1
|
Elvitigala KCML, Mohan L, Mubarok W, Sakai S. Phototuning of Hyaluronic-Acid-Based Hydrogel Properties to Control Network Formation in Human Vascular Endothelial Cells. Adv Healthc Mater 2024; 13:e2303787. [PMID: 38684108 PMCID: PMC11468695 DOI: 10.1002/adhm.202303787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/06/2024] [Indexed: 05/02/2024]
Abstract
In vitro network formation by endothelial cells serves as a fundamental model for studies aimed at understanding angiogenesis. The morphogenesis of these cells to form a network is intricately regulated by the mechanical and biochemical properties of the extracellular matrix. Here the effects of modulating these properties in hydrogels derived from phenolated hyaluronic acid (HA-Ph) and phenolated gelatin (Gelatin-Ph) are presented. Visible-light irradiation in the presence of tris(2,2'-bipyridyl)ruthenium(II) chloride hexahydrate and sodium persulfate induces the crosslinking of these polymers, thereby forming a hydrogel and degrading HA-Ph. Human vascular endothelial cells form networks on the hydrogel prepared by visible-light irradiation for 45 min (42 W cm-2 at 450 nm) but not on the hydrogels prepared by irradiation for 15, 30, or 60 min. The irradiation time-dependent degradation of HA-Ph and the changes in the mechanical stiffness of the hydrogels, coupled with the expressions of RhoA and β-actin genes and CD44 receptors in the cells, reveal that the network formation is synergistically influenced by the hydrogel stiffness and HA-Ph degradation. These findings highlight the potential of tailoring HA-based hydrogel properties to modulate human vascular endothelial cell responses, which is critical for advancing their application in vascular tissue engineering.
Collapse
Affiliation(s)
| | - Lakshmi Mohan
- Department of BioengineeringHenry Samueli School of EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Wildan Mubarok
- Department of Materials Engineering ScienceGraduate School of Engineering ScienceOsaka UniversityToyonakaOsaka560‐8531Japan
| | - Shinji Sakai
- Department of Materials Engineering ScienceGraduate School of Engineering ScienceOsaka UniversityToyonakaOsaka560‐8531Japan
| |
Collapse
|
2
|
Wu S, Tan Y, Li F, Han Y, Zhang S, Lin X. CD44: a cancer stem cell marker and therapeutic target in leukemia treatment. Front Immunol 2024; 15:1354992. [PMID: 38736891 PMCID: PMC11082360 DOI: 10.3389/fimmu.2024.1354992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
CD44 is a ubiquitous leukocyte adhesion molecule involved in cell-cell interaction, cell adhesion, migration, homing and differentiation. CD44 can mediate the interaction between leukemic stem cells and the surrounding extracellular matrix, thereby inducing a cascade of signaling pathways to regulate their various behaviors. In this review, we focus on the impact of CD44s/CD44v as biomarkers in leukemia development and discuss the current research and prospects for CD44-related interventions in clinical application.
Collapse
Affiliation(s)
- Shuang Wu
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yicheng Tan
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
| | - Fanfan Li
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixiang Han
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shenghui Zhang
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaofei Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Han J, Lee C, Jung Y. Current Evidence and Perspectives of Cluster of Differentiation 44 in the Liver's Physiology and Pathology. Int J Mol Sci 2024; 25:4749. [PMID: 38731968 PMCID: PMC11084344 DOI: 10.3390/ijms25094749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Cluster of differentiation 44 (CD44), a multi-functional cell surface receptor, has several variants and is ubiquitously expressed in various cells and tissues. CD44 is well known for its function in cell adhesion and is also involved in diverse cellular responses, such as proliferation, migration, differentiation, and activation. To date, CD44 has been extensively studied in the field of cancer biology and has been proposed as a marker for cancer stem cells. Recently, growing evidence suggests that CD44 is also relevant in non-cancer diseases. In liver disease, it has been shown that CD44 expression is significantly elevated and associated with pathogenesis by impacting cellular responses, such as metabolism, proliferation, differentiation, and activation, in different cells. However, the mechanisms underlying CD44's function in liver diseases other than liver cancer are still poorly understood. Hence, to help to expand our knowledge of the role of CD44 in liver disease and highlight the need for further research, this review provides evidence of CD44's effects on liver physiology and its involvement in the pathogenesis of liver disease, excluding cancer. In addition, we discuss the potential role of CD44 as a key regulator of cell physiology.
Collapse
Affiliation(s)
- Jinsol Han
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea;
| | - Chanbin Lee
- Institute of Systems Biology, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea;
| | - Youngmi Jung
- Department of Integrated Biological Science, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea;
- Department of Biological Sciences, College of Natural Science, Pusan National University, Pusan 46241, Republic of Korea
| |
Collapse
|
4
|
Ziranu P, Pretta A, Aimola V, Cau F, Mariani S, D’Agata AP, Codipietro C, Rizzo D, Dell’Utri V, Sanna G, Moledda G, Cadoni A, Lai E, Puzzoni M, Pusceddu V, Castagnola M, Scartozzi M, Faa G. CD44: A New Prognostic Marker in Colorectal Cancer? Cancers (Basel) 2024; 16:1569. [PMID: 38672650 PMCID: PMC11048923 DOI: 10.3390/cancers16081569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/19/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Cluster of differentiation 44 (CD44) is a non-kinase cell surface glycoprotein. It is overexpressed in several cell types, including cancer stem cells (CSCs). Cells overexpressing CD44 exhibit several CSC traits, such as self-renewal, epithelial-mesenchymal transition (EMT) capability, and resistance to chemo- and radiotherapy. The role of CD44 in maintaining stemness and the CSC function in tumor progression is accomplished by binding to its main ligand, hyaluronan (HA). The HA-CD44 complex activates several signaling pathways that lead to cell proliferation, adhesion, migration, and invasion. The CD44 gene regularly undergoes alternative splicing, resulting in the standard (CD44s) and variant (CD44v) isoforms. The different functional roles of CD44s and specific CD44v isoforms still need to be fully understood. The clinicopathological impact of CD44 and its isoforms in promoting tumorigenesis suggests that CD44 could be a molecular target for cancer therapy. Furthermore, the recent association observed between CD44 and KRAS-dependent carcinomas and the potential correlations between CD44 and tumor mutational burden (TMB) and microsatellite instability (MSI) open new research scenarios for developing new strategies in cancer treatment. This review summarises current research regarding the different CD44 isoform structures, their roles, and functions in supporting tumorigenesis and discusses its therapeutic implications.
Collapse
Affiliation(s)
- Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valentina Aimola
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Flaviana Cau
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Alessandra Pia D’Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Claudia Codipietro
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Daiana Rizzo
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Veronica Dell’Utri
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giorgia Sanna
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giusy Moledda
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Cadoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Massimo Castagnola
- Proteomics Laboratory, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00013 Rome, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Gavino Faa
- Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
5
|
Hu J, Hameed MR, Agaram NP, Whiting KA, Qin LX, Villano AM, O'Connor RB, Rozenberg JM, Cohen S, Prendergast K, Kryeziu S, White RL, Posner MC, Socci ND, Gounder MM, Singer S, Crago AM. PDGFRβ Signaling Cooperates with β-Catenin to Modulate c-Abl and Biologic Behavior of Desmoid-Type Fibromatosis. Clin Cancer Res 2024; 30:450-461. [PMID: 37943631 PMCID: PMC10792363 DOI: 10.1158/1078-0432.ccr-23-2313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/20/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
PURPOSE This study sought to identify β-catenin targets that regulate desmoid oncogenesis and determine whether external signaling pathways, particularly those inhibited by sorafenib (e.g., PDGFRβ), affect these targets to alter natural history or treatment response in patients. EXPERIMENTAL DESIGN In vitro experiments utilized primary desmoid cell lines to examine regulation of β-catenin targets. Relevance of results was assessed in vivo using Alliance trial A091105 correlative biopsies. RESULTS CTNNB1 knockdown inhibited hypoxia-regulated gene expression in vitro and reduced levels of HIF1α protein. ChIP-seq identified ABL1 as a β-catenin transcriptional target that modulated HIF1α and desmoid cell proliferation. Abrogation of either CTNNB1 or HIF1A inhibited desmoid cell-induced VEGFR2 phosphorylation and tube formation in endothelial cell co-cultures. Sorafenib inhibited this activity directly but also reduced HIF1α protein expression and c-Abl activity while inhibiting PDGFRβ signaling in desmoid cells. Conversely, c-Abl activity and desmoid cell proliferation were positively regulated by PDGF-BB. Reduction in PDGFRβ and c-Abl phosphorylation was commonly observed in biopsy samples from patients after treatment with sorafenib; markers of PDGFRβ/c-Abl pathway activation in baseline samples were associated with tumor progression in patients on the placebo arm and response to sorafenib in patients receiving treatment. CONCLUSIONS The β-catenin transcriptional target ABL1 is necessary for proliferation and maintenance of HIF1α in desmoid cells. Regulation of c-Abl activity by PDGF signaling and targeted therapies modulates desmoid cell proliferation, thereby suggesting a reason for variable biologic behavior between tumors, a mechanism for sorafenib activity in desmoids, and markers predictive of outcome in patients.
Collapse
Affiliation(s)
- Jia Hu
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meera R. Hameed
- Bone and Soft Tissue Pathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Narasimhan P. Agaram
- Bone and Soft Tissue Pathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Karissa A. Whiting
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anthony M. Villano
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rachael B. O'Connor
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julian M. Rozenberg
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sonia Cohen
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine Prendergast
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sara Kryeziu
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard L. White
- Department of Surgery, Levine Cancer Center, Atrium Health, Carolinas Medical Center, Charlotte, North Carolina
| | | | - Nicholas D. Socci
- Bioinformatics Core, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mrinal M. Gounder
- Sarcoma Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Samuel Singer
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Surgery, Weill Cornell Medical College, New York, New York
| | - Aimee M. Crago
- Kristen Ann Carr Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Gastric and Mixed Tumor Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Surgery, Weill Cornell Medical College, New York, New York
| |
Collapse
|
6
|
Menezes R, Vincent R, Osorno L, Hu P, Arinzeh TL. Biomaterials and tissue engineering approaches using glycosaminoglycans for tissue repair: Lessons learned from the native extracellular matrix. Acta Biomater 2023; 163:210-227. [PMID: 36182056 PMCID: PMC10043054 DOI: 10.1016/j.actbio.2022.09.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 01/30/2023]
Abstract
Glycosaminoglycans (GAGs) are an important component of the extracellular matrix as they influence cell behavior and have been sought for tissue regeneration, biomaterials, and drug delivery applications. GAGs are known to interact with growth factors and other bioactive molecules and impact tissue mechanics. This review provides an overview of native GAGs, their structure, and properties, specifically their interaction with proteins, their effect on cell behavior, and their mechanical role in the ECM. GAGs' function in the extracellular environment is still being understood however, promising studies have led to the development of medical devices and therapies. Native GAGs, including hyaluronic acid, chondroitin sulfate, and heparin, have been widely explored in tissue engineering and biomaterial approaches for tissue repair or replacement. This review focuses on orthopaedic and wound healing applications. The use of GAGs in these applications have had significant advances leading to clinical use. Promising studies using GAG mimetics and future directions are also discussed. STATEMENT OF SIGNIFICANCE: Glycosaminoglycans (GAGs) are an important component of the native extracellular matrix and have shown promise in medical devices and therapies. This review emphasizes the structure and properties of native GAGs, their role in the ECM providing biochemical and mechanical cues that influence cell behavior, and their use in tissue regeneration and biomaterial approaches for orthopaedic and wound healing applications.
Collapse
Affiliation(s)
- Roseline Menezes
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Richard Vincent
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Laura Osorno
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Phillip Hu
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Treena Livingston Arinzeh
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States; Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States.
| |
Collapse
|
7
|
A R, Kunimura N, Tominaga S, Hirata E, Nishioka S, Uesugi M, Yamazaki R, Ueki H, Kitagawa K, Fujisawa M, Shirakawa T. A recombinant adenovirus vector containing the synNotch receptor gene for the treatment of triple-negative breast cancer. Front Oncol 2023; 13:1147668. [PMID: 37064130 PMCID: PMC10090503 DOI: 10.3389/fonc.2023.1147668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is known as the most difficult molecular subtype of breast cancer to treat. Recent studies revealed that cancer stem cells (CSCs) play a critical role in TNBC recurrence and metastasis. In this study, we developed a recombinant replication-deficient adenoviral vector (Ad-CD44-N-HIF-3α4), which contains a gene encoding a synthetic Notch (synNotch) receptor composed of the extracellular domain of CD44 (CD44-ECD) and the hypoxia-inducible factor (HIF)-3α4 connected by the Notch core regulatory region. CD44 is a transmembrane glycoprotein and known as a CSC marker in breast cancer and other malignancies. HIF-3α4 is a dominant-negative regulator of HIF-1α and HIF-2α and inhibits hypoxia-inducing effect. Both CD44 and HIF signals contribute cancer stemness and maintaining CSCs in breast cancer. The CD44-ECD in the synNotch receptor acts as the CD44 decoy receptor, and after a ligand such as a hyaluronic acid binds to the CD44-ECD, HIF-3α4 is released from the Notch core domain. We performed an in vivo study using a mouse xenograft model of MDA-MB-231, a highly invasive TNBC cell, and confirmed the significant antitumor activity of the intratumoral injections of Ad-CD44-N-HIF3α4. Our findings in this study warrant the further development of Ad-CD44-N-HIF3α4 for the treatment of patients with TNBC.
Collapse
Affiliation(s)
- Ruhan A
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Naoto Kunimura
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Shoko Tominaga
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Erika Hirata
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Shunya Nishioka
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Misato Uesugi
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Rion Yamazaki
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Hideto Ueki
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Kitagawa
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiro Shirakawa
- Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
- *Correspondence: Toshiro Shirakawa,
| |
Collapse
|
8
|
Purushothaman A, Mohajeri M, Lele TP. The role of glycans in the mechanobiology of cancer. J Biol Chem 2023; 299:102935. [PMID: 36693448 PMCID: PMC9930169 DOI: 10.1016/j.jbc.2023.102935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Although cancer is a genetic disease, physical changes such as stiffening of the extracellular matrix also commonly occur in cancer. Cancer cells sense and respond to extracellular matrix stiffening through the process of mechanotransduction. Cancer cell mechanotransduction can enhance cancer-promoting cell behaviors such as survival signaling, proliferation, and migration. Glycans, carbohydrate-based polymers, have recently emerged as important mediators and/or modulators of cancer cell mechanotransduction. Stiffer tumors are characterized by increased glycan content on cancer cells and their associated extracellular matrix. Here we review the role of cancer-associated glycans in coupled mechanical and biochemical alterations during cancer progression. We discuss the recent evidence on how increased expression of different glycans, in the form of glycoproteins and proteoglycans, contributes to both mechanical changes in tumors and corresponding cancer cell responses. We conclude with a summary of emerging tools that can be used to modify glycans for future studies in cancer mechanobiology.
Collapse
Affiliation(s)
- Anurag Purushothaman
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA.
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Tanmay P Lele
- Department of Biomedical Engineering, Texas A&M University, Houston, Texas, USA; Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA; Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA; Department of Translational Medical Sciences, Texas A&M University, Houston, Texas, USA.
| |
Collapse
|
9
|
Carvalho AM, Soares da Costa D, Reis RL, Pashkuleva I. RHAMM expression tunes the response of breast cancer cell lines to hyaluronan. Acta Biomater 2022; 146:187-196. [PMID: 35577044 DOI: 10.1016/j.actbio.2022.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/12/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023]
Abstract
Hyaluronan (HA) synthesis and degradation are altered during carcinogenesis leading to an increased HA content in the tumor microenvironment, which correlates with poor prognosis and treatment outcomes. The main HA receptors, CD44 and RHAMM, are also overexpressed in tumors where they activate anti-apoptotic, proliferative, invasive, and migration signaling pathways. Herein, we used a unidirectional HA gradient to investigate in a high-throughput fashion the bi-directional communication between HA and breast cancer cell lines with different surface expression of CD44 and RHAMM. We found that the expression of CD44 and RHAMM depends on the HA density: the expression of these receptors is promoted at higher HA density and RHAMM is more sensitive to these changes when compared to CD44. Blocking either CD44 or RHAMM revealed different functions on binding and recognizing HA and a compensatory expression between these two receptors that maintains protumorigenic effectors such as cortactin. STATEMENT OF SIGNIFICANCE: We show that the expression of main hyaluronan (HA) receptors CD44 and RHAMM is enhanced in a HA concentration-dependent manner. Blocking activity experiments with either RHAMM or CD44 reveal the redundancy of these two receptors towards HA recognition and activation/recruitment of protumorigenic molecular effector, cortactin. These experiments also demonstrate that cells with overexpressed RHAMM are more sensitive to HA density than CD44 positive cells. The reported results are important for the development of therapies that target the hyaluronan signaling in the tumor microenvironment.
Collapse
Affiliation(s)
- Ana M Carvalho
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal.
| |
Collapse
|
10
|
Kciuk M, Gielecińska A, Mujwar S, Mojzych M, Marciniak B, Drozda R, Kontek R. Targeting carbonic anhydrase IX and XII isoforms with small molecule inhibitors and monoclonal antibodies. J Enzyme Inhib Med Chem 2022; 37:1278-1298. [PMID: 35506234 PMCID: PMC9090362 DOI: 10.1080/14756366.2022.2052868] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carbonic anhydrases IX and CAXII (CAIX/CAXII) are transmembrane zinc metalloproteins that catalyze a very basic but crucial physiological reaction: the conversion of carbon dioxide into bicarbonate with a release of the proton. CA, especially CAIX and CAXII isoforms gained the attention of many researchers interested in anticancer drug design due to pivotal functions of enzymes in the cancer cell metastasis and response to hypoxia, and their expression restricted to malignant cells. This offers an opportunity to develop new targeted therapies with fewer side effects. Continuous efforts led to the discovery of a series of diverse compounds with the most abundant sulphonamide derivatives. Here we review current knowledge considering small molecule and antibody-based targeting of CAIX/CAXII in cancer.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland.,Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Somdutt Mujwar
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, Lodz, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Laboratory of Cytogenetics, University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Oligosaccharides Ameliorate Acute Kidney Injury by Alleviating Cluster of Differentiation 44-Mediated Immune Responses in Renal Tubular Cells. Nutrients 2022; 14:nu14040760. [PMID: 35215410 PMCID: PMC8877265 DOI: 10.3390/nu14040760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 11/29/2022] Open
Abstract
Acute kidney injury (AKI) is a sudden episode of kidney damage that commonly occurs in patients admitted to hospitals. To date, no ideal treatment has been developed to reduce AKI severity. Oligo-fucoidan (FC) interferes with renal tubular cell surface protein cluster of differentiation 44 (CD44) to prevent renal interstitial fibrosis; however, the influence of oligosaccharides on AKI remains unknown. In this study, FC, galacto-oligosaccharide (GOS), and fructo-oligosaccharide (FOS) were selected to investigate the influence of oligosaccharides on AKI. All three oligosaccharides have been proven to be partially absorbed by the intestine. We found that the oligosaccharides dose-dependently reduced CD44 antigenicity and suppressed the hypoxia-induced expression of CD44, phospho-JNK, MCP-1, IL-1β, and TNF-α in NRK-52E renal tubular cells. Meanwhile, CD44 siRNA transfection and JNK inhibitor SP600125 reduced the hypoxia-induced expression of phospho-JNK and cytokines. The ligand of CD44, hyaluronan, counteracted the influence of oligosaccharides on CD44 and phospho-JNK. At 2 days post-surgery for ischemia–reperfusion injury, oligosaccharides reduced kidney inflammation, serum creatine, MCP-1, IL-1β, and TNF-α in AKI mice. At 7 days post-surgery, kidney recovery was promoted. These results indicate that FC, GOS, and FOS inhibit the hypoxia-induced CD44/JNK cascade and cytokines in renal tubular cells, thereby ameliorating AKI and kidney inflammation in AKI mice. Therefore, oligosaccharide supplementation is a potential healthcare strategy for patients with AKI.
Collapse
|
12
|
Identification of Chicken CD44 as a Novel B Lymphocyte Receptor for Infectious Bursal Disease Virus. J Virol 2022; 96:e0011322. [PMID: 35107370 DOI: 10.1128/jvi.00113-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infectious bursal disease virus (IBDV), which targets bursa B lymphocytes, causes severe immunosuppressive disease in chickens, inducing huge economic losses for the poultry industry. To date, the functional receptor for IBDV binding and entry into host cells remains unclear. This study used mass spectrometry to screen host proteins of chicken bursal lymphocytes interacting with VP2. The chicken transmembrane protein cluster of differentiation 44 (chCD44) was identified and evaluated for its interaction with IBDV VP2, the major capsid protein. Overexpression and knockdown experiments showed that chCD44 promotes replication of IBDV. Furthermore, soluble chCD44 and the anti-chCD44 antibody blocked virus binding. The results of receptor reconstitution indicated that chCD44 overexpression conferred viral binding capability in non-permissive cells. More important, although we found that IBDV could not replicate in the chCD44-overexpressed non-permissive cells, the virus could enter non-permissive cells using chCD44. Our finding reveals that chCD44 is a cellular receptor for IBDV, facilitating virus binding and entry in target cells by interacting with the IBDV VP2 protein. IMPORTANCE IBDV causes severe immunosuppressive disease in chickens, inducing huge economic losses for the poultry industry. However, the specific mechanism of IBDV invading host cells of IBDV was not very clear. This study shed light on which cellular protein component IBDV is used to bind and/or enter B lymphocytes. The results of our study revealed that chCD44 could promote both the binding and entry ability of IBDV in B lymphocytes, acting as a cellular receptor for IBDV. Besides, this is the first report about chicken CD44 function in viral replication. Our study impacts the understanding of the IBDV binding and entry process and sets the stage for further elucidation of the infection mechanism of IBDV.
Collapse
|
13
|
Chung H, Komada T, Lau A, Chappellaz M, Platnich JM, de Koning HD, Petri B, Luque Y, Walker S, Benediktsson H, Mesnard L, Chun J, Muruve DA. AIM2 Suppresses Inflammation and Epithelial Cell Proliferation during Glomerulonephritis. THE JOURNAL OF IMMUNOLOGY 2021; 207:2799-2812. [PMID: 34740957 DOI: 10.4049/jimmunol.2100483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/28/2021] [Indexed: 11/19/2022]
Abstract
Absent in melanoma-2 (AIM2) is an inflammasome-forming innate immune sensor for dsDNA but also exhibits inflammasome-independent functions such as restricting cellular proliferation. AIM2 is expressed in the kidney, but its localization and function are not fully characterized. In normal human glomeruli, AIM2 localized to podocytes. In patients with glomerulonephritis, AIM2 expression increased in CD44+-activated parietal epithelial cells within glomerular crescents. To explore AIM2 effects in glomerular disease, studies in Aim2 -/- mice were performed. Aim2-/- glomeruli showed reduced expression of Wilm tumor gene-1 (WT1), WT1-driven podocyte genes, and increased proliferation in outgrowth assays. In a nephrotoxic serum (NTS)-induced glomerulonephritis model, Aim2-/- (B6) mice exhibited more severe glomerular crescent formation, tubular injury, inflammation, and proteinuria compared with wild-type controls. Inflammasome activation markers were absent in both Aim2 -/- and wild-type kidneys, despite an increased inflammatory transcriptomic signature in Aim2 -/- mice. Aim2 -/- mice also demonstrated dysregulated cellular proliferation and an increase in CD44+ parietal epithelial cells during glomerulonephritis. The augmented inflammation and epithelial cell proliferation in Aim2 -/- (B6) mice was not due to genetic background, as Aim2 -/- (B6.129) mice demonstrated a similar phenotype during NTS glomerulonephritis. The AIM2-like receptor (ALR) locus was necessary for the inflammatory glomerulonephritis phenotype observed in Aim2 -/- mice, as NTS-treated ALR -/- mice displayed equal levels of injury as wild-type controls. Podocyte outgrowth from ALR -/- glomeruli was still increased, however, confirming that the ALR locus is dispensable for AIM2 effects on epithelial cell proliferation. These results identify a noncanonical role for AIM2 in suppressing inflammation and epithelial cell proliferation during glomerulonephritis.
Collapse
Affiliation(s)
- Hyunjae Chung
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Takanori Komada
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Lau
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mona Chappellaz
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jaye M Platnich
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Heleen D de Koning
- Department of Dermatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Björn Petri
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yosu Luque
- Soins Intensifs Néphrologiques et Rein Aigu (SINRA), Département de Néphrologie, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Inserm UMR_S1155, Sorbonne Université, Paris, France; and
| | - Simon Walker
- Department of Pathology and Laboratory Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hallgrimur Benediktsson
- Department of Pathology and Laboratory Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Laurent Mesnard
- Soins Intensifs Néphrologiques et Rein Aigu (SINRA), Département de Néphrologie, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Inserm UMR_S1155, Sorbonne Université, Paris, France; and
| | - Justin Chun
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniel A Muruve
- Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada;
| |
Collapse
|
14
|
Guillermin O, Angelis N, Sidor CM, Ridgway R, Baulies A, Kucharska A, Antas P, Rose MR, Cordero J, Sansom O, Li VSW, Thompson BJ. Wnt and Src signals converge on YAP-TEAD to drive intestinal regeneration. EMBO J 2021; 40:e105770. [PMID: 33950519 PMCID: PMC8246259 DOI: 10.15252/embj.2020105770] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Wnt signalling induces a gradient of stem/progenitor cell proliferation along the crypt-villus axis of the intestine, which becomes expanded during intestinal regeneration or tumour formation. The YAP transcriptional co-activator is known to be required for intestinal regeneration, but its mode of regulation remains controversial. Here we show that the YAP-TEAD transcription factor is a key downstream effector of Wnt signalling in the intestine. Loss of YAP activity by Yap/Taz conditional knockout results in sensitivity of crypt stem cells to apoptosis and reduced cell proliferation during regeneration. Gain of YAP activity by Lats1/2 conditional knockout is sufficient to drive a crypt hyperproliferation response. In particular, Wnt signalling acts transcriptionally to induce YAP and TEAD1/2/4 expression. YAP normally localises to the nucleus only in crypt base stem cells, but becomes nuclear in most intestinal epithelial cells during intestinal regeneration after irradiation, or during organoid growth, in a Src family kinase-dependent manner. YAP-driven crypt expansion during regeneration involves an elongation and flattening of the Wnt signalling gradient. Thus, Wnt and Src-YAP signals cooperate to drive intestinal regeneration.
Collapse
Affiliation(s)
- Oriane Guillermin
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Nikolaos Angelis
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Clara M Sidor
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Rachel Ridgway
- Colorectal Cancer and Wnt signalling LaboratoryCancer Research UK Beatson InstituteGlasgowUK
| | - Anna Baulies
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Anna Kucharska
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Pedro Antas
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Melissa R Rose
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Julia Cordero
- Institute of Cancer SciencesWolfson Wohl Cancer Research CentreBearsdenUK
| | - Owen Sansom
- Colorectal Cancer and Wnt signalling LaboratoryCancer Research UK Beatson InstituteGlasgowUK
| | - Vivian S W Li
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Barry J Thompson
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
- EMBL Australia ACRF Department of Cancer Biology & TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityActonACTAustralia
| |
Collapse
|
15
|
Nishikawa M, Inoue A, Ohnishi T, Yano H, Ozaki S, Kanemura Y, Suehiro S, Ohtsuka Y, Kohno S, Ohue S, Shigekawa S, Watanabe H, Kitazawa R, Tanaka J, Kunieda T. Hypoxia-induced phenotypic transition from highly invasive to less invasive tumors in glioma stem-like cells: Significance of CD44 and osteopontin as therapeutic targets in glioblastoma. Transl Oncol 2021; 14:101137. [PMID: 34052625 PMCID: PMC8175402 DOI: 10.1016/j.tranon.2021.101137] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/10/2021] [Accepted: 05/21/2021] [Indexed: 01/13/2023] Open
Abstract
CD44, upregulated by HIF-1α under 1%O2, induces highly invasive phenotype GSCs. HIF-2α-activated OPN under 5%O2 promotes less-invasive/proliferative type GSCs. CD44 and OPN knockdowns inhibit in vitro/vivo GSCs invasion and proliferation.
The poor prognosis of glioblastoma multiforme (GBM) is primarily due to highly invasive glioma stem-like cells (GSCs) in tumors. Upon GBM recurrence, GSCs with highly invasive and highly migratory activities must assume a less-motile state and proliferate to regenerate tumor mass. Elucidating the molecular mechanism underlying this transition from a highly invasive phenotype to a less-invasive, proliferative tumor could facilitate the identification of effective molecular targets for treating GBM. Here, we demonstrate that severe hypoxia (1% O2) upregulates CD44 expression via activation of hypoxia-inducible factor (HIF-1α), inducing GSCs to assume a highly invasive tumor. In contrast, moderate hypoxia (5% O2) upregulates osteopontin expression via activation of HIF-2α. The upregulated osteopontin inhibits CD44-promoted GSC migration and invasion and stimulates GSC proliferation, inducing GSCs to assume a less-invasive, highly proliferative tumor. These data indicate that the GSC phenotype is determined by interaction between CD44 and osteopontin. The expression of both CD44 and osteopontin is regulated by differential hypoxia levels. We found that CD44 knockdown significantly inhibited GSC migration and invasion both in vitro and in vivo. Mouse brain tumors generated from CD44-knockdown GSCs exhibited diminished invasiveness, and the mice survived significantly longer than control mice. In contrast, siRNA-mediated silencing of the osteopontin gene decreased GSC proliferation. These results suggest that interaction between CD44 and osteopontin plays a key role in tumor progression in GBM; inhibition of both CD44 and osteopontin may represent an effective therapeutic approach for suppressing tumor progression, thus resulting in a better prognosis for patients with GBM.
Collapse
Affiliation(s)
- Masahiro Nishikawa
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Akihiro Inoue
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan.
| | - Takanori Ohnishi
- Department of Neurosurgery, Washokai Sadamoto Hospital, Matsuyama, Ehime 790-0052, Japan
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Saya Ozaki
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka 540-0006, Japan; Department of Neurosurgery, National Hospital Organization Osaka National Hospital, Osaka 540-0006, Japan
| | - Satoshi Suehiro
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Yoshihiro Ohtsuka
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Shohei Kohno
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Shiro Ohue
- Department of Neurosurgery, Ehime Prefectural Central Hospital, Matsuyama, Ehime 790-0024, Japan
| | - Seiji Shigekawa
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Hideaki Watanabe
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Riko Kitazawa
- Division of Diagnostic Pathology, Ehime University Hospital, Toon, Ehime 791-0295, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Takeharu Kunieda
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| |
Collapse
|
16
|
Li HL, Li QY, Jin MJ, Lu CF, Mu ZY, Xu WY, Song J, Zhang Y, Zhang SY. A review: hippo signaling pathway promotes tumor invasion and metastasis by regulating target gene expression. J Cancer Res Clin Oncol 2021; 147:1569-1585. [PMID: 33864521 DOI: 10.1007/s00432-021-03604-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/16/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The Hippo pathway is widely considered to inhibit cell growth and play an important role in regulating the size of organs. However, recent studies have shown that abnormal regulation of the Hippo pathway can also affect tumor invasion and metastasis. Therefore, finding out how the Hippo pathway promotes tumor development by regulating the expression of target genes provides new ideas for future research on targeted drugs that inhibit tumor progression. METHODS PubMed, Embase, Web of Science, and the Cochrane Library were systematically searched. RESULTS The search strategy identified 1892 hits and 196 publications were finally included in this review. As the core molecule of the Hippo pathway, YAP/TAZ are usually highly expressed in tumors that undergo invasion and migration and are accompanied by abnormally strong nuclear metastasis. Through its interaction with nuclear transcription factors TEADs, it directly or indirectly regulates and the expressions of target genes related to tumor metastasis and invasion. These target genes can induce the formation of invasive pseudopodia in tumor cells, reduce intercellular adhesion, degrade extracellular matrix (ECM), and cause epithelial-mesenchymal transition (EMT), or indirectly promote through other signaling pathways, such as mitogen-activated protein kinases (MAPK), TGF/Smad, etc, which facilitate the invasion and metastasis of tumors. CONCLUSION This article mainly introduces the research progress of YAP/TAZ which are the core molecules of the Hippo pathway regulating related target genes to promote tumor invasion and metastasis. Focus on the target genes that affect tumor invasion and metastasis, providing the possibility for the selection of clinical drug treatment targets, to provide some help for a more in-depth study of tumor invasion and migration mechanism and the development of clinical drugs.
Collapse
Affiliation(s)
- Hong-Li Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Qian-Yu Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Min-Jie Jin
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chao-Fan Lu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhao-Yang Mu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wei-Yi Xu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China. .,School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Institute of Drug Discovery and Development, Zhengzhou, 450001, China.
| | - Yan Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China. .,School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Institute of Drug Discovery and Development, Zhengzhou, 450001, China. .,Zhengzhou University, Henan Institute of Advanced Technology, Zhengzhou, 450001, China.
| |
Collapse
|
17
|
Georgescu MM, Islam MZ, Li Y, Traylor J, Nanda A. Novel targetable FGFR2 and FGFR3 alterations in glioblastoma associate with aggressive phenotype and distinct gene expression programs. Acta Neuropathol Commun 2021; 9:69. [PMID: 33853673 PMCID: PMC8048363 DOI: 10.1186/s40478-021-01170-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023] Open
Abstract
Prognostic molecular subgrouping of glioblastoma is an ongoing effort and the current classification includes IDH-wild-type and IDH-mutant entities, the latter showing significantly better prognosis. We performed a comparative integrated analysis of the FGFR glioblastoma subgroup consisting of 5 cases from a prospective 101-patient-cohort. FGFR alterations included FGFR2-TACC2 and FGFR2 amplifications arising in a multifocal IDH-mutant glioblastoma with unexpected 2.5-month patient survival, novel FGFR3 carboxy-terminal duplication and FGFR3-TLN1 fusion, and two previously described FGFR3-TACC3 fusions. The FGFR2 tumors showed additional mutations in SERPINE1/PAI-1 and MMP16, as part of extensive extracellular matrix remodeling programs. Whole transcriptomic analysis revealed common proliferation but distinct morphogenetic gene expression programs that correlated with tumor histology. The kinase program revealed EPHA3, LTK and ALK receptor tyrosine kinase overexpression in individual FGFR tumors. Paradoxically, all FGFR-fused glioblastomas shared strong PI3K and MAPK pathway suppression effected by SPRY, DUSP and AKAP12 inhibitors, whereas the FGFR2-TACC2 tumor elicited also EGFR suppression by ERRFI1 upregulation. This integrated analysis outlined the proliferation and morphogenetic expression programs in FGFR glioblastoma, and identified four novel, clinically targetable FGFR2 and FGFR3 alterations that confer aggressive phenotype and trigger canonical pathway feedback inhibition, with important therapeutic implications.
Collapse
|
18
|
Hassan N, Greve B, Espinoza-Sánchez NA, Götte M. Cell-surface heparan sulfate proteoglycans as multifunctional integrators of signaling in cancer. Cell Signal 2020; 77:109822. [PMID: 33152440 DOI: 10.1016/j.cellsig.2020.109822] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
Proteoglycans (PGs) represent a large proportion of the components that constitute the extracellular matrix (ECM). They are a diverse group of glycoproteins characterized by a covalent link to a specific glycosaminoglycan type. As part of the ECM, heparan sulfate (HS)PGs participate in both physiological and pathological processes including cell recruitment during inflammation and the promotion of cell proliferation, adhesion and motility during development, angiogenesis, wound repair and tumor progression. A key function of HSPGs is their ability to modulate the expression and function of cytokines, chemokines, growth factors, morphogens, and adhesion molecules. This is due to their capacity to act as ligands or co-receptors for various signal-transducing receptors, affecting pathways such as FGF, VEGF, chemokines, integrins, Wnt, notch, IL-6/JAK-STAT3, and NF-κB. The activation of those pathways has been implicated in the induction, progression, and malignancy of a tumor. For many years, the study of signaling has allowed for designing specific drugs targeting these pathways for cancer treatment, with very positive results. Likewise, HSPGs have become the subject of cancer research and are increasingly recognized as important therapeutic targets. Although they have been studied in a variety of preclinical and experimental models, their mechanism of action in malignancy still needs to be more clearly defined. In this review, we discuss the role of cell-surface HSPGs as pleiotropic modulators of signaling in cancer and identify them as promising markers and targets for cancer treatment.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Biotechnology Program, Department of Chemistry, Faculty of Science, Cairo University, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany.
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.
| |
Collapse
|
19
|
Three-Dimensional Culture System of Cancer Cells Combined with Biomaterials for Drug Screening. Cancers (Basel) 2020; 12:cancers12102754. [PMID: 32987868 PMCID: PMC7601447 DOI: 10.3390/cancers12102754] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary For the research and development of drug discovery, it is of prime importance to construct the three-dimensional (3D) tissue models in vitro. To this end, the enhancement design of cell function and activity by making use of biomaterials is essential. In this review, 3D culture systems of cancer cells combined with several biomaterials for anticancer drug screening are introduced. Abstract Anticancer drug screening is one of the most important research and development processes to develop new drugs for cancer treatment. However, there is a problem resulting in gaps between the in vitro drug screening and preclinical or clinical study. This is mainly because the condition of cancer cell culture is quite different from that in vivo. As a trial to mimic the in vivo cancer environment, there has been some research on a three-dimensional (3D) culture system by making use of biomaterials. The 3D culture technologies enable us to give cancer cells an in vitro environment close to the in vivo condition. Cancer cells modified to replicate the in vivo cancer environment will promote the biological research or drug discovery of cancers. This review introduces the in vitro research of 3D cell culture systems with biomaterials in addition to a brief summary of the cancer environment.
Collapse
|
20
|
Luo Q, Wu T, Wu W, Chen G, Luo X, Jiang L, Tao H, Rong M, Kang S, Deng M. The Functional Role of Voltage-Gated Sodium Channel Nav1.5 in Metastatic Breast Cancer. Front Pharmacol 2020; 11:1111. [PMID: 32792949 PMCID: PMC7393602 DOI: 10.3389/fphar.2020.01111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Voltage-gated sodium channels (VGSCs), which are abnormally expressed in various types of cancers such as breast cancer, prostate cancer, lung cancer, and cervical cancer, are involved in the metastatic process of invasion and migration. Nav1.5 is a pore-forming α subunit of VGSC encoded by SCN5A. Various studies have demonstrated that Nav1.5, often as its neonatal splice form, is highly expressed in metastatic breast cancer cells. Abnormal activation and expression of Nav1.5 trigger a variety of cellular mechanisms, including changing H+ efflux, promoting epithelial-to-mesenchymal transition (EMT) and the expression of cysteine cathepsin, to potentiate the metastasis and invasiveness of breast cancer cells in vitro and in vivo. Here, we systematically review the latest available data on the pro-metastatic effect of Nav1.5 and its underlying mechanisms in breast cancer. We summarize the factors affecting Nav1.5 expression in breast cancer cells, and discuss the potential of Nav1.5 blockers serving as candidates for breast cancer treatment.
Collapse
Affiliation(s)
- Qianxuan Luo
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Ting Wu
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenfang Wu
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Gong Chen
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Xuan Luo
- Department of Biochemistry and Molecular Biology, Hunan Normal University, Changsha, China
| | - Liping Jiang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Huai Tao
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, China
| | - Mingqiang Rong
- Department of Biochemistry and Molecular Biology, Hunan Normal University, Changsha, China
| | - Shuntong Kang
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Meichun Deng
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
21
|
Liu A, Ildefonso CJ, Bond WS, Hurwitz MY, Hurwitz RL. Inhibitors of metalloprotease, γ-sectretase, protein kinase C and Rho kinase inhibit wild-type adenoviral replication. PLoS One 2020; 15:e0236175. [PMID: 32697798 PMCID: PMC7375579 DOI: 10.1371/journal.pone.0236175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/30/2020] [Indexed: 12/03/2022] Open
Abstract
Adenoviruses cause upper respiratory infections, conjunctivitis, keratitis, and gastrointestinal illness. These can be fatal in immunocompromised individuals. Adenoviruses have also been engineered into viral vectors to deliver therapeutic genes or induce immunity as vaccine carriers. The success of ocular gene therapy is driven partly by the immunologic and biochemical influences of the intraocular environment. We have shown that versican and hyaluronan modulate adenoviral vector transgene expression through CD44 signaling. Herein we explored the role of these pathways on virus replication and viral protein expression of wild type adenovirus. We report that the addition of vitreous humor (which contains both versican and hyaluronan) increases viral hexon protein levels. Vitreous humor also increased wild type adenovirus DNA replication in vitro. Metalloproteinase and γ-secretase inhibitors, which inhibit CD44 proteolytic activation, blocked adenoviral replication in vitro. Similarly, protein kinase C and RhoA kinase inhibitors, both proteins associated with CD44 mediated pathways, also inhibited wild type adenoviral replication in vitro. Application of metalloproteinase and γ-secretase inhibitors to human conjunctival explants sharply decreased adenoviral vector gene expression. Our results demonstrate that pharmacologic delivery of these inhibitors is easily achievable. The inhibition of these enzymes should be explored as potential therapies of wild type adenoviral infections.
Collapse
Affiliation(s)
- Alice Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Summer Undergraduate Research Training Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Cristhian J. Ildefonso
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wesley S. Bond
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mary Y. Hurwitz
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard L. Hurwitz
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Summer Undergraduate Research Training Program, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
22
|
Georgescu MM, Islam MZ, Li Y, Circu ML, Traylor J, Notarianni CM, Kline CN, Burns DK. Global activation of oncogenic pathways underlies therapy resistance in diffuse midline glioma. Acta Neuropathol Commun 2020; 8:111. [PMID: 32680567 PMCID: PMC7367358 DOI: 10.1186/s40478-020-00992-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022] Open
Abstract
Diffuse midline gliomas (DMGs) are aggressive pediatric brain tumors with dismal prognosis due to therapy-resistant tumor growth and invasion. We performed the first integrated histologic/genomic/proteomic analysis of 21 foci from three pontine DMG cases with supratentorial dissemination. Histone H3.3-K27M was the driver mutation, usually at high variant allele fraction due to recurrent chromosome 1q copy number gain, in combination with germline variants in ATM, FANCM and MYCN genes. Both previously reported and novel recurrent copy number variations and somatic pathogenic mutations in chromatin remodeling, DNA damage response and PI3K/MAPK growth pathways were variably detected, either in multiple or isolated foci. Proteomic analysis showed global upregulation of histone H3, lack of H3-K27 trimethylation, and further impairment of polycomb repressive complex 2 by ASXL1 downregulation. Activation of oncogenic pathways resulted from combined upregulation of N-MYC, SOX2, p65/p50 NF-κB and STAT3 transcription factors, EGFR, FGFR2, PDGFRα/β receptor tyrosine kinases, and downregulation of PHLPP1/2, PTEN and p16/INK4A tumor suppressors. Upregulation of SMAD4, PAI-1, CD44, and c-SRC in multiple foci most likely contributed to invasiveness. This integrated comprehensive analysis revealed a complex spatiotemporal evolution in diffuse intrisic pontine glioma, recommending pontine and cerebellar biopsies for accurate populational genetic characterization, and delineated common signaling pathways and potential therapeutic targets. It also revealed an unsuspected activation of a multitude of oncogenic pathways, including cancer cell reprogramming, explaining the resistance of DMG to current therapies.
Collapse
|
23
|
Wang C, Wang Z, Chen C, Fu X, Wang J, Fei X, Yan X, Xu R. A low MW inhibitor of CD44 dimerization for the treatment of glioblastoma. Br J Pharmacol 2020; 177:3009-3023. [PMID: 32080830 PMCID: PMC7280016 DOI: 10.1111/bph.15030] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE As a hallmark of glioblastoma multiforme (GBM), CD44 plays a crucial role in promoting glioblastoma stem cell (GSC) stemness phenotypes and multiple drug resistance. The therapeutic potential of CD44 has been validated by the clinical successes of several CD44 inhibitors, including antibodies and hyaluronan-related drugs. EXPERIMENTAL APPROACH We used systemsDock software to predict verbascoside as a candidate CD44 inhibitor. Microscale thermophoresis was used to confirm the interaction between CD44 and verbascoside. Four glioblastoma cell lines and a patient-derived glioblastoma cell line were used to test the influences of verbascoside on glioblastoma. CD44-overexpressing and CD44-knockout cell lines were also used. Real-time quantitative PCR and western blot analyses were performed. A xenograft mouse model was used to test verbascoside. KEY RESULTS Verbascoside bound to CD44 and suppressed its dimerization. By inhibiting CD44 dimerization, verbascoside decreased the release of the CD44 intracellular domain (CD44ICD) and suppressed the expression of CD44 downstream genes. Verbascoside treatment suppressed the stemness phenotypes of cells with high CD44 expression. In a mouse model of glioma, verbascoside treatment highly reduced the growth of intracranial tumours and inhibited CD44ICD release. Both stem cell marker and mesenchymal GBM subtype marker genes were down-regulated in verbascoside-treated mice. CONCLUSION AND IMPLICATIONS Verbascoside suppressed growth of glioblastoma cells by inhibiting CD44 dimerization. Stem cell-like cell properties and tumour cell growth were also suppressed by verbascoside, both in vitro and in vivo. Verbascoside significantly prolonged survival of xenografted mice.
Collapse
Affiliation(s)
- Chongwu Wang
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
| | - Zhaotao Wang
- Department of NeurosurgeryThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Chen Chen
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
| | - Xiaojun Fu
- Chinese PLA General HospitalMedicine School of Chinese PLABeijingChina
| | - Ji Wang
- School of basic medical scienceSouthern Medicine UniversityGuangzhouChina
| | - Xiaowei Fei
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
| | - Xiaojing Yan
- State Key Laboratory of Tree Genetics and BreedingChinese Academy of ForestryBeijingChina
| | - Ruxiang Xu
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
- School of basic medical scienceSouthern Medicine UniversityGuangzhouChina
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
24
|
Zhou Y, Feng Z, Cao F, Liu X, Xia X, Yu CH. Abl-mediated PI3K activation regulates macrophage podosome formation. J Cell Sci 2020; 133:jcs234385. [PMID: 32393599 DOI: 10.1242/jcs.234385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
Podosomes play crucial roles in macrophage adhesion and migration. Wiskott-Aldrich syndrome protein (WASP; also known as WAS)-mediated actin polymerization is one of the key events initiating podosome formation. Nevertheless, membrane signals to trigger WASP activation at macrophage podosomes remain unclear. Here, we show that phosphatidylinositol (3,4,5)-trisphosphate [PI(3,4,5)P3] lipids are enriched at the podosome and stably recruit WASP rather than the WASP-5KE mutant. Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit β (PIK3CB) is spatially located at the podosome core. Inhibition of PIK3CB and overexpression of phosphatase and tensin homolog (PTEN) impede F-actin polymerization of the podosome. PIK3CB activation is regulated by Abl1 and Src family kinases. At the podosome core, Src and Hck promote the phosphorylation of Tyr488 in the consensus Y-x-x-M motif of Abl1, which enables the association of phosphoinositide 3-kinase (PI3K) regulatory subunits. Knockdown of Abl1 rather than Abl2 suppresses the PI3K/Akt pathway, regardless of Src and Hck activities. Reintroduction of wild-type Abl1 rather than the Abl1-Y488F mutant rescues PI3KR1 recruitment and PI3K activation. When PIK3CB, Abl1 or Src/Hck is suppressed, macrophage podosome formation, matrix degradation and chemotactic migration are inhibited. Thus, Src/Hck-mediated phosphorylation of Abl1 Tyr488 triggers PIK3CB-dependent PI(3,4,5)P3 production and orchestrates the assembly and function of macrophage podosomes.
Collapse
Affiliation(s)
- Yuhuan Zhou
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Zhen Feng
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Fakun Cao
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Xiaoting Liu
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Xiaojie Xia
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Cheng-Han Yu
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
25
|
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
|
26
|
Lee MN, Song JH, Oh SH, Tham NT, Kim JW, Yang JW, Kim ES, Koh JT. The primary cilium directs osteopontin-induced migration of mesenchymal stem cells by regulating CD44 signaling and Cdc42 activation. Stem Cell Res 2020; 45:101799. [PMID: 32339903 DOI: 10.1016/j.scr.2020.101799] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/28/2020] [Accepted: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
The primary cilium acts as a sensory organelle with diverse receptors and ion channels to detect extracellular cues and regulate cellular functions, including cell migration. The migration of mesenchymal stem cells (MSCs) to bone remodeling sites is important for bone homeostasis. Recently, we have suggested that osteopontin (OPN) is a significant chemoattractant in MSC migration to bone remodeling sites. The objective of this study was to determine whether the primary cilium acts as a chemoattractant sensory unit to detect OPN cues and control MSC migration. We found that the loss of primary cilium induced by silencing of IFT88 reduced OPN-induced migration of MSCs. The effect of IFT88 silencing on cellular attachment, spreading, and proliferation was negligible. The loss of primary cilium did not affect the level of integrinβ1 or CD44, two known receptors for OPN. Interestingly, CD44 was localized to the primary cilium by OPN stimulus. Knockdown of IFT88 or CD44 dysregulated OPN-induced signaling activation and abolished OPN-induced Cdc42 activation. Our findings suggest that the primary cilium acts as a chemoattractant sensor for OPN to regulate MSC migration by controlling not only CD44-mediated OPN signaling, but also Cdc42-mediated actin cytoskeleton rearrangement.
Collapse
Affiliation(s)
- Mi Nam Lee
- Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Ju Han Song
- Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Sin-Hye Oh
- Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Nguyen Thi Tham
- Department of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, Korea
| | - Jung-Woo Kim
- Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Jin-Woo Yang
- Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Eung-Sam Kim
- Department of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, Korea
| | - Jeong-Tae Koh
- Hard-tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea.
| |
Collapse
|
27
|
Yang M, James AD, Suman R, Kasprowicz R, Nelson M, O'Toole PJ, Brackenbury WJ. Voltage-dependent activation of Rac1 by Na v 1.5 channels promotes cell migration. J Cell Physiol 2020; 235:3950-3972. [PMID: 31612502 PMCID: PMC6973152 DOI: 10.1002/jcp.29290] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022]
Abstract
Ion channels can regulate the plasma membrane potential (Vm ) and cell migration as a result of altered ion flux. However, the mechanism by which Vm regulates motility remains unclear. Here, we show that the Nav 1.5 sodium channel carries persistent inward Na+ current which depolarizes the resting Vm at the timescale of minutes. This Nav 1.5-dependent Vm depolarization increases Rac1 colocalization with phosphatidylserine, to which it is anchored at the leading edge of migrating cells, promoting Rac1 activation. A genetically encoded FRET biosensor of Rac1 activation shows that depolarization-induced Rac1 activation results in acquisition of a motile phenotype. By identifying Nav 1.5-mediated Vm depolarization as a regulator of Rac1 activation, we link ionic and electrical signaling at the plasma membrane to small GTPase-dependent cytoskeletal reorganization and cellular migration. We uncover a novel and unexpected mechanism for Rac1 activation, which fine tunes cell migration in response to ionic and/or electric field changes in the local microenvironment.
Collapse
Affiliation(s)
- Ming Yang
- Department of BiologyUniversity of YorkYorkUK
| | - Andrew D. James
- Department of BiologyUniversity of YorkYorkUK
- York Biomedical Research InstituteUniversity of YorkYorkUK
| | - Rakesh Suman
- Phase Focus Ltd, Electric WorksSheffield Digital CampusSheffieldUK
| | | | - Michaela Nelson
- Department of BiologyUniversity of YorkYorkUK
- York Biomedical Research InstituteUniversity of YorkYorkUK
| | - Peter J. O'Toole
- Bioscience Technology Facility, Department of BiologyUniversity of YorkYorkUK
| | - William J. Brackenbury
- Department of BiologyUniversity of YorkYorkUK
- York Biomedical Research InstituteUniversity of YorkYorkUK
| |
Collapse
|
28
|
Guo JY, Chiu CH, Wang MJ, Li FA, Chen JY. Proteoglycan serglycin promotes non-small cell lung cancer cell migration through the interaction of its glycosaminoglycans with CD44. J Biomed Sci 2020; 27:2. [PMID: 31898491 PMCID: PMC6939340 DOI: 10.1186/s12929-019-0600-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background Serglycin (SRGN), previously recognized as an intracellular proteoglycan involved in the storage processes of secretory granules, has recently been shown to be upregulated in several solid tumors. We have previously shown that SRGN in non-small cell lung cancer (NSCLC) promotes malignant phenotypes in a CD44-dependent manner and increased expression of SRGN predicts poor prognosis of primary lung adenocarcinomas. However, the underlying mechanism remains to be defined. Methods Overexpression, knockdown and knockout approaches were performed to assess the role of SRGN in cell motility using wound healing and Boyden chamber migration assays. SRGN devoid of glycosaminoglycan (GAG) modification was produced by site-directed mutagenesis or chondroitinase treatment. Liquid chromatography/tandem mass spectrometry was applied for quantitative analysis of the disaccharide compositions and sulfation extent of SRGN GAGs. Western blot and co-immunoprecipitation analyses were performed to determine the expression and interaction of proteins of interest. Actin cytoskeleton organization was monitored by immunofluorescence staining. Results SRGN expressed by NSCLC cells is readily secreted to the extracellular matrix in a heavily glycosylated form attached with mainly chondroitin sulfate (CS)-GAG chains, and to a lesser extent with heparin sulfate (HS). The CS-GAG moiety serves as the structural motif for SRGN binding to tumor cell surface CD44 and promotes cell migration. SRGN devoid of CS-GAG modification fails to interact with CD44 and has lost the ability to promote cell migration. SRGN/CD44 interaction promotes focal adhesion turnover via Src-mediated paxillin phosphorylation and disassembly of paxillin/FAK adhesion complex, facilitating cell migration. In support, depletion of Src activity or removal of CS-GAGs efficiently blocks SRGN-mediated Src activation and cell migration. SRGN also promotes cell migration via inducing cytoskeleton reorganization mediated through RAC1 and CDC42 activation accompanied with increased lamellipodia and filopodia formation. Conclusions Proteoglycan SRGN promotes NSCLC cell migration via the binding of its GAG motif to CD44. SRGN/CD44 interaction induces Rho-family GTPase-mediated cytoskeleton reorganization and facilitates Src-mediated focal adhesion turnover, leading to increased cell migration. These findings suggest that targeting specific glycans in tumor microenvironment that serve as ligands for oncogenic pathways may be a potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Jing-You Guo
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, 115, Taiwan
| | - Chu-Hsuan Chiu
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, 115, Taiwan
| | - Mei-Jung Wang
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, 115, Taiwan
| | - Fu-An Li
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, 115, Taiwan
| | - Jeou-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Academia Road, Section 2, Taipei, 115, Taiwan. .,Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, Republic of China.
| |
Collapse
|
29
|
Xiao W, Wang S, Zhang R, Sohrabi A, Yu Q, Liu S, Ehsanipour A, Liang J, Bierman RD, Nathanson DA, Seidlits SK. Bioengineered scaffolds for 3D culture demonstrate extracellular matrix-mediated mechanisms of chemotherapy resistance in glioblastoma. Matrix Biol 2020; 85-86:128-146. [PMID: 31028838 PMCID: PMC6813884 DOI: 10.1016/j.matbio.2019.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
Abstract
Originating in the brain, glioblastoma (GBM) is a highly lethal and virtually incurable cancer, in large part because it readily develops resistance to treatments. While numerous studies have investigated mechanisms enabling GBM cells to evade chemotherapy-induced apoptosis, few have addressed how their surrounding extracellular matrix (ECM) acts to promote their survival. Here, we employed a biomaterial-based, 3D culture platform to investigate systematically how interactions between patient-derived GBM cells and the brain ECM promote resistance to alkylating chemotherapies - including temozolomide, which is used routinely in clinical practice. Scaffolds for 3D culture were fabricated from hyaluronic acid (HA) - a major structural and bioactive component of the brain ECM - and functionalized with the RGD (arginine-glycine-aspartic acid) tripeptide to provide sites for integrin engagement. Data demonstrate that cooperative engagement of CD44, through HA, and integrin αV, through RGD, facilitates resistance to alkylating chemotherapies through co-activation of Src, which inhibited downstream expression of BCL-2 family pro-apoptotic factors. In sum, a bioengineered, 3D culture platform was used to gain new mechanistic insights into how ECM in the brain tumor microenvironment promotes resistance to chemotherapy and suggests potential avenues for the development of novel, matrix-targeted combination therapies designed to suppress chemotherapy resistance in GBM.
Collapse
Affiliation(s)
- Weikun Xiao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Shanshan Wang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rongyu Zhang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Alireza Sohrabi
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Qi Yu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sihan Liu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Arshia Ehsanipour
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jesse Liang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rebecca D Bierman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - David A Nathanson
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Stephanie K Seidlits
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA 90095, USA; Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Heldin P, Kolliopoulos C, Lin CY, Heldin CH. Involvement of hyaluronan and CD44 in cancer and viral infections. Cell Signal 2019; 65:109427. [PMID: 31654718 DOI: 10.1016/j.cellsig.2019.109427] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
Hyaluronan and its major receptor CD44 are ubiquitously distributed. They have important structural as well as signaling roles, regulating tissue homeostasis, and their expression levels are tightly regulated. In addition to signaling initiated by the interaction of the intracellular domain of CD44 with cytoplasmic signaling molecules, CD44 has important roles as a co-receptor for different types of receptors of growth factors and cytokines. Dysregulation of hyaluronan-CD44 interactions is seen in diseases, such as inflammation and cancer. In the present communication, we discuss the mechanism of hyaluronan-induced signaling via CD44, as well as the involvement of hyaluronan-engaged CD44 in malignancies and in viral infections.
Collapse
Affiliation(s)
- Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden.
| | - Constantinos Kolliopoulos
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Chun-Yu Lin
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden; Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University Department of Surgery, Uppsala University, Sweden; Department of Surgical Sciences, Uppsala University, Akademiska Hospital, 751 85 Uppsala, Sweden
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
31
|
Zargaran M, Baghaei F, Moghimbeigi A, Baghai F. Explanation for different behavior of peripheral and central giant cell granuloma by CD44 and CD34 immunostaining. ACTA ACUST UNITED AC 2019; 10:e12451. [DOI: 10.1111/jicd.12451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 04/12/2019] [Accepted: 07/29/2019] [Indexed: 12/01/2022]
Affiliation(s)
- Massoumeh Zargaran
- Dental Research Center, Department of Oral and Maxillofacial Pathology, School of Dentistry Hamadan University of Medical Sciences Hamadan Iran
| | - Fahimeh Baghaei
- Department of Oral and Maxillofacial Pathology, School of Dentistry Hamadan University of Medical Sciences Hamadan Iran
| | - Abbas Moghimbeigi
- Modeling of Noncommunicable Diseases Research Center, Department of Biostatistics, School of Public Health Hamadan University of Medical Sciences Hamadan Iran
| | - Fereshteh Baghai
- Dental Research Center, Dentistry Research Institute, Department of Oral and Maxillofacial Pathology School of Dentistry Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
32
|
Proinvasive extracellular matrix remodeling for tumor progression. Arch Pharm Res 2018; 42:40-47. [PMID: 30515725 DOI: 10.1007/s12272-018-1097-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
Abstract
Cancer is a systemic disease in which neoplastic cells interact with multiple types of non-neoplastic stromal cells as well as non-cellular components. The extracellular matrix (ECM) is a non-cellular component that is aberrantly regulated in many types of tumor microenvironments. Since the ECM generally maintains the tissue structure and provides mechanical forces in the tumor microenvironment, it has been simply assumed to act as a physical barrier for cancer metastasis and have a passive role in cancer progression. However, a substantial body of evidence has suggested that ECM remodeling influences many aspects of cancer cell behaviors and its importance has attracted attention in cancer biology. Abnormal ECM affects cancer progression through several ways such as inducing hypoxia, immune cells interaction by promoting mesenchymal shift and cell transformation. Accordingly, in this review we summarize and discusses the role of the ECM in modulating epithelial cells and surrounding stomatal cell components and considers its prospects in cancer biology.
Collapse
|
33
|
Fernández S, Córdoba M. Hyaluronic acid-induced capacitation involves protein kinase C and tyrosine kinase activity modulation with a lower oxidative metabolism in cryopreserved bull sperm. Theriogenology 2018; 122:68-73. [DOI: 10.1016/j.theriogenology.2018.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 10/28/2022]
|
34
|
Ouhtit A, Rizeq B, Saleh HA, Rahman MM, Zayed H. Novel CD44-downstream signaling pathways mediating breast tumor invasion. Int J Biol Sci 2018; 14:1782-1790. [PMID: 30443182 PMCID: PMC6231220 DOI: 10.7150/ijbs.23586] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/04/2018] [Indexed: 01/06/2023] Open
Abstract
CD44, also known as homing cell adhesion molecule is a multi-structural cell molecule involved in cell-cell and cell-extracellular matrix communications. CD44 regulates a number of central signaling pathways, including PI3K/AKT, Rho GTPases and the Ras-MAPK pathways, but also acts as a growth/arrest sensor, and inhibitor of angiogenesis and invasion, in response to signals from the microenvironment. The function of CD44 has been very controversial since it acts as both, a suppressor and a promoter of tumor growth and progression. To address this discrepancy, we have previously established CD44-inducible system both in vitro and in vivo. Next, using microarray analysis, we have identified and validated Survivin, Cortactin and TGF-β2 as novel CD44-downstream target genes, and characterized their signaling pathways underpinning CD44-promoted breast cancer (BC) cell invasion. This report aims to update the literature by adding and discussing the impact of these novel three signaling pathways to better understand the CD44-signaling pathways involved in BC tumor cell invasion.
Collapse
Affiliation(s)
- Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar
| | - Balsam Rizeq
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Haissam Abou Saleh
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar
| | - Md Mizanur Rahman
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
35
|
The biology and role of CD44 in cancer progression: therapeutic implications. J Hematol Oncol 2018; 11:64. [PMID: 29747682 PMCID: PMC5946470 DOI: 10.1186/s13045-018-0605-5] [Citation(s) in RCA: 762] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
CD44, a non-kinase transmembrane glycoprotein, is overexpressed in several cell types including cancer stem cells and frequently shows alternative spliced variants that are thought to play a role in cancer development and progression. Hyaluronan, the main ligand for CD44, binds to and activates CD44 resulting in activation of cell signaling pathways that induces cell proliferation, increases cell survival, modulates cytoskeletal changes, and enhances cellular motility. The different functional roles of CD44 standard (CD44s) and specific CD44 variant (CD44v) isoforms are not fully understood. CD44v contain additional peptide motifs that can interact with and sequester growth factors and cytokines at the cell surface thereby functioning as coreceptors to facilitate cell signaling. Moreover, CD44v were expressed in metastasized tumors, whereas switching between CD44v and CD44s may play a role in regulating epithelial to mesenchymal transition (EMT) and in the adaptive plasticity of cancer cells. Here, we review current data on the structural and functional properties of CD44, the known roles for CD44 in tumorigencity, the regulation of CD44 expression, and the potential for targeting CD44 for cancer therapy.
Collapse
|
36
|
Caccuri F, Ronca R, Laimbacher AS, Berenzi A, Steimberg N, Campilongo F, Mazzuca P, Giacomini A, Mazzoleni G, Benetti A, Caselli E, Presta M, Di Luca D, Fraefel C, Caruso A. U94 of human herpesvirus 6 down-modulates Src, promotes a partial mesenchymal-to-epithelial transition and inhibits tumor cell growth, invasion and metastasis. Oncotarget 2018; 8:44533-44549. [PMID: 28562350 PMCID: PMC5546500 DOI: 10.18632/oncotarget.17817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/21/2017] [Indexed: 12/31/2022] Open
Abstract
U94, the latency gene of human herpesvirus 6, was found to inhibit migration, invasion and proliferation of vascular endothelial cells (ECs). Because of its potent anti-migratory activity on ECs, we tested the capability of U94 to interfere with the individual steps of the metastatic cascade. We examined the U94 biological activity on the human breast cancer cell line MDA-MB 231, as a model of highly aggressive cancer cell. Here we show that the expression of U94 delivered by an HSV-1-based amplicon promoted down-modulation of Src and downstream molecules linked to cell motility and proliferation. Indeed, U94 expression strongly inhibited cell migration, invasiveness and clonogenicity. We investigated the effects of U94 in a three-dimensional rotary cell-culture system and observed the ability of U94 to modify tumor cell morphology by inducing a partial mesenchymal-to-epithelial transition. In fact, despite U94 did not induce any expression of the epithelial marker E-cadherin, it down-modulated different mesenchymal markers as β-catenin, Vimentin, TWIST, Snail1, and MMP2. In vivo data on the tumorigenicity of MDA-MB 231 displayed the capability of U94 to control tumor growth, invasiveness and metastasis, as well as tumor-driven angiogenesis. The antitumor U94 activity was also confirmed on the human cervical cancer cell line HeLa. The ability of U94 to inhibit cell growth, invasion and metastasis opens the way to a promising field of research aimed to develop new therapeutic approaches for treating tumor and cancer metastasis.
Collapse
Affiliation(s)
- Francesca Caccuri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Angiola Berenzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Nathalie Steimberg
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Federica Campilongo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Pietro Mazzuca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giovanna Mazzoleni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Anna Benetti
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Dario Di Luca
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
37
|
Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix Biol 2018; 78-79:292-313. [PMID: 29625181 DOI: 10.1016/j.matbio.2018.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The tissue microenvironment contributes to local immunity and to the pathogenesis of autoimmune diseases - a diverse set of conditions characterized by sterile inflammation, immunity against self-antigens, and destruction of tissues. However, the specific factors within the tissue microenvironment that contribute to local immune dysregulation in autoimmunity are poorly understood. One particular tissue component implicated in multiple autoimmune diseases is hyaluronan (HA), an extracellular matrix (ECM) polymer. HA is abundant in settings of chronic inflammation and contributes to lymphocyte activation, polarization, and migration. Here, we first describe what is known about the size, amount, and distribution of HA at sites of autoimmunity and in associated lymphoid structures in type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. Next, we examine the recent literature on HA and its impact on adaptive immunity, particularly in regards to the biology of lymphocytes and Foxp3+ regulatory T-cells (Treg), a T-cell subset that maintains immune tolerance in healthy individuals. We propose that HA accumulation at sites of chronic inflammation creates a permissive environment for autoimmunity, characterized by CD44-mediated inhibition of Treg expansion. Finally, we address potential tools and strategies for targeting HA and its receptor CD44 in chronic inflammation and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton L Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Wang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
38
|
Proinvasive extracellular matrix remodeling in tumor microenvironment in response to radiation. Oncogene 2018; 37:3317-3328. [DOI: 10.1038/s41388-018-0199-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/31/2017] [Accepted: 01/02/2018] [Indexed: 11/08/2022]
|
39
|
Liu HN, Guo NN, Wang TT, Guo WW, Lin MT, Huang-Fu MY, Vakili MR, Xu WH, Chen JJ, Wei QC, Han M, Lavasanifar A, Gao JQ. Mitochondrial Targeted Doxorubicin-Triphenylphosphonium Delivered by Hyaluronic Acid Modified and pH Responsive Nanocarriers to Breast Tumor: in Vitro and in Vivo Studies. Mol Pharm 2018; 15:882-891. [PMID: 29357260 DOI: 10.1021/acs.molpharmaceut.7b00793] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Multidrug resistance (MDR) is the major obstacle for chemotherapy. In a previous study, we have successfully synthesized a novel doxorubicin (DOX) derivative modified by triphenylphosphonium (TPP) to realize mitochondrial delivery of DOX and showed the potential of this compound to overcome DOX resistance in MDA-MB-435/DOX cells. (1) To introduce specificity for DOX-TPP to cancer cells, here we report on the conjugation of DOX-TPP to hyaluronic acid (HA) by hydrazone bond with adipic acid dihydrazide (ADH) as the acid-responsive linker, producing HA- hydra-DOX-TPP nanoparticles. Hyaluronic acid (HA) is a natural water-soluble linear glycosaminoglycan, which was hypothesized to increase the accumulation of nanoparticles containing DOX-TPP in the mitochondria of tumor cells upon systemic administration, overcoming DOX resistance, in vivo. Our results showed HA- hydra-DOX-TPP to self-assemble to core/shell nanoparticles of good dispersibility and effective release of DOX-TPP from the HA- hydra-DOX-TPP conjugate in cancer cells, which was followed by enhanced DOX mitochondria accumulation. The HA- hydra-DOX-TPP nanoparticles also showed improved anticancer effects, better tumor cell apoptosis, and better safety profile compared to free DOX in MCF-7/ADR bearing mice.
Collapse
Affiliation(s)
- Hui-Na Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Ning-Ning Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Tian-Tian Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Wang-Wei Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Meng-Ting Lin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Ming-Yi Huang-Fu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Mohammad Reza Vakili
- Faculty of Pharmacy and Pharmaceutical Sciences , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | - Wen-Hong Xu
- Department of Radiation Oncology, Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, the Second Affiliated Hospital , Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Jie-Jian Chen
- Department of Radiation Oncology, Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, the Second Affiliated Hospital , Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Qi-Chun Wei
- Department of Radiation Oncology, Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, the Second Affiliated Hospital , Zhejiang University, College of Medicine , Hangzhou , Zhejiang , China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou 310058 , China
| |
Collapse
|
40
|
|
41
|
Synthetic peptide TEKKRRETVEREKE derived from ezrin induces differentiation of NIH/3T3 fibroblasts. Eur J Pharmacol 2017; 811:249-259. [DOI: 10.1016/j.ejphar.2017.06.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 01/02/2023]
|
42
|
Delcourt V, Franck J, Leblanc E, Narducci F, Robin YM, Gimeno JP, Quanico J, Wisztorski M, Kobeissy F, Jacques JF, Roucou X, Salzet M, Fournier I. Combined Mass Spectrometry Imaging and Top-down Microproteomics Reveals Evidence of a Hidden Proteome in Ovarian Cancer. EBioMedicine 2017; 21:55-64. [PMID: 28629911 PMCID: PMC5514399 DOI: 10.1016/j.ebiom.2017.06.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recently, it was demonstrated that proteins can be translated from alternative open reading frames (altORFs), increasing the size of the actual proteome. Top-down mass spectrometry-based proteomics allows the identification of intact proteins containing post-translational modifications (PTMs) as well as truncated forms translated from reference ORFs or altORFs. METHODS Top-down tissue microproteomics was applied on benign, tumor and necrotic-fibrotic regions of serous ovarian cancer biopsies, identifying proteins exhibiting region-specific cellular localization and PTMs. The regions of interest (ROIs) were determined by MALDI mass spectrometry imaging and spatial segmentation. FINDINGS Analysis with a customized protein sequence database containing reference and alternative proteins (altprots) identified 15 altprots, including alternative G protein nucleolar 1 (AltGNL1) found in the tumor, and translated from an altORF nested within the GNL1 canonical coding sequence. Co-expression of GNL1 and altGNL1 was validated by transfection in HEK293 and HeLa cells with an expression plasmid containing a GNL1-FLAG(V5) construct. Western blot and immunofluorescence experiments confirmed constitutive co-expression of altGNL1-V5 with GNL1-FLAG. CONCLUSIONS Taken together, our approach provides means to evaluate protein changes in the case of serous ovarian cancer, allowing the detection of potential markers that have never been considered.
Collapse
Affiliation(s)
- Vivian Delcourt
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France; Département de Biochimie Lab. Z8-2001, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada
| | - Julien Franck
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Eric Leblanc
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France; Centre Oscar-Lambret, 3 Rue Frédéric Combemale, 59000 Lille, France
| | - Fabrice Narducci
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France; Centre Oscar-Lambret, 3 Rue Frédéric Combemale, 59000 Lille, France
| | - Yves-Marie Robin
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France; Centre Oscar-Lambret, 3 Rue Frédéric Combemale, 59000 Lille, France
| | - Jean-Pascal Gimeno
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France; ONCOLille, Maison Régionale de la Recherche Clinique, Lille, France
| | - Jusal Quanico
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Maxence Wisztorski
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Jean-François Jacques
- Département de Biochimie Lab. Z8-2001, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada
| | - Xavier Roucou
- Département de Biochimie Lab. Z8-2001, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Canada
| | - Michel Salzet
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France.
| | - Isabelle Fournier
- Université de Lille 1, INSERM, U1192, Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France.
| |
Collapse
|
43
|
Carvalho MP, Costa EC, Correia IJ. Assembly of breast cancer heterotypic spheroids on hyaluronic acid coated surfaces. Biotechnol Prog 2017; 33:1346-1357. [PMID: 28547896 DOI: 10.1002/btpr.2497] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/04/2017] [Indexed: 12/20/2022]
Abstract
Drug screening is currently demanding for realistic models that are able to reproduce the structural features of solid tumors. 3D cell culture systems, namely spheroids, emerged as a promising approach to provide reliable results during drug development. So far, liquid overlay technique (LOT) is one of the most used methods for spheroids assembly. It comprises cellular aggregation due to their limited adhesion to certain biomaterials, like agarose. However, researchers are currently improving this technique in order to obtain spheroids on surfaces that mimic cancer extracellular matrix (ECM), since cell-ECM interactions modulate cells behavior and their drug resistance profile. Herein, hyaluronic acid (HA) coated surfaces were used, for the first time, for the production of reproducible heterotypic breast cancer spheroids. The obtained results revealed that it is possible to control the size, shape, and number of spheroids gotten per well by changing the HA concentration and the number of cells initially seeded in each well. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1346-1357, 2017.
Collapse
Affiliation(s)
- Marco P Carvalho
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| | - Elisabete C Costa
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| | - Ilídio J Correia
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, 6200-506, Portugal
| |
Collapse
|
44
|
Fernández S, Córdoba M. A membrane-associated adenylate cyclase modulates lactate dehydrogenase and creatine kinase activities required for bull sperm capacitation induced by hyaluronic acid. Anim Reprod Sci 2017; 179:80-87. [DOI: 10.1016/j.anireprosci.2017.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 02/07/2023]
|
45
|
Kuo YZ, Fang WY, Huang CC, Tsai ST, Wang YC, Yang CL, Wu LW. Hyaluronan synthase 3 mediated oncogenic action through forming inter-regulation loop with tumor necrosis factor alpha in oral cancer. Oncotarget 2017; 8:15563-15583. [PMID: 28107185 PMCID: PMC5362506 DOI: 10.18632/oncotarget.14697] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/27/2016] [Indexed: 12/31/2022] Open
Abstract
Hyaluronan (HA) is a major extracellular matrix component. However, its role and mediation in oral cancer remains elusive. Hyaluronan synthase 3 (HAS3), involved in pro-inflammatory short chain HA synthesis, was the predominant synthase in oral cancer cells and tissues. HAS3 overexpression significantly increased oral cancer cell migration, invasion and xenograft tumorigenesis accompanied with the increased expression of tumor necrosis factor alpha (TNF-α) and monocyte chemoattractant protein 1 (MCP-1). Conversely, HAS3 depletion abrogated HAS3-mediated stimulation. HAS3 induced oncogenic actions partly through activating EGFR-SRC signaling. HAS3-derived HA release into extracellular milieu enhanced transendothelial monocyte migration and MCP-1 expression, which was attenuated by anti-HAS3 antibodies or a HAS inhibitor, 4-Methylumbelliferone (4-MU). The NF-κB-binding site III at -1692 to -1682 bp upstream from the transcript 1 start site in HAS3 proximal promoter was the most responsive to TNF-α-stimulated transcription. ChIP-qPCR analysis confirmed the highest NF-κB-p65 enrichment on site III. Increased HAS3 mRNA expression was negatively correlated with the overall survival of oral cancer patients. A concomitant increase of TNF-α, a stimulus for HAS3 expression, with HAS3 expression was not only associated with lymph node metastasis but also negated clinical outcome. Together, HAS3 and TNF-α formed an inter-regulation loop to enhance tumorigenesis in oral cancer.
Collapse
Affiliation(s)
- Yi-Zih Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C
| | - Wei-Yu Fang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C
| | - Cheng-Chih Huang
- Department of Otolaryngology, National Cheng Kung University Hospital, Tainan 70428, Taiwan, R.O.C
| | - Sen-Tien Tsai
- Department of Otolaryngology, National Cheng Kung University Hospital, Tainan 70428, Taiwan, R.O.C.,Department of Radiation Oncology, National Cheng Kung University Hospital, Tainan 70428, Taiwan, R.O.C
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C
| | - Chih-Li Yang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C
| | - Li-Wha Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C.,Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, R.O.C.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| |
Collapse
|
46
|
NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling. Oncotarget 2016; 7:10228-42. [PMID: 26849234 PMCID: PMC4891116 DOI: 10.18632/oncotarget.7169] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 01/21/2016] [Indexed: 11/25/2022] Open
Abstract
Neuromedin U (NMU) was originally named based on its strong uterine contractile activity, but little is known regarding its signaling/functions in utero. We identified that NMU and one of its receptors, NMUR2, are not only present in normal uterine endometrium but also co-expressed in endometrial cancer tissues, where the NMU level is correlated with the malignant grades and survival of patients. Cell-based assays further confirmed that NMU signaling can promote cell motility and proliferation of endometrial cancer cells derived from grade II tumors. Activation of NMU pathway in these endometrial cancer cells is required in order to sustain expression of various adhesion molecules, such as CD44 and integrin alpha1, as well as production of their corresponding extracellular matrix ligands, hyaluronan and collagen IV; it also increased the activity of SRC and its downstream proteins RHOA and RAC1. Thus, it is concluded that NMU pathway positively controls the adhesion signaling-SRC-Rho GTPase axis in the tested endometrial cancer cells and that changes in cell motility and proliferation can occur when there is manipulation of NMU signaling in these cells either in vitro or in vivo. Intriguingly, this novel mechanism also explains how NMU signaling promotes the EGFR-driven and TGFβ receptor-driven mesenchymal transitions. Through the above axis, NMU signaling not only can promote malignancy of the tested endometrial cancer cells directly, but also helps these cells to become more sensitive to niche growth factors in their microenvironment.
Collapse
|
47
|
Yang Y, Zhao X, Li X, Yan Z, Liu Z, Li Y. Effects of anti-CD44 monoclonal antibody IM7 carried with chitosan polylactic acid-coated nano-particles on the treatment of ovarian cancer. Oncol Lett 2016; 13:99-104. [PMID: 28123528 PMCID: PMC5245159 DOI: 10.3892/ol.2016.5413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/11/2016] [Indexed: 12/28/2022] Open
Abstract
Failure in early diagnosis and ineffective treatment are the major causes of ovarian cancer mortality. Hyaluronan and its receptor, cluster of differentiation (CD)44, have been considered to be valid targets for treating cancer. The anti-CD44 monoclonal antibody IM7 is effective in treating ovarian cancer; however, its toxicity should not be ignored. The present study has developed a new drug carrier system composed of chitosan nano-particles coated with polylactic acid (PLA) to improve the treatment efficacy and reduce toxicity. An ionic crosslinking method and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride/N-hydroxysuccinimide were used to prepare the IM7 antibody, which was loaded with chitosan nano-particles. The surfaces of the nano-particles were coated with PLA to generate PLA-chitosan-IM7. Subsequently, transmission electron microscopy (TEM) was used to observe the size and zeta potential of the nano-particles. In addition, a spectrophotometer was used to calculate the loading rate and release rate of the nano-particles in acidic and neutral environments. MTT assay was used to evaluate the anti-proliferative effect of PLA-chitosan-IM7 on the human ovarian cancer cell line HO-8910PM. In addition, an in vivo imaging system was used to further investigate the effect of PLA-chitosan-IM7 on the treatment of mice with ovarian cancer. A total of 35 days subsequent to PLA-chitosan-IM7 treatment, all animals were sacrificed by CO2, and the tumors were removed and weighted. The PLA-chitosan-IM7 nano-particles were successfully prepared, since TEM revealed that their size was 300–400 nm and their zeta potential was +25 mV. According to the spectrophotometry results, the loading rate was 52%, and PLA-chitosan-IM7 exhibited good resistance to acids. MTT assay demonstrated that PLA-chitosan-IM7 could suppress the proliferation of HO-8910PM cells in vitro. The in vivo imaging system revealed that PLA-chitosan-IM7 was effective in controlling the development of human ovarian cancer cells and the tumor weight. These results suggest that PLA-chitosan-IM7 could be effective in treating cancers in vitro and in vivo, which may provide a novel approach to enhance the effectiveness of anti-CD44 treatment while reducing its toxicity.
Collapse
Affiliation(s)
- Yizhuo Yang
- Department of Obstetrics and Gynecology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Xinghui Zhao
- State Key Laboratory of Pathogens and Biosecurity, Laboratory of Applied Molecular Biology, Beijing Institute of Microbiology and Epidemiology, Beijing 100853, P.R. China
| | - Xiuli Li
- Department of Obstetrics and Gynecology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Zhifeng Yan
- Department of Obstetrics and Gynecology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Zhongyu Liu
- Department of Obstetrics and Gynecology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Yali Li
- Department of Obstetrics and Gynecology, General Hospital of PLA, Beijing 100853, P.R. China
| |
Collapse
|
48
|
Ablation of CD44 induces glycolysis-to-oxidative phosphorylation transition via modulation of the c-Src–Akt–LKB1–AMPKα pathway. Biochem J 2016; 473:3013-30. [DOI: 10.1042/bcj20160613] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/25/2016] [Indexed: 01/16/2023]
Abstract
Cluster of differentiation 44 (CD44) is a transmembrane glycoprotein that has been identified as a cancer stem cell marker in various cancer cells. Although many studies have focused on CD44 as a cancer stem cell marker, its effect on cancer cell metabolism remains unclear. To investigate the role of CD44 on cancer cell metabolism, we established CD44 knock-down cells via retroviral delivery of shRNA against CD44 in human breast cancer cells. Silencing of CD44 decreased the glycolytic phenotype of cancer cells, affecting glucose uptake, ATP production, and lactate production. We also found that ablation of the CD44-induced lactate dehydrogenase (LDH) isoenzyme results in a shift to LDH1 due to LDHA down-regulation and LDHB up-regulation, implying the importance of LDH isoenzyme modulation on cancer metabolism. The expression of glycolysis-related proteins including hypoxia inducible factor-1α (HIF-1α) and LDHA was decreased by CD44 silencing. These effects were due to the up-regulation of liver kinase B1 (LKB1)/AMP-activated protein kinase (AMPK)α activity by reduction in c-Src and Akt activity in CD44 knock-down cells. Finally, induction of LKB1/AMPKα activity blocked the expression of HIF-1α and its target gene, LDHA. Inversely, LDHB expression was repressed by HIF-1α. Collectively, these results indicate that the CD44 silencing-induced metabolic shift is mediated by the regulation of c-Src/Akt/LKB1/AMPKα/HIF-1α signaling in human breast cancer cells.
Collapse
|
49
|
Nelson M, Yang M, Millican-Slater R, Brackenbury WJ. Nav1.5 regulates breast tumor growth and metastatic dissemination in vivo. Oncotarget 2016; 6:32914-29. [PMID: 26452220 PMCID: PMC4741739 DOI: 10.18632/oncotarget.5441] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/25/2015] [Indexed: 02/07/2023] Open
Abstract
Voltage-gated Na+ channels (VGSCs) mediate action potential firing and regulate adhesion and migration in excitable cells. VGSCs are also expressed in cancer cells. In metastatic breast cancer (BCa) cells, the Nav1.5 α subunit potentiates migration and invasion. In addition, the VGSC-inhibiting antiepileptic drug phenytoin inhibits tumor growth and metastasis. However, the functional activity of Nav1.5 and its specific contribution to tumor progression in vivo has not been delineated. Here, we found that Nav1.5 is up-regulated at the protein level in BCa compared with matched normal breast tissue. Na+ current, reversibly blocked by tetrodotoxin, was retained in cancer cells in tumor tissue slices, thus directly confirming functional VGSC activity in vivo. Stable down-regulation of Nav1.5 expression significantly reduced tumor growth, local invasion into surrounding tissue, and metastasis to liver, lungs and spleen in an orthotopic BCa model. Nav1.5 down-regulation had no effect on cell proliferation or angiogenesis within the in tumors, but increased apoptosis. In vitro, Nav1.5 down-regulation altered cell morphology and reduced CD44 expression, suggesting that VGSC activity may regulate cellular invasion via the CD44-src-cortactin signaling axis. We conclude that Nav1.5 is functionally active in cancer cells in breast tumors, enhancing growth and metastatic dissemination. These findings support the notion that compounds targeting Nav1.5 may be useful for reducing metastasis.
Collapse
Affiliation(s)
- Michaela Nelson
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Ming Yang
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | | | | |
Collapse
|
50
|
Liu RM, Sun RG, Zhang LT, Zhang QF, Chen DX, Zhong JJ, Xiao JH. Hyaluronic acid enhances proliferation of human amniotic mesenchymal stem cells through activation of Wnt/β-catenin signaling pathway. Exp Cell Res 2016; 345:218-29. [PMID: 27237096 DOI: 10.1016/j.yexcr.2016.05.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/15/2016] [Accepted: 05/23/2016] [Indexed: 01/11/2023]
|