HA in the tumor context has been implicated in many aspects of tumor biology. In particular, the accumulation of small HA oligosaccharides (sHA) in the tumor interstitial space may play a decisive role, due to the ability of sHA to activate a number of biological processes that are not modulated by high molecular weight (HMW)-HA. In this article, we review the normal physiological role and metabolism of HA and then survey the evidence implicating HA in tumor growth and progression, focusing in particular on the potential contribution of sHA to these processes.
Collapse
Affiliation(s)
- Anja Schmaus
- Institut für Toxikologie und Genetik, Karlsruhe Institute for Technology (KIT), Campus Nord, Postfach 3640, 76021, Karlsruhe, Germany
| | | | | |
Collapse
|
27
|
Skandalis SS, Gialeli C, Theocharis AD, Karamanos NK. Advances and advantages of nanomedicine in the pharmacological targeting of hyaluronan-CD44 interactions and signaling in cancer. Adv Cancer Res 2015; 123:277-317. [PMID: 25081534 DOI: 10.1016/b978-0-12-800092-2.00011-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Extensive experimental evidence in cell and animal tumor models show that hyaluronan-CD44 interactions are crucial in both malignancy and resistance to cancer therapy. Because of the intimate relationship between the hyaluronan-CD44 system and tumor cell survival and growth, it is an increasingly investigated area for applications to anticancer chemotherapeutics. Interference with the hyaluronan-CD44 interaction by targeting drugs to CD44, targeting drugs to the hyaluronan matrix, or interfering with hyaluronan matrix/tumor cell-associated CD44 interactions is a viable strategy for cancer treatment. Many of these methods can decrease tumor burden in animal models but have yet to show significant clinical utility. Recent advances in nanomedicine have offered new valuable tools for cancer detection, prevention, and treatment. The enhanced permeability and retention effect has served as key rationale for using nanoparticles to treat solid tumors. However, the targeted and uniform delivery of these particles to all regions of tumors in sufficient quantities requires optimization. An ideal nanocarrier should be equipped with selective ligands that are highly or exclusively expressed on target cells and thus endow the carriers with specific targeting capabilities. In this review, we describe how the hyaluronan-CD44 system may provide such an alternative in tumors expressing specific CD44 variants.
Collapse
Affiliation(s)
- Spyros S Skandalis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Chrisostomi Gialeli
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), Patras, Greece.
| |
Collapse
|
28
|
Abstract
Hyaluronic acid or hyaluronan (HA) is perhaps one of the most uncomplicated large polymers that regulates several normal physiological processes and, at the same time, contributes to the manifestation of a variety of chronic and acute diseases, including cancer. Members of the HA signaling pathway (HA synthases, HA receptors, and HYAL-1 hyaluronidase) have been experimentally shown to promote tumor growth, metastasis, and angiogenesis, and hence each of them is a potential target for cancer therapy. Furthermore, as these members are also overexpressed in a variety of carcinomas, targeting of the HA family is clinically relevant. A variety of targeted approaches have been developed to target various HA family members, including small-molecule inhibitors and antibody and vaccine therapies. These treatment approaches inhibit HA-mediated intracellular signaling that promotes tumor cell proliferation, motility, and invasion, as well as induction of endothelial cell functions. Being nontoxic, nonimmunogenic, and versatile for modifications, HA has been used in nanoparticle preparations for the targeted delivery of chemotherapy drugs and other anticancer compounds to tumor cells through interaction with cell-surface HA receptors. This review discusses basic and clinical translational aspects of targeting each HA family member and respective treatment approaches that have been described in the literature.
Collapse
|
29
|
Synergism of arsenic trioxide and MG132 in Raji cells attained by targeting BNIP3, autophagy, and mitochondria with low doses of valproic acid and vincristine. Eur J Cancer 2014; 50:3243-61. [DOI: 10.1016/j.ejca.2014.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 08/30/2014] [Accepted: 09/20/2014] [Indexed: 12/20/2022]
|
30
|
Silva R, Vilas-Boas V, Carmo H, Dinis-Oliveira RJ, Carvalho F, de Lourdes Bastos M, Remião F. Modulation of P-glycoprotein efflux pump: induction and activation as a therapeutic strategy. Pharmacol Ther 2014; 149:1-123. [PMID: 25435018 DOI: 10.1016/j.pharmthera.2014.11.013] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/03/2023]
Abstract
P-glycoprotein (P-gp) is an ATP-dependent efflux pump encoded by the MDR1 gene in humans, known to mediate multidrug resistance of neoplastic cells to cancer therapy. For several decades, P-gp inhibition has drawn many significant research efforts in an attempt to overcome this phenomenon. However, P-gp is also constitutively expressed in normal human epithelial tissues and, due to its broad substrate specificity, to its cellular polarized expression in many excretory and barrier tissues, and to its great efflux capacity, it can play a crucial role in limiting the absorption and distribution of harmful xenobiotics, by decreasing their intracellular accumulation. Such a defense mechanism can be of particular relevance at the intestinal level, by significantly reducing the intestinal absorption of the xenobiotic and, consequently, avoiding its access to the target organs. In this review, the current knowledge on this important efflux pump is summarized, and a new focus is brought on the therapeutic interest of inducing and/or activating P-gp for limiting the toxicity caused by its substrates. Several in vivo and in vitro studies validating the use of such a therapeutic strategy are discussed. An extensive literature search for reported P-gp inducers/activators and for the experimental models used in their characterization was conducted. Those studies demonstrate that effective antidotal pathways can be achieved by efficiently promoting the P-gp-mediated efflux of deleterious xenobiotics, resulting in a significant reduction in their intracellular levels and, consequently, in a significant reduction of their toxicity.
Collapse
Affiliation(s)
- Renata Silva
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Vânia Vilas-Boas
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Helena Carmo
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Ricardo Jorge Dinis-Oliveira
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; INFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, Advanced Institute of Health Sciences - North (ISCS-N), CESPU, CRL, Gandra, Portugal; Department of Legal Medicine and Forensic Sciences, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Félix Carvalho
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
31
|
Bonneau C, Rouzier R, Geyl C, Cortez A, Castela M, Lis R, Daraï E, Touboul C. Predictive markers of chemoresistance in advanced stages epithelial ovarian carcinoma. Gynecol Oncol 2014; 136:112-20. [PMID: 25449309 DOI: 10.1016/j.ygyno.2014.10.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/22/2014] [Accepted: 10/27/2014] [Indexed: 01/06/2023]
Abstract
OBJECTIVE DNA repair mechanisms, environment-mediated drug resistance and cancer initiating cells (CIC) are three major research concepts that can explain the chemoresistance of epithelial ovarian cancer (EOC). The objective was to test if changes in the expression of potential markers associated with drug resistance before and after chemotherapy would correlate with platinum resistance, defined as a recurrence within the first year after chemotherapy cessation, and with survival, in advanced EOC. METHODS We included 32 patients with stage IIIC-IV EOC who underwent laparoscopy to evaluate the extent of carcinomatosis, neoadjuvant chemotherapy (carboplatin/taxol) and interval surgery. Biopsies taken during the initial laparoscopies and interval surgeries were evaluated using immunohistochemistry for the expression of 7 proteins: CD117, CD44 and ALDH1 to evaluate CIC; IL-6, IL-8 and BMP2 to evaluate environment-mediated drug resistance; and ERCC1 to evaluate DNA repair. Expression measurements were correlated with platin resistance and survival. The markers' relevance was confirmed in vitro using chemoresistance tests and flow cytometric measurements of the proportion of CD44+ cells. RESULTS 17 patients were chemoresistant and 15 patients were chemosensitive. We observed increases in CD44, IL-6 and ERCC1 expression and stable ALDH1, CD117, IL-8, and BMP2 expression. Reduced expression of cancer initiating cell markers and increased expression of environment-mediated drug resistance markers were associated with poor prognosis. We also demonstrated that CD44+ cells had survival advantages in vitro. CONCLUSIONS Changes in CD44 and IL-8 expression on tumor cells appeared to correlate with overall survival and should be further tested as predictors of chemoresistance using larger cohort.
Collapse
Affiliation(s)
- Claire Bonneau
- UMRS 938 INSERM, Université Pierre et Marie Curie-Paris 6, 27 rue de Chaligny, 75571 Paris cedex 12, France; Department of Gynecology-Obstetrics and Reproductive Medicine, Hôpitaux Universitaires Paris-Est, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Institut Universitaire de Cancérologie, Université Pierre et Marie Curie, 4 Rue de la Chine, 75020 Paris, France
| | - Roman Rouzier
- Department of Surgery, Institut Curie, Paris Saint-Cloud, Université Versailles-Saint-Quentin-en-Yvelines, 35 Rue Dailly, 92210 Saint-Cloud, France
| | - Caroline Geyl
- UMR INSERM U965, Angiogenèse et Recherche translationnelle, Hôpital Lariboisière, 49 bd de la chapelle, 75010 Paris, France
| | - Annie Cortez
- Department of Pathology, Hôpital Tenon, 4 Rue de la Chine, 75020 Paris, France
| | - Mathieu Castela
- UMRS 938 INSERM, Université Pierre et Marie Curie-Paris 6, 27 rue de Chaligny, 75571 Paris cedex 12, France
| | - Raphael Lis
- Weill Cornell Medical College, Department of Genetic Medicine, Shahin Rafii' Laboratory, Ansary Stem Cell Institute, 510 East 70th Street, Building A, New York, NY 10065, USA
| | - Emile Daraï
- UMRS 938 INSERM, Université Pierre et Marie Curie-Paris 6, 27 rue de Chaligny, 75571 Paris cedex 12, France; Department of Gynecology-Obstetrics and Reproductive Medicine, Hôpitaux Universitaires Paris-Est, Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Institut Universitaire de Cancérologie, Université Pierre et Marie Curie, 4 Rue de la Chine, 75020 Paris, France
| | - Cyril Touboul
- UMR INSERM U965, Angiogenèse et Recherche translationnelle, Hôpital Lariboisière, 49 bd de la chapelle, 75010 Paris, France; Department of Obstetrics and Gynecology, Hôpital Intercommunal de Créteil, Université Paris Est, Paris XII, 40 avenue de Verdun, 94000 Créteil, France.
| |
Collapse
|
32
|
Misra S, Ghatak S, Vyas A, O’Brien P, Markwald RR, Khetmalas M, Hascall VC, McCarthy JB, Karamanos NK, Tammi MI, Tammi RH, Prestwitch GD, Padhye S. Isothiocyanate analogs targeting CD44 receptor as an effective strategy against colon cancer. Med Chem Res 2014; 23:3836-3851. [PMID: 25013352 PMCID: PMC4084864 DOI: 10.1007/s00044-014-0958-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammatory pathway plays an important role in tumor cell progression of colorectal cancers. Although colon cancer is considered as one of the leading causes of death worldwide, very few drugs are available for its effective treatment. Many studies have examined the effects of specific COX-2 and 5-LOX inhibitors on human colorectal cancer, but the role of isothiocyanates (ITSCs) as COX-LOX dual inhibitors engaged in hyaluronan-CD44 interaction has not been studied. In the present work, we report series of ITSC analogs incorporating bioisosteric thiosemicarbazone moiety. These inhibitors are effective against panel of human colon cancer cell lines including COX-2 positive HCA-7, HT-29 cells lines, and hyaluronan synthase-2 (Has2) enzyme over-expressing transformed intestinal epithelial Apc10.1Has2 cells. Specifically, our findings indicate that HA-CD44v6-mediated COX-2/5-LOX signaling mediate survivin production, which in turn, supports anti-apoptosis and chemo-resistance leading to colon cancer cell survival. The over-expression of CD44v6shRNA as well as ITSC treatment significantly decreases the survival of colon cancer cells. The present results thus offer an opportunity to evolve potent inhibitors of HA synthesis and CD44v6 pathway and thus underscoring the importance of the ITSC analogs as chemopreventive agents for targeting HA/CD44v6 pathway.
Collapse
Affiliation(s)
- Suniti Misra
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Shibnath Ghatak
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alok Vyas
- ISTRA, Department of Chemistry, Abeda Inamdar College, University of Pune, Pune 411001, India
- Department of Bioinformatics and Computer Science, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Paul O’Brien
- Hematology/Oncology Division, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Roger R. Markwald
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Madhukar Khetmalas
- Department of Bioinformatics and Computer Science, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, India
| | - Vincent C. Hascall
- Department of Biomedical Engineering/ND20, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - James B. McCarthy
- Department of Laboratory Medicine and Pathology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, USA
| | - Nikos K. Karamanos
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Markku I. Tammi
- University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Raija H. Tammi
- University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Glenn D. Prestwitch
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Subhash Padhye
- ISTRA, Department of Chemistry, Abeda Inamdar College, University of Pune, Pune 411001, India
| |
Collapse
|
33
|
Ma H, Zhou H, Li P, Song X, Miao X, Li Y, Jia L. Effect of ST3GAL 4 and FUT 7 on sialyl Lewis X synthesis and multidrug resistance in human acute myeloid leukemia. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1681-92. [PMID: 24953795 DOI: 10.1016/j.bbadis.2014.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/07/2014] [Accepted: 06/12/2014] [Indexed: 01/12/2023]
Abstract
Sialyl Lewis X (sLe X, CD15s) is a key antigen produced on tumor cell surfaces during multidrug resistance (MDR) development. The present study investigated the effect of α1, 3 fucosyltransferase VII (FucT VII) and α2, 3 sialyltransferase IV (ST3Gal IV) on sLe X oligosaccharides synthesis as well as their impact on MDR development in acute myeloid leukemia cells (AML). FUT7 and ST3GAL4 were overexpressed in three AML MDR cells and bone marrow mononuclear cells (BMMC) of AML patients with MDR by real-time polymerase chain reaction (PCR). A close association was found between the expression levels of FUT7 and ST3GAL4 and the amount of sLe X oligosaccharides, as well as the phenotypic variation of MDR of HL60 and HL60/ADR cells both in vitro and in vivo. Manipulation of these two genes' expression modulated the activity of phosphoinositide-3 kinase (PI3K)/Akt signaling pathway, thereby regulating the proportionally mutative expression of P-glycoprotein (P-gp) and multidrug resistance related protein 1 (MRP1), both of which are known to be involved in MDR. Blocking the PI3K/Akt pathway by its specific inhibitor LY294002 or Akt short hairpin RNA (shRNA) resulted in the reduced MDR of HL60/ADR cells. This study indicated that sLe X involved in the development of MDR of AML cells probably through FUT7 and ST3GAL4 regulating the activity of PI3K/Akt signaling pathway and the expression of P-gp and MRP1.
Collapse
Affiliation(s)
- Hongye Ma
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Huimin Zhou
- Department of Microbiology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Peng Li
- Department of Bone Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Xiaobo Song
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Xiaoyan Miao
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Yanping Li
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China.
| |
Collapse
|
34
|
Modification of sialylation is associated with multidrug resistance in human acute myeloid leukemia. Oncogene 2014; 34:726-40. [PMID: 24531716 DOI: 10.1038/onc.2014.7] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/07/2013] [Accepted: 01/06/2014] [Indexed: 12/15/2022]
Abstract
Aberrant cell surface sialylation patterns have been shown to correlate with tumor progression and metastasis. However, the role of sialylation regulation of cancer multidrug resistance (MDR) remains poorly understood. This study investigated sialylation in modification on MDR in acute myeloid leukemia (AML). Using mass spectrometry (MS) analysis, the composition profiling of sialylated N-glycans differed in three pairs of AML cell lines. Real-time PCR showed the differential expressional profiles of 20 sialyltransferase (ST) genes in the both AML cell lines and bone marrow mononuclear cells (BMMCs) of AML patients. The expression levels of ST3GAL5 and ST8SIA4 were detected, which were overexpressed in HL60 and HL60/adriamycin-resistant (ADR) cells. The altered levels of ST3GAL5 and ST8SIA4 were found in close association with the MDR phenotype changing of HL60 and HL60/ADR cells both in vitro and in vivo. Further data demonstrated that manipulation of these two genes' expression modulated the activity of phosphoinositide-3 kinase (PI3K)/Akt signaling pathway and its downstream target thus regulated the proportionally mutative expression of P-glycoprotein (P-gp) and MDR-related protein 1 (MRP1), both of which are known to be involved in MDR. Blocking the PI3K/Akt pathway by its specific inhibitor LY294002 or by Akt small interfering RNA resulted in the reduced chemosensitivity of HL60/ADR cells. Therefore, this study indicated that sialylation involved in the development of MDR of AML cells probably through ST3GAL5 or ST8SIA4 regulating the activity of PI3K/Akt signaling and the expression of P-gp and MRP1.
Collapse
|
35
|
Ma H, Cheng L, Hao K, Li Y, Song X, Zhou H, Jia L. Reversal effect of ST6GAL 1 on multidrug resistance in human leukemia by regulating the PI3K/Akt pathway and the expression of P-gp and MRP1. PLoS One 2014; 9:e85113. [PMID: 24454800 PMCID: PMC3894187 DOI: 10.1371/journal.pone.0085113] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 12/01/2013] [Indexed: 01/06/2023] Open
Abstract
β-galactoside α2, 6-sialyltransferse gene (ST6GAL) family has two members, which encode corresponding enzymes ST6Gal I and ST6Gal II. The present atudy was to investigate whether and how ST6GAL family involved in multidrug resistance (MDR) in human leukemia cell lines and bone marrow mononuclear cells (BMMC) of leukemia patients. Real-time PCR showed a high expression level of ST6GAL1 gene in both MDR cells and BMMCs (*P<0.05). Alternation of ST6GAL1 levels had a significant impact on drug-resistant phenotype changing of K562 and K562/ADR cells both in vitro and in vivo. However, no significant changes were observed of ST6GAL2 gene. Further data revealed that manipulation of ST6GAL1 modulated the activity of phosphoinositide 3 kinase (PI3K)/Akt signaling and consequently regulated the expression of P-glycoprotein (P-gp, *P<0.05) and multidrug resistance related protein 1 (MRP1, *P<0.05), which are both known to be associated with MDR. Therefore we postulate that ST6GAL1 is responsible for the development of MDR in human leukemia cells probably through medicating the activity of PI3K/Akt signaling and the expression of P-gp and MRP1.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adolescent
- Adult
- Aged
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Child
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Female
- Gene Expression
- Gene Expression Regulation, Leukemic
- Gene Knockdown Techniques
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myeloid, Acute/enzymology
- Male
- Middle Aged
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering/genetics
- Sialyltransferases/genetics
- Sialyltransferases/metabolism
- Young Adult
Collapse
Affiliation(s)
- Hongye Ma
- College of Laboratory Medicine, Medical University, Dalian, Liaoning Province, China
| | - Lei Cheng
- Department of Laparoscopic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Keji Hao
- Department of Nuclear Medicine, People's Hospital of Peking University, Beijing, China
| | - Yanping Li
- College of Laboratory Medicine, Medical University, Dalian, Liaoning Province, China
| | - Xiaobo Song
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | - Huimin Zhou
- Department of Microbiology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Jia
- College of Laboratory Medicine, Medical University, Dalian, Liaoning Province, China
- * E-mail:
| |
Collapse
|
36
|
Ricciardelli C, Ween MP, Lokman NA, Tan IA, Pyragius CE, Oehler MK. Chemotherapy-induced hyaluronan production: a novel chemoresistance mechanism in ovarian cancer. BMC Cancer 2013; 13:476. [PMID: 24124770 PMCID: PMC3852938 DOI: 10.1186/1471-2407-13-476] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/01/2013] [Indexed: 12/15/2022] Open
Abstract
Background Hyaluronan (HA) an important component of the extracellular matrix, has been linked to tumor progression and drug resistance in several malignancies. However, limited data is available for ovarian cancer. This study investigated the role of hyaluronan (HA) and a potential link between the HA-CD44 pathway and membrane ATP binding cassette (ABC) transporter proteins in ovarian cancer chemoresistance. Methods We investigated the ability of HA to block the cytotoxic effects of the chemotherapy drug carboplatin, and to regulate the expression of ABC transporters in ovarian cancer cells. We also examined HA serum levels in ovarian cancer patients prior to and following chemotherapy and assessed its prognostic relevance. Results HA increased the survival of carboplatin treated ovarian cancer cells expressing the HA receptor, CD44 (OVCAR-5 and OV-90). Carboplatin significantly increased expression of HAS2, HAS3 and ABCC2 and HA secretion in ovarian cancer cell conditioned media. Serum HA levels were significantly increased in patients following platinum based chemotherapy and at both 1st and 2nd recurrence when compared with HA levels prior to treatment. High serum HA levels (>50 μg/ml) prior to chemotherapy treatment were associated with significantly reduced progression-free (P = 0.014) and overall survival (P = 0.036). HA production in ovarian cancer cells was increased in cancer tissues collected following chemotherapy treatment and at recurrence. Furthermore HA treatment significantly increased the expression of ABC drug transporters (ABCB3, ABCC1, ABCC2, and ABCC3), but only in ovarian cancer cells expressing CD44. The effects of HA and carboplatin on ABC transporter expression in ovarian cancer cells could be abrogated by HA oligomer treatment. Importantly, HA oligomers increased the sensitivity of chemoresistant SKOV3 cells to carboplatin. Conclusions Our findings indicate that carboplatin chemotherapy induces HA production which can contribute to chemoresistance by regulating ABC transporter expression. The HA-CD44 signaling pathway is therefore a promising target in platinum resistant ovarian cancer.
Collapse
Affiliation(s)
- Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, Research Centre for Reproductive Health, Robinson Institute, University of Adelaide, Adelaide 5005, South Australia, Australia.
| | | | | | | | | | | |
Collapse
|
37
|
Lompardía SL, Papademetrio DL, Mascaró M, Álvarez EMDC, Hajos SE. Human leukemic cell lines synthesize hyaluronan to avoid senescence and resist chemotherapy. Glycobiology 2013; 23:1463-76. [PMID: 24013961 DOI: 10.1093/glycob/cwt074] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hyaluronan (HA) is one of the major components of the extracellular matrix. Several solid tumors produce high levels of HA, which promotes survival and multidrug resistance (MDR). HA oligomers (oHAs) can block HA effects. However, little is known about the role of HA in hematological malignancies. The aim of this work was to determine whether HA or its oligomers can modulate the proliferation of leukemia cells as well as their effect on MDR. Receptors and signaling pathways involved were also analyzed. For this purpose, the human leukemic cell lines K562 and Kv562, which are sensitive and resistant to Vincristine (VCR), respectively, were used. We demonstrated that HA induced cell proliferation in both cell lines. On K562 cells, this effect was mediated by cluster differentiation 44 (CD44) and activation of both phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways, whereas on Kv562 cells, the effect was mediated by receptor for hyaluronan-mediated motility (RHAMM) and PI3K/Akt activation. The inhibition of HA synthesis by 4-methylumbelliferone (4MU) decreased cell line proliferation and sensitized Kv562 to the effect of VCR through P-glycoprotein (Pgp) inhibition, in both cases with senescence induction. Moreover, oHAs inhibited K562 proliferation mediated by CD44 as well as Akt and ERK down-regulation. Furthermore, oHAs sensitized Kv562 cells to VCR by Pgp inhibition inducing senescence. We postulate that the synthesis of HA would promote leukemia progression mediated by the triggering of the above-mentioned proliferative signals. These findings highlight the potential use of oHAs and 4MU as coadjuvant for drug-resistant leukemia.
Collapse
Affiliation(s)
- Silvina Laura Lompardía
- Department of Immunology, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), IDEHU-CONICET, Buenos Aires 1113, Argentina
| | | | | | | | | |
Collapse
|
38
|
Cai Q, Huang H, Qian D, Chen K, Luo J, Tian Y, Lin T, Lin T. 13-methyltetradecanoic acid exhibits anti-tumor activity on T-cell lymphomas in vitro and in vivo by down-regulating p-AKT and activating caspase-3. PLoS One 2013; 8:e65308. [PMID: 23762338 PMCID: PMC3676434 DOI: 10.1371/journal.pone.0065308] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 04/29/2013] [Indexed: 12/11/2022] Open
Abstract
13-Methyltetradecanoic acid (13-MTD), a saturated branched-chain fatty acid purified from soy fermentation products, induces apoptosis in human cancer cells. We investigated the inhibitory effects and mechanism of action of 13-MTD on T-cell non-Hodgkin’s lymphoma (T-NHL) cell lines both in vitro and in vivo. Growth inhibition in response to 13-MTD was evaluated by the cell counting kit-8 (CCK-8) assay in three T-NHL cell lines (Jurkat, Hut78, EL4 cells). Flow cytometry analyses were used to monitor the cell cycle and apoptosis. Proteins involved in 13-MTD-induced apoptosis were examined in Jurkat cells by western blotting. We found that 13-MTD inhibited proliferation and induced the apoptosis of T-NHL cell lines. 13-MTD treatment also induced a concentration-dependent arrest of Jurkat cells in the G1-phase. During 13-MTD-induced apoptosis in Jurkat cells, the cleavage of caspase-3 and poly ADP-ribose polymerase (PARP, a caspase enzymolysis product) were detected after incubation for 2 h, and increased after extending the incubation time. However, there was no change in the expression of Bcl-2 or c-myc proteins. The appearance of apoptotic Jurkat cells was accompanied by the inhibition of AKT and nuclear factor-kappa B (NF-κB) phosphorylation. In addition, 13-MTD could also effectively inhibit the growth of T-NHL tumors in vivo in a xenograft model. The tumor inhibition rate in the experimental group was 40%. These data indicate that 13-MTD inhibits proliferation and induces apoptosis through the down-regulation of AKT phosphorylation followed by caspase activation, which may provide a new approach for treating T-cell lymphomas.
Collapse
Affiliation(s)
- Qingqing Cai
- Department of Medical Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory of Oncology in Southern China, Guangzhou, P. R. China
| | - Huiqiang Huang
- Department of Medical Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory of Oncology in Southern China, Guangzhou, P. R. China
| | - Dong Qian
- State Key Laboratory of Oncology in Southern China, Guangzhou, P. R. China
| | - Kailin Chen
- Department of Medical Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory of Oncology in Southern China, Guangzhou, P. R. China
| | - Junhua Luo
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P. R. China
- Lin Bai-xin Medical Research Center, the Second Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Ying Tian
- State Key Laboratory of Oncology in Southern China, Guangzhou, P. R. China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, P. R. China
- Lin Bai-xin Medical Research Center, the Second Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
- * E-mail: (Tongyu Lin); (Tianxin Lin)
| | - Tongyu Lin
- Department of Medical Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory of Oncology in Southern China, Guangzhou, P. R. China
- * E-mail: (Tongyu Lin); (Tianxin Lin)
| |
Collapse
|
39
|
Baeva LF, Lyle DB, Rios M, Langone JJ, Lightfoote MM. Different molecular weight hyaluronic acid effects on human macrophage interleukin 1β production. J Biomed Mater Res A 2013; 102:305-14. [PMID: 23533059 DOI: 10.1002/jbm.a.34704] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 02/01/2013] [Accepted: 02/19/2013] [Indexed: 02/04/2023]
Abstract
This study examined the effect of hyaluronan (HA) molecular weight on immune response. HA with molecular weights ranging from the unitary disaccharide unit (400 Da) up to 1.7 × 10(6) Da and with very low endotoxin contamination level (less than 0.03 EU/mg) was used. Primary human monocyte/macrophage cultures were assayed for IL-1β production under a variety of inflammatory conditions with or without HA. Under the highest inflammatory states, production of interleukin 1β (IL-1β) was suppressed in the presence of high molecular weight hyaluronan (HMW-HA) and in the presence of low molecular weight hyaluronan (LMW-HA) at mg/mL concentrations. There was variability in the sensitivity of the response to HA fragments with MW below 5000 Da at micromolar concentrations. There was variability in IL-1β cytokine productions from donor to donor in unstimulated human cell cultures. This study supplements our previous published study that investigated the immunogenic effect of HA molecular weights using murine cell line RAW264.6, rat splenocytes, and rat adherent differentiated primary macrophages. These data support the hypothesis that if the amount of endotoxin is reduced to an extremely low level, LMW-HA may not directly provoke normal tissue macrophage-mediated inflammatory reactions.
Collapse
Affiliation(s)
- Larissa F Baeva
- Division of Biology, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, FDA, 10903 New Hampshire Ave, Silver Spring, Maryland, 20993-002
| | | | | | | | | |
Collapse
|
40
|
Tsubaki M, Satou T, Itoh T, Imano M, Komai M, Nishinobo M, Yamashita M, Yanae M, Yamazoe Y, Nishida S. Overexpression of MDR1 and survivin, and decreased Bim expression mediate multidrug-resistance in multiple myeloma cells. Leuk Res 2012; 36:1315-22. [PMID: 22819074 DOI: 10.1016/j.leukres.2012.07.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 06/07/2012] [Accepted: 07/02/2012] [Indexed: 01/12/2023]
Abstract
Multidrug resistance represents a major obstacle for the chemotherapy of a wide variety of human tumors. To investigate the underlying mechanisms associated with resistance to anti-cancer drugs, we established anti-cancer drug-resistant multiple myeloma (MM) cell lines RPMI8226/ADM, RPMI8226/VCR, RPMI8226/DEX, and RPMI8226/L-PAM, the 50% inhibitory concentration values of which were 77-, 58-, 79-, and 30-fold higher than their parental cell lines, respectively. The resistant cell lines overexpressed MDR1 and survivin, or showed decreased Bim expression. These results indicated that regulating these factors with inhibitors might be a viable approach to increasing the susceptibility of quiescent MM cells to chemotherapy.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kinki University School of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cai Q, Deng H, Xie D, Lin T, Lin T. Phosphorylated AKT Protein Is Overexpressed in Human Peripheral T-cell Lymphomas and Predicts Decreased Patient Survival. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:106-12. [DOI: 10.1016/j.clml.2011.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 11/15/2011] [Accepted: 12/12/2011] [Indexed: 01/06/2023]
|
42
|
Urakawa H, Nishida Y, Knudson W, Knudson CB, Arai E, Kozawa E, Futamura N, Wasa J, Ishiguro N. Therapeutic potential of hyaluronan oligosaccharides for bone metastasis of breast cancer. J Orthop Res 2012; 30:662-72. [PMID: 21913222 DOI: 10.1002/jor.21557] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 08/24/2011] [Indexed: 02/04/2023]
Abstract
Hyaluronan (HA) oligosaccharides were reported to have suppressive effects on various malignant tumors via disruption of receptor HA interactions. However, no studies have focused on the effects of HA oligosaccharides on bone metastasis of breast cancer. In this study, we clarified the effective size of HA oligosaccharides required to inhibit cell growth in the highly invasive breast cancer cell line, MDA-MB-231 cells. Based on the results of cell growth assay, we subsequently analyzed the effects of HA tetrasaccharides, HA decasaccharides, and high molecular weight HA on the other breast cancer cell behaviors in vitro and breast cancer bone metastasis in vivo. HA decasaccharides significantly inhibited cell growth, motility, and invasion, whereas tetrasaccharides did not. HAS2 mRNA expression was altered after the treatment with both tetrasaccharides and decasaccharides. Phosphorylation of Akt was suppressed after 1 h treatment with HA decasaccharides, and the effect was partially reversed by anti-CD44 monoclonal antibody. In vivo, local application of HA decasaccharides inhibited the expansion of osteolytic lesions in tibia on soft X-rays using mouse bone metastasis model of breast cancer. Histological analysis revealed HA accumulation in bone metastatic lesions was perturbed by decasaccharides. These results suggest that HA oligosaccharides suppressed progression of bone metastasis in breast cancer via interruption of endogenous HA-CD44 interaction, and as such, can be a novel therapeutic candidate to limit bone metastasis of breast cancer.
Collapse
Affiliation(s)
- Hiroshi Urakawa
- Department of Orthopedic Surgery, Nagoya University Graduate School and School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8550, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Facompre N, Nakagawa H, Herlyn M, Basu D. Stem-like cells and therapy resistance in squamous cell carcinomas. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 65:235-65. [PMID: 22959028 DOI: 10.1016/b978-0-12-397927-8.00008-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs) within squamous cell carcinomas (SCCs) are hypothesized to contribute to chemotherapy and radiation resistance and represent potentially useful pharmacologic targets. Hallmarks of the stem cell phenotype that may contribute to therapy resistance of CSCs include quiescence, evasion of apoptosis, resistance to DNA damage, and expression of drug transporter pumps. A variety of CSC populations within SCCs of the head and neck and esophagus have been defined tentatively, based on diverse surface markers and functional assays. Stem-like self-renewal and differentiation capacities of these SCC subpopulations are supported by sphere formation and clonogenicity assays in vitro as well as limiting dilution studies in xenograft models. Early evidence supports a role for SCC CSCs in intrinsic therapy resistance, while detailed mechanisms by which these subpopulations evade treatment remain to be defined. Development of novel SCC therapies will be aided by pursuing such mechanisms as well as refining current definitions for CSCs and clarifying their relevance to hierarchical versus dynamic models of stemness.
Collapse
Affiliation(s)
- Nicole Facompre
- Department of Otorhinolaryngology--Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA; The Wistar Institute, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
44
|
Han ME, Jeon TY, Hwang SH, Lee YS, Kim HJ, Shim HE, Yoon S, Baek SY, Kim BS, Kang CD, Oh SO. Cancer spheres from gastric cancer patients provide an ideal model system for cancer stem cell research. Cell Mol Life Sci 2011; 68:3589-605. [PMID: 21448722 PMCID: PMC11114917 DOI: 10.1007/s00018-011-0672-z] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 02/06/2011] [Accepted: 02/24/2011] [Indexed: 12/14/2022]
Abstract
Cancer stem cells have been hypothesized to drive the growth and metastasis of tumors. Because they need to be targeted for cancer treatment, they have been isolated from many solid cancers. However, cancer stem cells from primary human gastric cancer tissues have not been isolated as yet. For the isolation, we used two cell surface markers: the epithelial cell adhesion molecule (EpCAM) and CD44. When analyzed by flow cytometry, the EpCAM(+)/CD44(+) population accounts for 4.5% of tumor cells. EpCAM(+)/CD44(+) gastric cancer cells formed tumors in immunocompromised mice; however, EpCAM(-)/CD44(-), EpCAM(+)/CD44(-) and EpCAM(-)/CD44(+) cells failed to do so. Xenografts of EpCAM(+)/CD44(+) gastric cancer cells maintained a differentiated phenotype and reproduced the morphological and phenotypical heterogeneity of the original gastric tumor tissues. The tumorigenic subpopulation was serially passaged for several generations without significant phenotypic alterations. Moreover, EpCAM(+)/CD44(+), but not EpCAM(-)/CD44(-), EpCAM(+)/CD44(-) or EpCAM(-)/CD44(+) cells grew exponentially in vitro as cancer spheres in serum-free medium, maintaining the tumorigenicity. Interestingly, a single cancer stem cell generated a cancer sphere that contained various differentiated cells, supporting multi-potency and self-renewal of a cancer stem cell. EpCAM(+)/CD44(+) cells had greater resistance to anti-cancer drugs than other subpopulation cells. The above in vivo and in vitro results suggest that cancer stem cells, which are enriched in the EpCAM(+)/CD44(+) subpopulation of gastric cancer cells, provide an ideal model system for cancer stem cell research.
Collapse
Affiliation(s)
- Myoung-Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| | - Tae-Yong Jeon
- Department of Surgery, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sun-Hwi Hwang
- Department of Surgery, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Young-Suk Lee
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Hyun-Jung Kim
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| | - Hye-Eun Shim
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Sun-Yong Baek
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Bong-Seon Kim
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Chi-Dug Kang
- Department of Biochemistry, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
45
|
Fang XJ, Jiang H, Zhao XP, Jiang WM. The role of a new CD44st in increasing the invasion capability of the human breast cancer cell line MCF-7. BMC Cancer 2011; 11:290. [PMID: 21749678 PMCID: PMC3161032 DOI: 10.1186/1471-2407-11-290] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 07/12/2011] [Indexed: 12/18/2022] Open
Abstract
Background CD44, a hyaluronan (HA) receptor, is a multistructural and multifunctional cell surface molecule involved in cell proliferation, cell differentiation, cell migration, angiogenesis, presentation of cytokines, chemokines and growth factors to the corresponding receptors, and docking of proteases at the cell membrane, as well as in signaling for cell survival. The CD44 gene contains 20 exons that are alternatively spliced, giving rise to many CD44 isoforms, perhaps including tumor-specific sequences. Methods Reverse transcriptase polymerase chain reaction (RT-PCR) and Western blotting were used to detect CD44st mRNA and CD44 protein in sensitive MCF-7, Lovo, K562 and HL-60 cell lines as well as their parental counterparts, respectively. The full length cDNA encoding CD44st was obtained from the total RNA isolated from MCF-7/Adr cells by RT-PCR, and subcloned into the pMD19-T vector. The CD44st gene sequence and open reading frame were confirmed by restriction enzyme analysis and nucleotide sequencing, and then inserted into the eukaryotic expression vector pcDNA3.1. The pcDNA3.1-CD44st was transfected into MCF-7 cells using Lipofectamine. After transfection, the positive clones were obtained by G418 screening. The changes of the MMP-2 and MMP-9 genes and protein levels were detected by RT-PCR and gelatin zymography, respectively. The number of the cells penetrating through the artificial matrix membrane in each group (MCF-7, MCF-7+HA, MCF-7/neo, MCF-7/neo+HA, MCF-7/CD44st, MCF-7/CD44st+HA and MCF-7/CD44st+Anti-CD44+HA) was counted to compare the change of the invasion capability regulated by the CD44st. Erk and P-Erk were investigated by Western blotting to approach the molecular mechanisms of MMP-2 and MMP-9 expression regulated by the CD44st. Results Sensitive MCF-7, Lovo, K562 and HL-60 cells did not contain CD44st mRNA and CD44 protein. In contrast, the multidrug resistance MCF-7/Adr, Lovo/Adr, K562/Adr and HL-60/Adr cells expressed CD44st mRNA and CD44 protein. The CD44st mRNA gene sequence was successfully cloned into the recombinant vector pcDNA3.1 and identified by the two restriction enzymes. It was confirmed that the reconstructed plasmid contained the gene sequence of CD44st that was composed of exons 1 to 4, 16 to 17, and 1 to 205 bases of exons 18. The new gene sequence was sent to NCBI for publication, and obtained the registration number FJ216964. The up-regulated level of the mRNA of the CD44 gene and the CD44 protein were detected, respectively, by RT-PCR and flow cytometry in MCF-7 cells transfected with pcDNA3.1-CD44st. The invasiveness of the cells and the activity of MMP-2 and MMP-9 were clearly activated by HA treatment, and blocked by CD44 neutralizing antibody. MCF-7/CD44st cells pretreated with the neutralizing antibody against CD44, and the inhibitor of MAPKs signaling pathway, could strongly block the expression of P-Erk. Conclusions A new CD44st was expressed in multidrug resistant MCF-7/Adr, Lovo/Adr, K562/Adr and HL-60/Adr cells. The expression vector pcDNA3.1-CD44st was cloned and constructed successfully, and stably transfected into MCF-7 cells. HA could interact with the new CD44st and regulate the expression of MMP-2 and MMP-9, which could increase the invasion capability of MCF-7 cells through the Ras/MAPK signaling pathway.
Collapse
Affiliation(s)
- Xin Jian Fang
- Department of Medical Oncology, The second People's Hospital of Lianyungang (Lianyungang Hospital affiliated to Bengbu medical college), No. 41, Hailian east Road, Lianyungang, Jiangsu 222000, The People's Republic of China
| | | | | | | |
Collapse
|
46
|
Qin Z, Dai L, Bratoeva M, Slomiany MG, Toole BP, Parsons C. Cooperative roles for emmprin and LYVE-1 in the regulation of chemoresistance for primary effusion lymphoma. Leukemia 2011; 25:1598-609. [PMID: 21660043 DOI: 10.1038/leu.2011.144] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Kaposi's sarcoma-associated herpesvirus is the causative agent of primary effusion lymphoma (PEL), for which cytotoxic chemotherapy represents the standard of care. The high mortality associated with PEL may be explained in part by resistance of these tumors to chemotherapy. The membrane-bound glycoprotein emmprin (CD147) enhances chemoresistance in tumors through effects on transporter expression, trafficking and interactions. Interactions between hyaluronan and hyaluronan receptors on the cell surface also facilitate emmprin-mediated chemoresistance. Whether emmprin or hyaluronan-receptor interactions regulate chemotherapeutic resistance for virus-associated malignancies is unknown. Using human PEL tumor cells, we found that PEL sensitivity to chemotherapy is directly proportional to expression of emmprin, the lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) and a drug transporter known as the breast cancer resistance protein/ABCG2 (BCRP), and that emmprin, LYVE-1 and BCRP interact with each other and colocalize on the PEL cell surface. In addition, we found that emmprin induces chemoresistance in PEL cells through upregulation of BCRP expression, and RNA interference targeting of emmprin, LYVE-1 or BCRP enhances PEL cell apoptosis induced by chemotherapy. Finally, disruption of hyaluronan-receptor interactions using small hyaluronan oligosaccharides reduces expression of emmprin and BCRP while sensitizing PEL cells to chemotherapy. Collectively, these data support interdependent roles for emmprin, LYVE-1 and BCRP in chemotherapeutic resistance for PEL.
Collapse
Affiliation(s)
- Z Qin
- Department of Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
47
|
Urakawa H, Nishida Y, Wasa J, Arai E, Zhuo L, Kimata K, Kozawa E, Futamura N, Ishiguro N. Inhibition of hyaluronan synthesis in breast cancer cells by 4-methylumbelliferone suppresses tumorigenicity in vitro and metastatic lesions of bone in vivo. Int J Cancer 2011; 130:454-66. [PMID: 21387290 DOI: 10.1002/ijc.26014] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 02/01/2011] [Accepted: 02/11/2011] [Indexed: 01/26/2023]
Abstract
Hyaluronan (HA) has been shown to play crucial roles in the tumorigenicity of malignant tumors. Previous studies demonstrated that inhibition of HA suppressed the tumorigenicity of various malignant tumors including breast cancer. 4-methylumbelliferone (MU) has been reported to inhibit HA synthesis in several cell types. However, few studies have focused on the effects of HA inhibition in breast cancer cells by MU, nor the effects on bone metastasis. We hypothesized that MU would suppress the progression of bone metastasis via inhibition of HA synthesis. Here, we investigated the effects of MU on HA expression in MDA-MB-231 breast cancer cell line in addition to their tumorigenicity in vitro and in vivo. HAS2 mRNA expression was downregulated after 6 and 24 hr treatment with MU. Quantitative analysis of HA revealed that MU significantly inhibited the intracellular and cell surface HA. MU significantly inhibited cell growth and induced apoptosis as determined by cell proliferation and TUNEL assays, respectively. Phosphorylation of Akt was suppressed after 12 and 24 hr treatment with MU. MU treatment also inhibited cell motility as well as cell invasiveness. MU also inhibited cell growth and motility in murine fibroblast cell line NIH3T3. In vivo, administration of MU inhibited the expansion of osteolytic lesions on soft X-rays in mouse breast cancer xenograft models. HA accumulation in bone metastatic lesions was perturbed peripherally. These data suggest that MU might be a therapeutic candidate for bone metastasis of breast cancer via suppression of HA synthesis and accumulation.
Collapse
Affiliation(s)
- Hiroshi Urakawa
- Department of Orthopedic Surgery, Nagoya University Graduate School and School of Medicine, Showa, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The CD44 protein family spans a large group of transmembrane glycoproteins acquired by alternative splicing and post-translational modifications. The great heterogeneity in molecular structure is reflected in its various important functions: CD44 mediates (1) interaction between cell and extracellular matrix, (2) signal submission, e.g., by acting as co-receptor for membrane-spanning receptor tyrosine kinases or by association with intracellular molecules initiating several signaling pathways, and (3) anchor function connecting to the cytoskeleton via the ezrin-radixin-moesin protein family. The expression pattern of the different CD44 isoforms display strong variations dependent on cell type, state of activation, and differentiation stage. In hematopoietic cells, CD44 mediates interaction of progenitor cells and bone marrow stroma during hematopoiesis, regulates maturation, and activation-induced cell death in T cells, influences neutrophil and macrophage migration as well as cytokine production, and participates in lymphocyte extravasation and migration. CD44 is involved in development and progress of hematological neoplasias by enhancement of apoptotic resistance, invasiveness, as well as regulation of bone marrow homing, and mobilization of leukemia-initiating cells into the peripheral blood. Thereby altered CD44 expression functions as marker for worse prognosis in most hematological malignancies. Additionally, CD44 expression levels can be used to distinguish between different hematological neoplasias and subtypes. Concerning new treatment strategies, CD44 displays promising potential either by direct targeting of CD44 expressed on the malignant cells or reversing an acquired resistance to primary treatment mediated through altered CD44 expression. The former can be achieved by antibody or hyaluronan-based immunotherapy.
Collapse
|
49
|
Ween MP, Hummitzsch K, Rodgers RJ, Oehler MK, Ricciardelli C. Versican induces a pro-metastatic ovarian cancer cell behavior which can be inhibited by small hyaluronan oligosaccharides. Clin Exp Metastasis 2010; 28:113-25. [DOI: 10.1007/s10585-010-9363-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 11/11/2010] [Indexed: 12/21/2022]
|
50
|
Cordo-Russo RI, Alaniz LD, Saccodossi N, Lompardía S, Blanco G, Alvarez E, García MG, Hajos SE. Hyaluronan induces migration of multidrug-resistant lymphoma cell lines in vitro through Tiam1 activation by a PI3K-dependent mechanism. Leuk Res 2010; 34:1525-32. [PMID: 20299090 DOI: 10.1016/j.leukres.2010.02.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 02/21/2010] [Accepted: 02/21/2010] [Indexed: 12/21/2022]
Abstract
Hyaluronan (HA) modulates multidrug resistance (MDR) as well as cell migration. Tiam1 is involved in cytoskeleton reorganization during tumor invasion. In this report we show the relationship among HA, Tiam1, migration and MDR in murine lymphoma cell lines. We observed that MDR cells presented higher migratory capacity towards HA in vitro as well as higher constitutive active Tiam1 expression than the sensitive cell line. Besides, HA treatment induced migration towards HA of MDR cell lines through Tiam1 activation by a PI3K-dependent mechanism, showing that disruption of HA signaling would be useful in treatment of MDR hematological malignancies.
Collapse
Affiliation(s)
- Rosalía I Cordo-Russo
- Department of Immunology, School of Pharmacy and Biochemistry, University of Buenos Aires, IDEHU-CONICET, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | |
Collapse
|