1
|
Sheida A, Farshadi M, Mirzaei A, Najjar Khalilabad S, Zarepour F, Taghavi SP, Hosseini Khabr MS, Ravaei F, Rafiei S, Mosadeghi K, Yazdani MS, Fakhraie A, Ghattan A, Zamani Fard MM, Shahyan M, Rafiei M, Rahimian N, Talaei Zavareh SA, Mirzaei H. Potential of Natural Products in the Treatment of Glioma: Focus on Molecular Mechanisms. Cell Biochem Biophys 2024; 82:3157-3208. [PMID: 39150676 DOI: 10.1007/s12013-024-01447-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Despite the waning of traditional treatments for glioma due to possible long-term issues, the healing possibilities of substances derived from nature have been reignited in the scientific community. These natural substances, commonly found in fruits and vegetables, are considered potential alternatives to pharmaceuticals, as they have been shown in prior research to impact pathways surrounding cancer progression, metastases, invasion, and resistance. This review will explore the supposed molecular mechanisms of different natural components, such as berberine, curcumin, coffee, resveratrol, epigallocatechin-3-gallate, quercetin, tanshinone, silymarin, coumarin, and lycopene, concerning glioma treatment. While the benefits of a balanced diet containing these compounds are widely recognized, there is considerable scope for investigating the efficacy of these natural products in treating glioma.
Collapse
Affiliation(s)
- Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Mirzaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Najjar Khalilabad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Rafiei
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kimia Mosadeghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sepehr Yazdani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Fakhraie
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Ghattan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Shahyan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
Memari E, Khan D, Alkins R, Helfield B. Focused ultrasound-assisted delivery of immunomodulating agents in brain cancer. J Control Release 2024; 367:283-299. [PMID: 38266715 DOI: 10.1016/j.jconrel.2024.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Focused ultrasound (FUS) combined with intravascularly circulating microbubbles can transiently increase the permeability of the blood-brain barrier (BBB) to enable targeted therapeutic delivery to the brain, the clinical testing of which is currently underway in both adult and pediatric patients. Aside from traditional cancer drugs, this technique is being extended to promote the delivery of immunomodulating therapeutics to the brain, including antibodies, immune cells, and cytokines. In this manner, FUS approaches are being explored as a tool to improve and amplify the effectiveness of immunotherapy for both primary and metastatic brain cancer, a particularly challenging solid tumor to treat. Here, we present an overview of the latest groundbreaking research in FUS-assisted delivery of immunomodulating agents to the brain in pre-clinical models of brain cancer, and place it within the context of the current immunotherapy approaches. We follow this up with a discussion on new developments and emerging strategies for this rapidly evolving approach.
Collapse
Affiliation(s)
- Elahe Memari
- Department of Physics, Concordia University, Montreal H4B 1R6, Canada
| | - Dure Khan
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Ryan Alkins
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Division of Neurosurgery, Department of Surgery, Kingston Health Sciences Centre, Queen's University, Kingston, ON, Canada
| | - Brandon Helfield
- Department of Physics, Concordia University, Montreal H4B 1R6, Canada; Department of Biology, Concordia University, Montreal H4B 1R6, Canada.
| |
Collapse
|
3
|
Nahar Metu CL, Sutihar SK, Sohel M, Zohora F, Hasan A, Miah MT, Rani Kar T, Hossain MA, Rahman MH. Unraveling the signaling mechanism behind astrocytoma and possible therapeutics strategies: A comprehensive review. Cancer Rep (Hoboken) 2023; 6:e1889. [PMID: 37675821 PMCID: PMC10598261 DOI: 10.1002/cnr2.1889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/09/2023] [Accepted: 07/28/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND A form of cancer called astrocytoma can develop in the brain or spinal cord and sometimes causes death. A detailed overview of the precise signaling cascade underlying astrocytoma formation has not yet been revealed, although various factors have been investigated. Therefore, our objective was to unravel and summarize our current understanding of molecular genetics and associated signaling pathways with some possible therapeutic strategies for astrocytoma. RECENT FINDINGS In general, four different forms of astrocytoma have been identified in individuals, including circumscribed, diffuse, anaplastic, and multiforme glioblastoma, according to a recent literature review. All types of astrocytoma have a direct connection with some oncogenic signaling cascade. Common signaling is MAPK cascade, including Ras-Raf-ERK, up-regulated with activating EGFR/AKT/PTEN/mTOR and PDGFR. Recent breakthrough studies found that BRAF mutations, including KIAA1549: BRAF and BRAF V600E are responsible for astrocytoma progression. Additionally, cancer progression is influenced by mutations in some tumor suppressor genes, such as the Tp53/ATRX and MGMT mutant. As synthetic medications must cross the blood-brain barrier (BBB), modulating signal systems such as miRNA is the primary option for treating patients with astrocytoma. However, available surgery, radiation therapy, and experimental therapies such as adjuvant therapy, anti-angiogenic therapy, and EGFR-targeting antibody drug are the usual treatment for most types of astrocytoma. Similar to conventional anticancer medications, some phytochemicals slow tumor growth by simultaneously controlling several cellular proteins, including those involved in cell cycle regulation, apoptosis, metastatic spread, tyrosine kinase, growth factor receptor, and antioxidant-related proteins. CONCLUSION In conclusion, cellular and molecular signaling is directly associated with the development of astrocytoma, and a combination of conventional and alternative therapies can improve the malignancy of cancer patients.
Collapse
Affiliation(s)
- Chowdhury Lutfun Nahar Metu
- Biochemistry and Molecular BiologyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganjBangladesh
| | - Sunita Kumari Sutihar
- Biochemistry and Molecular BiologyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganjBangladesh
| | - Md Sohel
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
- Department of Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Fatematuz Zohora
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Akayed Hasan
- Department of PharmacyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Md. Thandu Miah
- Department of PharmacyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Tanu Rani Kar
- Department of Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Md. Arju Hossain
- Department of Biotechnology and Genetic EngineeringMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Md Habibur Rahman
- Department of Computer Science and EngineeringIslamic UniversityKushtiaBangladesh
| |
Collapse
|
4
|
Erices JI, Bizama C, Niechi I, Uribe D, Rosales A, Fabres K, Navarro-Martínez G, Torres Á, San Martín R, Roa JC, Quezada-Monrás C. Glioblastoma Microenvironment and Invasiveness: New Insights and Therapeutic Targets. Int J Mol Sci 2023; 24:ijms24087047. [PMID: 37108208 PMCID: PMC10139189 DOI: 10.3390/ijms24087047] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 04/29/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain cancer in adults. Without treatment the mean patient survival is approximately 6 months, which can be extended to 15 months with the use of multimodal therapies. The low effectiveness of GBM therapies is mainly due to the tumor infiltration into the healthy brain tissue, which depends on GBM cells' interaction with the tumor microenvironment (TME). The interaction of GBM cells with the TME involves cellular components such as stem-like cells, glia, endothelial cells, and non-cellular components such as the extracellular matrix, enhanced hypoxia, and soluble factors such as adenosine, which promote GBM's invasiveness. However, here we highlight the role of 3D patient-derived glioblastoma organoids cultures as a new platform for study of the modeling of TME and invasiveness. In this review, the mechanisms involved in GBM-microenvironment interaction are described and discussed, proposing potential prognosis biomarkers and new therapeutic targets.
Collapse
Affiliation(s)
- José Ignacio Erices
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Carolina Bizama
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Ignacio Niechi
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Daniel Uribe
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Arnaldo Rosales
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Karen Fabres
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Giovanna Navarro-Martínez
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Ángelo Torres
- Escuela de Medicina Veterinaria, Facultad de Recursos Naturales y Medicina Veterinaria, Universidad Santo Tomás, Talca 8370003, Chile
| | - Rody San Martín
- Laboratorio de Patología Molecular, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Claudia Quezada-Monrás
- Laboratorio de Biología Tumoral, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia 5090000, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| |
Collapse
|
5
|
Pratt J, Haidara K, Annabi B. MT1-MMP Expression Levels and Catalytic Functions Dictate LDL Receptor-Related Protein-1 Ligand Internalization Capacity in U87 Glioblastoma Cells. Int J Mol Sci 2022; 23:ijms232214214. [PMID: 36430705 PMCID: PMC9692856 DOI: 10.3390/ijms232214214] [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: 09/02/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Modulations in cell surface receptor ectodomain proteolytic shedding impact on receptor function and cancer biomarker expression. As such, heavily pursued therapeutic avenues have exploited LDL receptor-related protein-1 (LRP-1)-mediated capacity in internalizing Angiopep-2 (An2), a brain-penetrating peptide that allows An2-drug conjugates to cross the blood-brain tumor barrier (BBTB). Given that LRP-1 is proteolytically shed from the cell surface through matrix metalloproteinase (MMP) activity, the balance between MMP expression/function and LRP-1-mediated An2 internalization is unknown. In this study, we found that membrane type-1 (MT1)-MMP expression increased from grade 1 to 4 brain tumors, while that of LRP-1 decreased inversely. MMP pharmacological inhibitors such as Ilomastat, Doxycycline and Actinonin increased in vitro An2 internalization by up to 2.5 fold within a human grade IV-derived U87 glioblastoma cell model. Transient siRNA-mediated MT1-MMP gene silencing resulted in increased basal An2 cell surface binding and intracellular uptake, while recombinant MT1-MMP overexpression reduced both cell surface LRP-1 expression as well as An2 internalization. The addition of Ilomastat to cells overexpressing recombinant MT1-MMP restored LRP-1 expression at the cell surface and An2 uptake to levels comparable to those observed in control cells. Collectively, our data suggest that MT1-MMP expression status dictates An2-mediated internalization processes in part by regulating cell surface LRP-1 functions. Such evidence prompts preclinical evaluations of combined MMP inhibitors/An2-drug conjugate administration to potentially increase the treatment of high-MT1-MMP-expressing brain tumors.
Collapse
|
6
|
Meesaragandla B, Hayet S, Fine T, Janke U, Chai L, Delcea M. Inhibitory Effect of Epigallocatechin Gallate-Silver Nanoparticles and Their Lysozyme Bioconjugates on Biofilm Formation and Cytotoxicity. ACS APPLIED BIO MATERIALS 2022; 5:4213-4221. [PMID: 35977081 PMCID: PMC9490750 DOI: 10.1021/acsabm.2c00409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022]
Abstract
Biofilms are multicellular communities of microbial cells that grow on natural and synthetic surfaces. They have become the major cause for hospital-acquired infections because once they form, they are very difficult to eradicate. Nanotechnology offers means to fight biofilm-associated infections. Here, we report on the synthesis of silver nanoparticles (AgNPs) with the antibacterial ligand epigallocatechin gallate (EGCG) and the formation of a lysozyme protein corona on AgNPs, as shown by UV-vis, dynamic light scattering, and circular dichroism analyses. We further tested the activity of EGCG-AgNPs and their lysozyme bioconjugates on the viability of Bacillus subtilis cells and biofilm formation. Our results showed that, although EGCG-AgNPs presented no antibacterial activity on planktonic B. subtilis cells, they inhibited B. subtilis biofilm formation at concentrations larger than 40 nM, and EGCG-AgNP-lysozyme bioconjugates inhibited biofilms at concentrations above 80 nM. Cytotoxicity assays performed with human cells showed a reverse trend, where EGCG-AgNPs barely affected human cell viability while EGCG-AgNP-lysozyme bioconjugates severely hampered viability. Our results therefore demonstrate that EGCG-AgNPs may be used as noncytotoxic antibiofilm agents.
Collapse
Affiliation(s)
- Brahmaiah Meesaragandla
- Institute
of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489 Greifswald, Germany
- ZIK
HIKE—Zentrum für Innovationskompetenz “Humorale
Immunreaktionen bei kardiovaskulären Erkrankungen”, Fleischmannstraße 42, 17489 Greifswald, Germany
| | - Shahar Hayet
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, 91904 Jerusalem, Israel
- The
Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, 91904 Jerusalem, Israel
| | - Tamir Fine
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, 91904 Jerusalem, Israel
- The
Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, 91904 Jerusalem, Israel
| | - Una Janke
- Institute
of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489 Greifswald, Germany
- ZIK
HIKE—Zentrum für Innovationskompetenz “Humorale
Immunreaktionen bei kardiovaskulären Erkrankungen”, Fleischmannstraße 42, 17489 Greifswald, Germany
| | - Liraz Chai
- Institute
of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, 91904 Jerusalem, Israel
- The
Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond J. Safra Campus, 91904 Jerusalem, Israel
| | - Mihaela Delcea
- Institute
of Biochemistry, University of Greifswald, Felix-Hausdorff-Straße 4, 17489 Greifswald, Germany
- ZIK
HIKE—Zentrum für Innovationskompetenz “Humorale
Immunreaktionen bei kardiovaskulären Erkrankungen”, Fleischmannstraße 42, 17489 Greifswald, Germany
- DZHK
(Deutsches Zentrum für Herz-Kreislauf-Forschung), Partner Site
Greifswald, 17489 Greifswald, Germany
| |
Collapse
|
7
|
Becker KN, Pettee KM, Sugrue A, Reinard KA, Schroeder JL, Eisenmann KM. The Cytoskeleton Effectors Rho-Kinase (ROCK) and Mammalian Diaphanous-Related (mDia) Formin Have Dynamic Roles in Tumor Microtube Formation in Invasive Glioblastoma Cells. Cells 2022; 11:1559. [PMID: 35563863 PMCID: PMC9103681 DOI: 10.3390/cells11091559] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/21/2022] [Accepted: 04/30/2022] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is a progressive and lethal brain cancer. Malignant control of actin and microtubule cytoskeletal mechanics facilitates two major GBM therapeutic resistance strategies-diffuse invasion and tumor microtube network formation. Actin and microtubule reorganization is controlled by Rho-GTPases, which exert their effects through downstream effector protein activation, including Rho-associated kinases (ROCK) 1 and 2 and mammalian diaphanous-related (mDia) formins (mDia1, 2, and 3). Precise spatial and temporal balancing of the activity between these effectors dictates cell shape, adhesion turnover, and motility. Using small molecules targeting mDia, we demonstrated that global agonism (IMM02) was superior to antagonism (SMIFH2) as anti-invasion strategies in GBM spheroids. Here, we use IDH-wild-type GBM patient-derived cell models and a novel semi-adherent in vitro system to investigate the relationship between ROCK and mDia in invasion and tumor microtube networks. IMM02-mediated mDia agonism disrupts invasion in GBM patient-derived spheroid models, in part by inducing mDia expression loss and tumor microtube network collapse. Pharmacological disruption of ROCK prevented invasive cell-body movement away from GBM spheres, yet induced ultralong, phenotypically abnormal tumor microtube formation. Simultaneously targeting mDia and ROCK did not enhance the anti-invasive/-tumor microtube effects of IMM02. Our data reveal that targeting mDia is a viable GBM anti-invasion/-tumor microtube networking strategy, while ROCK inhibition is contraindicated.
Collapse
Affiliation(s)
- Kathryn N. Becker
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (K.N.B.); (K.M.P.)
| | - Krista M. Pettee
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (K.N.B.); (K.M.P.)
| | - Amanda Sugrue
- Department of Chemistry and Biochemistry, University of Heidelberg, Tiffin, OH 44883, USA;
| | - Kevin A. Reinard
- Division of Neurosurgery, ProMedica Toledo Hospital, Toledo, OH 43606, USA; (K.A.R.); (J.L.S.)
| | - Jason L. Schroeder
- Division of Neurosurgery, ProMedica Toledo Hospital, Toledo, OH 43606, USA; (K.A.R.); (J.L.S.)
- Department of Surgery, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Kathryn M. Eisenmann
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (K.N.B.); (K.M.P.)
| |
Collapse
|
8
|
Persano F, Gigli G, Leporatti S. Natural Compounds as Promising Adjuvant Agents in The Treatment of Gliomas. Int J Mol Sci 2022; 23:3360. [PMID: 35328780 PMCID: PMC8955269 DOI: 10.3390/ijms23063360] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023] Open
Abstract
In humans, glioblastoma is the most prevalent primary malignant brain tumor. Usually, glioblastoma has specific characteristics, such as aggressive cell proliferation and rapid invasion of surrounding brain tissue, leading to a poor patient prognosis. The current therapy-which provides a multidisciplinary approach with surgery followed by radiotherapy and chemotherapy with temozolomide-is not very efficient since it faces clinical challenges such as tumor heterogeneity, invasiveness, and chemoresistance. In this respect, natural substances in the diet, integral components in the lifestyle medicine approach, can be seen as potential chemotherapeutics. There are several epidemiological studies that have shown the chemopreventive role of natural dietary compounds in cancer progression and development. These heterogeneous compounds can produce anti-glioblastoma effects through upregulation of apoptosis and autophagy; allowing the promotion of cell cycle arrest; interfering with tumor metabolism; and permitting proliferation, neuroinflammation, chemoresistance, angiogenesis, and metastasis inhibition. Although these beneficial effects are promising, the efficacy of natural compounds in glioblastoma is limited due to their bioavailability and blood-brain barrier permeability. Thereby, further clinical trials are necessary to confirm the in vitro and in vivo anticancer properties of natural compounds. In this article, we overview the role of several natural substances in the treatment of glioblastoma by considering the challenges to be overcome and future prospects.
Collapse
Affiliation(s)
- Francesca Persano
- Department of Mathematics and Physics, University of Salento, Via Per Arnesano, 73100 Lecce, Italy;
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| | - Giuseppe Gigli
- Department of Mathematics and Physics, University of Salento, Via Per Arnesano, 73100 Lecce, Italy;
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| | - Stefano Leporatti
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
9
|
Pibuel MA, Poodts D, Díaz M, Hajos SE, Lompardía SL. The scrambled story between hyaluronan and glioblastoma. J Biol Chem 2021; 296:100549. [PMID: 33744285 PMCID: PMC8050860 DOI: 10.1016/j.jbc.2021.100549] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Advances in cancer biology are revealing the importance of the cancer cell microenvironment on tumorigenesis and cancer progression. Hyaluronan (HA), the main glycosaminoglycan in the extracellular matrix, has been associated with the progression of glioblastoma (GBM), the most frequent and lethal primary tumor in the central nervous system, for several decades. However, the mechanisms by which HA impacts GBM properties and processes have been difficult to elucidate. In this review, we provide a comprehensive assessment of the current knowledge on HA's effects on GBM biology, introducing its primary receptors CD44 and RHAMM and the plethora of relevant downstream signaling pathways that can scramble efforts to directly link HA activity to biological outcomes. We consider the complexities of studying an extracellular polymer and the different strategies used to try to capture its function, including 2D and 3D in vitro studies, patient samples, and in vivo models. Given that HA affects not only migration and invasion, but also cell proliferation, adherence, and chemoresistance, we highlight the potential role of HA as a therapeutic target. Finally, we review the different existing approaches to diminish its protumor effects, such as the use of 4-methylumbelliferone, HA oligomers, and hyaluronidases and encourage further research along these lines in order to improve the survival and quality of life of GBM patients.
Collapse
Affiliation(s)
- Matías Arturo Pibuel
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| | - Daniela Poodts
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvia Elvira Hajos
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvina Laura Lompardía
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| |
Collapse
|
10
|
Aggarwal V, Tuli HS, Tania M, Srivastava S, Ritzer EE, Pandey A, Aggarwal D, Barwal TS, Jain A, Kaur G, Sak K, Varol M, Bishayee A. Molecular mechanisms of action of epigallocatechin gallate in cancer: Recent trends and advancement. Semin Cancer Biol 2020; 80:256-275. [PMID: 32461153 DOI: 10.1016/j.semcancer.2020.05.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/08/2020] [Accepted: 05/17/2020] [Indexed: 12/22/2022]
Abstract
Epigallocatechin gallate (EGCG), also known as epigallocatechin-3-gallate, is an ester of epigallocatechin and gallic acid. EGCG, abundantly found in tea, is a polyphenolic flavonoid that has the potential to affect human health and disease. EGCG interacts with various recognized cellular targets and inhibits cancer cell proliferation by inducing apoptosis and cell cycle arrest. In addition, scientific evidence has illustrated the promising role of EGCG in inhibiting tumor cell metastasis and angiogenesis. It has also been found that EGCG may reverse drug resistance of cancer cells and could be a promising candidate for synergism studies. The prospective importance of EGCG in cancer treatment is owed to its natural origin, safety, and low cost which presents it as an attractive target for further development of novel cancer therapeutics. A major challenge with EGCG is its low bioavailability which is being targeted for improvement by encapsulating EGCG in nano-sized vehicles for further delivery. However, there are major limitations of the studies on EGCG, including study design, experimental bias, and inconsistent results and reproducibility among different study cohorts. Additionally, it is important to identify specific EGCG pharmacological targets in the tumor-specific signaling pathways for development of novel combined therapeutic treatments with EGCG. The present review highlights the ongoing development to identify cellular and molecular targets of EGCG in cancer. Furthermore, the role of nanotechnology-mediated EGCG combinations and delivery systems will also be discussed.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh 160 012, Punjab, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133 207, Haryana, India.
| | - Mousumi Tania
- Division of Molecular Cancer, Red Green Research Center, Dhaka 1205, Bangladesh
| | - Saumya Srivastava
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211 004, Uttar Pradesh, India
| | - Erin E Ritzer
- Lake Erie College of Osteopathic Medicine, Bradenton 34211, FL, USA
| | - Anjana Pandey
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211 004, Uttar Pradesh, India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133 207, Haryana, India
| | - Tushar Singh Barwal
- Department of Zoology, Central University of Punjab, Bathinda 151 001, Punjab, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda 151 001, Punjab, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Mumbai 400 056, Maharastra, India
| | | | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Muğla TR48000, Turkey
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton 34211, FL, USA.
| |
Collapse
|
11
|
Cao H, Li X, Wang F, Zhang Y, Xiong Y, Yang Q. Phytochemical-Mediated Glioma Targeted Treatment: Drug Resistance and Novel Delivery Systems. Curr Med Chem 2020; 27:599-629. [PMID: 31400262 DOI: 10.2174/0929867326666190809221332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 03/15/2019] [Accepted: 07/23/2019] [Indexed: 02/08/2023]
Abstract
Glioma, especially its most malignant type, Glioblastoma (GBM), is the most common and the most aggressive malignant tumour in the central nervous system. Currently, we have no specific therapies that can significantly improve its dismal prognosis. Recent studies have reported promising in vitro experimental results of several novel glioma-targeting drugs; these studies are encouraging to both researchers and patients. However, clinical trials have revealed that novel compounds that focus on a single, clear glioma genetic alteration may not achieve a satisfactory outcome or have side effects that are unbearable. Based on this consensus, phytochemicals that exhibit multiple bioactivities have recently attracted much attention. Traditional Chinese medicine and traditional Indian medicine (Ayurveda) have shown that phytocompounds inhibit glioma angiogenesis, cancer stem cells and tumour proliferation; these results suggest a novel drug therapeutic strategy. However, single phytocompounds or their direct usage may not reverse comprehensive malignancy due to poor histological penetrability or relatively unsatisfactory in vivo efficiency. Recent research that has employed temozolomide combination treatment and Nanoparticles (NPs) with phytocompounds has revealed a powerful dual-target therapy and a high blood-brain barrier penetrability, which is accompanied by low side effects and strong specific targeting. This review is focused on major phytocompounds that have contributed to glioma-targeting treatment in recent years and their role in drug resistance inhibition, as well as novel drug delivery systems for clinical strategies. Lastly, we summarize a possible research strategy for the future.
Collapse
Affiliation(s)
- Hang Cao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Feiyifan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yueqi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
The Role of Rho GTPases in Motility and Invasion of Glioblastoma Cells. Anal Cell Pathol (Amst) 2020; 2020:9274016. [PMID: 32089990 PMCID: PMC7013281 DOI: 10.1155/2020/9274016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 12/27/2022] Open
Abstract
Astrocytomas are primary malignant brain tumors that originate from astrocytes. Grade IV astrocytoma or glioblastoma is a highly invasive tumor that occur within the brain parenchyma. The Rho family of small GTPases, which includes Rac1, Cdc42, and RhoA, is an important family whose members are key regulators of the invasion and migration of glioblastoma cells. In this review, we describe the role played by the Rho family of GTPases in the regulation of the invasion and migration of glioblastoma cells. Specifically, we focus on the role played by RhoA, Rac1, RhoG, and Cdc42 in cell migration through rearrangement of actin cytoskeleton, cell adhesion, and invasion. Finally, we highlight the importance of potentially targeting Rho GTPases in the treatment of glioblastoma.
Collapse
|
13
|
Eser Ocak P, Ocak U, Tang J, Zhang JH. The role of caveolin-1 in tumors of the brain - functional and clinical implications. Cell Oncol (Dordr) 2019; 42:423-447. [PMID: 30993541 DOI: 10.1007/s13402-019-00447-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Caveolin-1 (cav-1) is the major structural protein of caveolae, the flask-shaped invaginations of the plasma membrane mainly involved in cell signaling. Today, cav-1 is believed to play a role in a variety of disease processes including cancer, owing to the variations of its expression in association with tumor progression, invasive behavior, metastasis and therapy resistance. Since first detected in the brain, a number of studies has particularly focused on the role of cav-1 in the various steps of brain tumorigenesis. In this review, we discuss the different roles of cav-1 and its contributions to the molecular mechanisms underlying the pathobiology and natural behavior of brain tumors including glial, non-glial and metastatic subtypes. These contributions could be attributed to its co-localization with important players in tumorigenesis within the lipid-enriched domains of the plasma membrane. In that regard, the ability of cav-1 to interact with various cell signaling molecules as well as the impact of caveolae depletion on important pathways acting in brain tumor pathogenesis are noteworthy. We also discuss conversant causes hampering the treatment of malignant glial tumors such as limited transport of chemotherapeutics across the blood tumor barrier and resistance to chemoradiotherapy, by focusing on the molecular fundamentals involving cav-1 participation. CONCLUSIONS Cav-1 has the potential to pivot the molecular basis underlying the pathobiology of brain tumors, particularly the malignant glial subtype. In addition, the regulatory effect of cav-1-dependent and caveola-mediated transcellular transport on the permeability of the blood tumor barrier could be of benefit to overcome the restricted transport across brain barriers when applying chemotherapeutics. The association of cav-1 with tumors of the brain other than malignant gliomas deserves to be underlined, as well given the evidence suggesting its potential in predicting tumor grade and recurrence rates together with determining patient prognosis in oligodendrogliomas, ependymomas, meningiomas, vestibular schwannomas and brain metastases.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
14
|
Desjarlais M, Annabi B. Dual functions of ARP101 in targeting membrane type-1 matrix metalloproteinase: Impact on U87 glioblastoma cell invasion and autophagy signaling. Chem Biol Drug Des 2018; 93:272-282. [PMID: 30291676 DOI: 10.1111/cbdd.13410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/24/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022]
Abstract
Membrane type-1 matrix metalloproteinase (MT1-MMP) possesses both extracellular proteolytic and intracellular signal-transducing functions in tumorigenesis. An imbalance in MT1-MMP expression and/or function triggers a metastatic, invasive, and therapy resistance phenotype. MT1-MMP is involved in extracellular matrix (ECM) proteolysis, activation of latent MMPs, as well as in autophagy signaling in human hepatoma and glioblastoma cells. A low autophagy index in tumorigenesis has been inferred by recent studies where autophagic capacity was decreased during tumor progression. Here, we establish ARP101 as a dual-function small-molecule inhibitor against MT1-MMP ECM hydrolysis and autophagy signal-transducing functions in a model of grade IV glioblastoma cells. ARP101 inhibited concanavalin-A-mediated proMMP-2 activation into MMP-2, as well as MT1-MMP auto-proteolytic processing. When overexpressing recombinant Wt MT1-MMP, ARP101 inhibited proMMP-2 activation and triggered the formation of MT1-MMP oligomers that required trafficking to the plasma membrane. ARP101 further induced cell autophagy as reflected by increased formation of acidic vacuole organelles, LC3 puncta, and autophagy-related protein ATG9 transcription. These were all significantly reversed upon siRNA-mediated gene silencing of MT1-MMP. ARP101 can thus concomitantly inhibit MT1-MMP extracellular catalytic function and exploit its intracellular transducing signal function to trigger autophagy-mediated cell death in U87 glioblastoma cancer cells.
Collapse
Affiliation(s)
- Michel Desjarlais
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre de recherche BIOMED, Université du Québec à Montréal, Montréal, Quebec, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre de recherche BIOMED, Université du Québec à Montréal, Montréal, Quebec, Canada
| |
Collapse
|
15
|
Le CT, Leenders WPJ, Molenaar RJ, van Noorden CJF. Effects of the Green Tea Polyphenol Epigallocatechin-3-Gallate on Glioma: A Critical Evaluation of the Literature. Nutr Cancer 2018; 70:317-333. [PMID: 29570984 DOI: 10.1080/01635581.2018.1446090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The review discusses the effects of Epigallocatechin-3-gallate Gallate (EGCG) on glioma as a basis for future research on clinical application of EGCG. Epidemiological studies on the effects of green tea or EGCG on the risk of glioma is inconclusive due to the limited number of studies, the inclusion of all tea types in these studies, and the focus on caffeine rather than EGCG. In vivo experiments using EGCG monotherapy are inconclusive. Nevertheless, EGCG induces cell death, prevents cellular proliferation, and limits invasion in multiple glioma cell lines. Furthermore, EGCG enhances the efficacy of anti-glioma therapies, including irradiation, temozolomide, carmustine, cisplatin, tamoxifen, and TNF-related apoptosis-inducing ligand, but reduces the effect of bortezomib. Pro-drugs, co-treatment, and encapsulation are being investigated to enhance clinical applicability of EGCG. Mechanisms of actions of EGCG have been partly elucidated. EGCG has both anti-oxidant and oxidant properties. EGCG inhibits pro-survival proteins, such as telomerase, survivin, GRP78, PEA15, and P-gp. EGCG inhibits signaling of PDGFR, IGF-1R, and 67LR. EGCG reduces invasiveness of cancer cells by inhibiting the activities of various metalloproteinases, cytokines, and chemokines. Last, EGCG inhibits some NADPH-producing enzymes, thus disturbing redox status and metabolism of glioma cells. In conclusion, EGCG may be a suitable adjuvant to potentiate anti-glioma therapies.
Collapse
Affiliation(s)
- Chung T Le
- a Department of Medical Biology , Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| | | | - Remco J Molenaar
- a Department of Medical Biology , Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| | - Cornelis J F van Noorden
- a Department of Medical Biology , Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| |
Collapse
|
16
|
Biophysical evidence for differential gallated green tea catechins binding to membrane type-1 matrix metalloproteinase and its interactors. Biophys Chem 2018; 234:34-41. [PMID: 29407769 DOI: 10.1016/j.bpc.2018.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
Membrane type-1 matrix metalloproteinase (MT1-MMP) is a transmembrane MMP which triggers intracellular signaling and regulates extracellular matrix proteolysis, two functions that are critical for tumor-associated angiogenesis and inflammation. While green tea catechins, particularly epigallocatechin gallate (EGCG), are considered very effective in preventing MT1-MMP-mediated functions, lack of structure-function studies and evidence regarding their direct interaction with MT1-MMP-mediated biological activities remain. Here, we assessed the impact in both cellular and biophysical assays of four ungallated catechins along with their gallated counterparts on MT1-MMP-mediated functions and molecular binding partners. Concanavalin-A (ConA) was used to trigger MT1-MMP-mediated proMMP-2 activation, expression of MT1-MMP and of endoplasmic reticulum stress biomarker GRP78 in U87 glioblastoma cells. We found that ConA-mediated MT1-MMP induction was inhibited by EGCG and catechin gallate (CG), that GRP78 induction was inhibited by EGCG, CG, and gallocatechin gallate (GCG), whereas proMMP-2 activation was inhibited by EGCG and GCG. Surface plasmon resonance was used to assess direct interaction between catechins and MT1-MMP interactors. We found that gallated catechins interacted better than their ungallated analogs with MT1-MMP as well as with MT1-MMP binding partners MMP-2, TIMP-2, MTCBP-1 and LRP1-clusterIV. Overall, current structure-function evidence supports a role for the galloyl moiety in both direct and indirect interactions of green tea catechins with MT1-MMP-mediated oncogenic processes.
Collapse
|
17
|
Sheehy S, Annabi B. A Transcriptional Regulatory Role for the Membrane Type-1 Matrix Metalloproteinase in Carcinogen-Induced Inflammasome Gene Expression. GENE REGULATION AND SYSTEMS BIOLOGY 2017. [PMID: 28634425 PMCID: PMC5467917 DOI: 10.1177/1177625017713996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Signal-transducing functions driven by the cytoplasmic domain of membrane type-1 matrix metalloproteinase (MT1-MMP) are believed to regulate many inflammation-associated cancer cell functions including migration, proliferation, and survival. Aside from upregulation of the inflammation biomarker cyclooxygenase-2 (COX-2) expression, MT1-MMP’s role in relaying intracellular signals triggered by extracellular pro-inflammatory cues remains poorly understood. Here, we triggered inflammation in HT1080 fibrosarcoma cells with phorbol-12-myristate-13-acetate (PMA), an inducer of COX-2 and of MT1-MMP. To assess the global transcriptional regulatory role that MT1-MMP may exert on inflammation biomarkers, we combined gene array screens with a transient MT1-MMP gene silencing strategy. Expression of MT1-MMP was found to exert both stimulatory and repressive transcriptional control of several inflammasome-related biomarkers such as interleukin (IL)-1B, IL-6, IL-12A, and IL-33, as well as of transcription factors such as EGR1, ELK1, and ETS1/2 in PMA-treated cells. Among the signal-transducing pathways explored, the silencing of MT1-MMP prevented PMA from phosphorylating extracellular signal–regulated kinase, inhibitor of κB, and p105 nuclear factor κB (NF-κB) intermediates. We also found a signaling axis linking MT1-MMP to MMP-9 transcriptional regulation. Altogether, our data indicate a significant involvement of MT1-MMP in the transcriptional regulation of inflammatory biomarkers consolidating its contribution to signal transduction functions in addition to its classical hydrolytic activity.
Collapse
Affiliation(s)
- Samuel Sheehy
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre de recherche BIOMED, Université du Québec à Montréal, Montréal, QC, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre de recherche BIOMED, Université du Québec à Montréal, Montréal, QC, Canada
| |
Collapse
|
18
|
Berardinelli F, Coluzzi E, Sgura A, Antoccia A. Targeting telomerase and telomeres to enhance ionizing radiation effects in in vitro and in vivo cancer models. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:204-219. [PMID: 28927529 DOI: 10.1016/j.mrrev.2017.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 01/05/2023]
Abstract
One of the hallmarks of cancer consists in the ability of tumor cells to divide indefinitely, and to maintain stable telomere lengths throughout the activation of specific telomere maintenance mechanisms (TMM). Therefore in the last fifteen years, researchers proposed to target telomerase or telomeric structure in order to block limitless replicative potential of cancer cells providing a fascinating strategy for a broad-spectrum cancer therapy. In the present review, we report in vitro and in vivo evidence regarding the use of chemical agents targeting both telomerase or telomere structure and showing promising antitumor effects when used in combination with ionizing radiation (IR). RNA interference, antisense oligonucleotides (e.g., GRN163L), non-nucleoside inhibitors (e.g., BIBR1532) and nucleoside analogs (e.g., AZT) represent some of the most potent strategies to inhibit telomerase activity used in combination with IR. Furthermore, radiosensitizing effects were demonstrated also for agents acting directly on the telomeric structure such as G4-ligands (e.g., RHPS4 and Telomestatin) or telomeric-oligos (T-oligos). To date, some of these compounds are under clinical evaluation (e.g., GRN163L and KML001). Advantages of Telomere/Telomerase Targeting Compounds (T/TTCs) coupled with radiotherapy may be relevant in the treatment of radioresistant tumors and in the development of new optimized treatment plans with reduced dose adsorbed by patients and consequent attenuation of short- end long-term side effects. Pros and cons of possible future applications in cancer therapy based on the combination of T/TCCs and radiation treatment are discussed.
Collapse
Affiliation(s)
- F Berardinelli
- Dipartimento di Scienze, Università Roma Tre, Rome Italy; Istituto Nazionale di Fisica Nucleare, INFN, Sezione di Roma Tre, Rome, Italy.
| | - E Coluzzi
- Dipartimento di Scienze, Università Roma Tre, Rome Italy
| | - A Sgura
- Dipartimento di Scienze, Università Roma Tre, Rome Italy; Istituto Nazionale di Fisica Nucleare, INFN, Sezione di Roma Tre, Rome, Italy
| | - A Antoccia
- Dipartimento di Scienze, Università Roma Tre, Rome Italy; Istituto Nazionale di Fisica Nucleare, INFN, Sezione di Roma Tre, Rome, Italy
| |
Collapse
|
19
|
Nanni SB, Pratt J, Beauchemin D, Haidara K, Annabi B. Impact of Concanavalin-A-Mediated Cytoskeleton Disruption on Low-Density Lipoprotein Receptor-Related Protein-1 Internalization and Cell Surface Expression in Glioblastomas. BIOMARKERS IN CANCER 2016; 8:77-87. [PMID: 27226736 PMCID: PMC4874747 DOI: 10.4137/bic.s38894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/17/2016] [Accepted: 04/26/2016] [Indexed: 01/13/2023]
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP-1) is a multiligand endocytic receptor, which plays a pivotal role in controlling cytoskeleton dynamics during cancer cell migration. Its rapid endocytosis further allows efficient clearance of extracellular ligands. Concanavalin-A (ConA) is a lectin used to trigger in vitro physiological cellular processes, including cytokines secretion, nitric oxide production, and T-lymphocytes activation. Given that ConA exerts part of its effects through cytoskeleton remodeling, we questioned whether it affected LRP-1 expression, intracellular trafficking, and cell surface function in grade IV U87 glioblastoma cells. Using flow cytometry and confocal microscopy, we found that loss of the cell surface 600-kDa mature form of LRP-1 occurs upon ConA treatment. Consequently, internalization of the physiological α2-macroglobulin and the synthetic angiopep-2 ligands of LRP-1 was also decreased. Silencing of known mediators of ConA, such as the membrane type-1 matrix metalloproteinase, and the Toll-like receptors (TLR)-2 and TLR-6 was unable to rescue ConA-mediated LRP-1 expression decrease, implying that the loss of LRP-1 was independent of cell surface relayed signaling. The ConA-mediated reduction in LRP-1 expression was emulated by the actin cytoskeleton-disrupting agent cytochalasin-D, but not by the microtubule inhibitor nocodazole, and required both lysosomal- and ubiquitin-proteasome system-mediated degradation. Our study implies that actin cytoskeleton integrity is required for proper LRP-1 cell surface functions and that impaired trafficking leads to specialized compartmentation and degradation. Our data also strengthen the biomarker role of cell surface LRP-1 functions in the vectorized transport of therapeutic angiopep bioconjugates into brain cancer cells.
Collapse
Affiliation(s)
- Samuel Burke Nanni
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, QC, Canada
| | - Jonathan Pratt
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, QC, Canada
| | - David Beauchemin
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, QC, Canada
| | - Khadidja Haidara
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, QC, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, QC, Canada
| |
Collapse
|
20
|
MacKay JL, Kumar S. Simultaneous and independent tuning of RhoA and Rac1 activity with orthogonally inducible promoters. Integr Biol (Camb) 2015; 6:885-94. [PMID: 25044255 DOI: 10.1039/c4ib00099d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The GTPases RhoA and Rac1 are key regulators of cell spreading, adhesion, and migration, and they exert distinct effects on the actin cytoskeleton. While RhoA classically stimulates stress fiber assembly and contraction, Rac1 promotes branched actin polymerization and membrane protrusion. These competing influences are reinforced by antagonistic crosstalk between RhoA and Rac1, which has complicated efforts to identify the specific mechanisms by which each GTPase regulates cell behavior. We therefore wondered whether RhoA and Rac1 are intrinsically coupled or whether they can be manipulated independently. To address this question, we placed constitutively active (CA) RhoA under a doxycycline-inducible promoter and CA Rac1 under an orthogonal cumate-inducible promoter, and we stably introduced both constructs into glioblastoma cells. We found that doxycycline addition increased RhoA activity without altering Rac1, and similarly cumate addition increased Rac1 activity without altering RhoA. Furthermore, co-expression of both mutants enabled high activation of RhoA and Rac1 simultaneously. When cells were cultured on collagen hydrogels, RhoA activation prevented cell spreading and motility, whereas Rac1 activation stimulated migration and dynamic cell protrusions. Interestingly, high activation of both GTPases induced a third phenotype, in which cells migrated at intermediate speeds similar to control cells but also aggregated into large, contractile clusters. In addition, we demonstrate dynamic and reversible switching between high RhoA and high Rac1 phenotypes. Overall, this approach represents a unique way to access different combinations of RhoA and Rac1 activity levels in a single cell and may serve as a valuable tool for multiplexed dissection and control of mechanobiological signals.
Collapse
Affiliation(s)
- Joanna L MacKay
- Department of Chemical and Biomolecular Engineering, University of California-Berkeley, Berkeley, California 94720, USA
| | | |
Collapse
|
21
|
Gundimeda U, McNeill TH, Barseghian BA, Tzeng WS, Rayudu DV, Cadenas E, Gopalakrishna R. Polyphenols from green tea prevent antineuritogenic action of Nogo-A via 67-kDa laminin receptor and hydrogen peroxide. J Neurochem 2015; 132:70-84. [PMID: 25314656 DOI: 10.1111/jnc.12964] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/22/2014] [Accepted: 10/06/2014] [Indexed: 02/02/2023]
Abstract
Axonal regeneration after injury to the CNS is hampered by myelin-derived inhibitors, such as Nogo-A. Natural products, such as green tea, which are neuroprotective and safe for long-term therapy, would complement ongoing various pharmacological approaches. In this study, using nerve growth factor-differentiated neuronal-like Neuroscreen-1 cells, we show that extremely low concentrations of unfractionated green tea polyphenol mixture (GTPP) and its active ingredient, epigallocatechin-3-gallate (EGCG), prevent both the neurite outgrowth-inhibiting activity and growth cone-collapsing activity of Nogo-66 (C-terminal domain of Nogo-A). Furthermore, a synergistic interaction was observed among GTPP constituents. This preventive effect was dependent on 67-kDa laminin receptor (67LR) to which EGCG binds with high affinity. The antioxidants N-acetylcysteine and cell-permeable catalase abolished this preventive effect of GTPP and EGCG, suggesting the involvement of sublethal levels of H2 O2 in this process. Accordingly, exogenous sublethal concentrations of H2 O2 , added as a bolus dose (5 μM) or more effectively through a steady-state generation (1-2 μM), mimicked GTPP in counteracting the action of Nogo-66. Exogenous H2 O2 mediated this action by bypassing the requirement of 67LR. Taken together, these results show for the first time that GTPP and EGCG, acting through 67LR and elevating intracellular sublethal levels of H2 O2 , inhibit the antineuritogenic action of Nogo-A. Currently, several agents are being evaluated for overcoming axonal growth inhibitors to promote functional recovery after stroke and spinal cord injury. Epigallocatechin-3-gallate (EGCG), present in green tea polyphenol mixture (GTPP), prevents antineuritogenic activity of Nogo-A, a myelin-derived axonal growth inhibitor. The preventive action of EGCG involves the cell-surface-associated 67-kDa laminin receptor and H2 O2 . GTPP may complement ongoing efforts to treat neuronal injuries.>
Collapse
Affiliation(s)
- Usha Gundimeda
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Pratt J, Iddir M, Bourgault S, Annabi B. Evidence of MTCBP-1 interaction with the cytoplasmic domain of MT1-MMP: Implications in the autophagy cell index of high-grade glioblastoma. Mol Carcinog 2015; 55:148-60. [PMID: 25640948 DOI: 10.1002/mc.22264] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/17/2014] [Indexed: 01/01/2023]
Abstract
Progression of astrocytic tumors is, in part, related to their dysregulated autophagy capacity. Recent evidence indicates that upstream autophagy signaling events can be triggered by MT1-MMP, a membrane-bound matrix metalloproteinase that contributes to the invasive phenotype of brain cancer cells. The signaling functions of MT1-MMP require its intracellular domain, and recent identification of MTCBP-1, a cytoplasmic 19 kDa protein involved in the inhibition of MT1-MMP-mediated cell migration, suggests that modulation of MT1-MMP cytoplasmic domain-mediated signaling may affect other carcinogenic processes. Using qPCR and screening of cDNA generated from brain tumor tissues of grades I, II, III, and IV, MT1-MMP gene expression was found to correlate with increased grade of tumors. Inversely, MTCBP-1 expression decreased with increasing grade of brain tumor. Confocal microscopy and fluorescence resonance energy transfer (FRET) analysis revealed that overexpressing a cytoplasmic-deleted MT1-MMP recombinant protein mutant prevented MTCBP-1 recruitment to the intracellular leaf of plasma membrane in U87 glioblastoma cells. The interaction between MTCBP-1 and the 20 amino acids peptide representing the MT1-MMP cytoplasmic domain was confirmed by surface plasmon resonance. Overexpression of a full-length Wt-MT1-MMP triggered acidic autophagy vesicle formation and autophagic puncta formation for green fluorescent microtubule-associated protein 1 light chain 3 (GFP-LC3). Autophagic vesicles and GFP-LC3 puncta formation were abrogated in the presence of MTCBP-1. Our data elucidate a new role for MTCBP-1 regulating the intracellular function of MT1-MMP-mediated autophagy. The inverse correlation between MTCBP-1 and MT1-MMP expression with brain tumor grades could also contribute to the decreased autophagic index observed in high-grade tumors.
Collapse
Affiliation(s)
- Jonathan Pratt
- Laboratoire d'Oncologie Moléculaire, Centre de recherche Biomed, Quebec, Canada
| | - Mustapha Iddir
- Laboratoire d'Oncologie Moléculaire, Centre de recherche Biomed, Quebec, Canada
| | - Steve Bourgault
- Centre de recherche Pharmaqam, Département de Chimie, Université du Québec à Montréal, Quebec, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Centre de recherche Biomed, Quebec, Canada
| |
Collapse
|
23
|
Gupta R, Toufaily C, Annabi B. Caveolin and cavin family members: dual roles in cancer. Biochimie 2014; 107 Pt B:188-202. [PMID: 25241255 DOI: 10.1016/j.biochi.2014.09.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 09/04/2014] [Indexed: 12/16/2022]
Abstract
Caveolae are specialized plasma membrane subdomains with distinct lipid and protein compositions, which play an essential role in cell physiology through regulation of trafficking and signaling functions. The structure and functions of caveolae have been shown to require the proteins caveolins. Recently, members of the cavin protein family were found to be required, in concert with caveolins, for the formation and function of caveolae. Caveolins have a paradoxical role in the development of cancer formation. They have been involved in both tumor suppression and oncogenesis, depending on tumor type and progress stage. High expression of caveolins and cavins leads to inhibition of cancer-related pathways, such as growth factor signaling pathways. However, certain cancer cells that express caveolins and cavins have been shown to be more aggressive and metastatic because of their increased potential for anchorage-independent growth. Here, we will survey the functional roles of caveolins and of different cavin family members in cancer regulation.
Collapse
Affiliation(s)
- Reshu Gupta
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMed, Département de Chimie, Université du Québec à Montréal, Québec H3C 3P8, Canada.
| | - Chirine Toufaily
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMed, Département de Chimie, Université du Québec à Montréal, Québec H3C 3P8, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMed, Département de Chimie, Université du Québec à Montréal, Québec H3C 3P8, Canada
| |
Collapse
|
24
|
Li Z, Ha J, Zou T, Gu L. Fabrication of coated bovine serum albumin (BSA)-epigallocatechin gallate (EGCG) nanoparticles and their transport across monolayers of human intestinal epithelial Caco-2 cells. Food Funct 2014; 5:1278-85. [PMID: 24741679 DOI: 10.1039/c3fo60500k] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The bovine serum albumin (BSA)-epigallocatechin gallate (EGCG) nanoparticles were fabricated using a desolvation method, and coated with poly-ε-lysine or chitosan. BSA-EGCG nanoparticles (BEN), poly-ε-lysine coated BSA-EGCG nanoparticles (PBEN), and chitosan coated BSA-EGCG nanoparticles (CBEN) had a spherical morphology and a size of 186, 259, and 300 nm, respectively. The loading efficiency of EGCG in these nanoparticles was 32.3%, 35.4%, and 32.7%, whereas the loading capacity was 18.9%, 17.0%, and 16.0% (w/w), respectively. Poly-ε-lysine or chitosan coating prevented the aggregation of nanoparticles at pH 4.5-5.0. However, they caused particle aggregation at pH 6.5-7.0. BEN had negative zeta-potentials between pH 4.5 and 6.0. Poly-ε-lysine or chitosan coating changed the zeta-potentials to positive. The release study of EGCG from the nanoparticles in the simulated gastric or intestinal fluid with or without digestive enzymes showed that poly-ε-lysine and chitosan coatings delayed EGCG release from the nanoparticles. Poly-ε-lysine or chitosan coating improved the stability of EGCG during storage at 60 °C compared with EGCG in the uncoated particles. EGCG in BEN, PBEN, and CBEN had a decreasing apparent permeability coefficient (Papp) on Caco-2 monolayers, whereas pure EGCG showed relatively stable Papp during the incubation over time. EGCG in CBEN showed significantly higher Papp, suggesting that chitosan coated BSA-EGCG nanoparticles may improve the absorption of EGCG.
Collapse
Affiliation(s)
- Zheng Li
- Food Science and Human Nutrition Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, Florida 32611, USA.
| | | | | | | |
Collapse
|
25
|
Li Z, Gu L. Fabrication of self-assembled (-)-epigallocatechin gallate (EGCG) ovalbumin-dextran conjugate nanoparticles and their transport across monolayers of human intestinal epithelial Caco-2 cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:1301-1309. [PMID: 24446922 DOI: 10.1021/jf404621f] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Nanoparticles have the potential to increase bioavailability of nutraceutical compounds such as (-)-epigallocatechin gallate (EGCG). Ovalbumin was conjugated with dextran using the Maillard reaction. The resultant ovalbumin-dextran (O-D) conjugates were self-assembled with EGCG to form EGCG O-D conjugate nanoparticles at pH 5.2 after heating at 80 °C for 60 min. Ovalbumin in EGCG O-D conjugate nanoparticles was further cross-linked by glutaraldehyde for 24 h at room temperature. EGCG O-D conjugate nanoparticles and cross-linked EGCG O-D conjugate nanoparticles in aqueous suspension had particle sizes of 285 and 339 nm, respectively, and showed a spherical morphology. The loading efficiencies of EGCG in these two nanoparticles were 23.4 and 30.0%, whereas the loading capacities were 19.6 and 20.9%, respectively. These nanoparticles showed positive zeta-potentials in a pH range from 2.5 to 4.0 but had negative charges at pH ≥5.0. EGCG O-D conjugate nanoparticles maintained a particle size of 183-349 nm in simulated gastric fluid (SGF) and 188-291 nm in simulated intestinal fluid (SIF) at 37 °C for 2 h, whereas cross-linked nanoparticles had particle sizes of 294-527 nm in SGF and 206-300 nm in SIF. Limited release of EGCG was observed in both nanoparticle systems in simulated gastric and intestinal fluids without and with digestive enzymes. EGCG O-D conjugate nanoparticles significantly enhanced the apparent permeability coefficient (Papp) of EGCG on Caco-2 monolayers compared with EGCG solution, suggesting that these nanoparticles may improve the absorption of EGCG.
Collapse
Affiliation(s)
- Zheng Li
- Food Science and Human Nutrition Department, Institute of Food and Agricultural Sciences, University of Florida , Gainesville, Florida 32611, United States
| | | |
Collapse
|
26
|
Induction of autophagy biomarker BNIP3 requires a JAK2/STAT3 and MT1-MMP signaling interplay in Concanavalin-A-activated U87 glioblastoma cells. Cell Signal 2014; 26:917-24. [PMID: 24462646 DOI: 10.1016/j.cellsig.2014.01.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 01/13/2014] [Indexed: 11/20/2022]
Abstract
Plant lectins have been considered as possible anti-tumor drugs because of their property to induce autophagic cell death. Given that expression of membrane type-1 matrix metalloproteinase (MT1-MMP) has been found to regulate expression of the autophagy biomarker Bcl-2/adenovirus E1B 19kDa interacting protein 3 (BNIP3), we sought to investigate possible signaling interplay mechanisms between MT1-MMP and BNIP3 in Concanavalin-A (ConA) lectin-activated U87 glioblastoma cells. ConA induced acidic vacuole organelle formation as well as BNIP3 and MT1-MMP gene and protein expressions, whereas only BNIP3 expression was dose-dependently inhibited by the JAK2 tyrosine kinase inhibitor AG490 suggesting a requirement for some STAT-mediated signaling. Gene silencing of MT1-MMP and of STAT3 abrogated ConA-induced STAT3 phosphorylation and BNIP3 expression. Correlative analysis shows that STAT3 signaling events occur downstream from MT1-MMP induction. Overexpression of a full length MT1-MMP recombinant protein led to increased BNIP3 gene and protein expressions. The cytoplasmic domain of MT1-MMP was also found necessary for transducing STAT3 phosphorylation. Among JAK1, JAK2, JAK3, and TYK2, only JAK2 gene silencing abrogated ConA's effects on MT1-MMP and BNIP3 gene and protein expressions. Our study elucidates how MT1-MMP signals autophagy, a process which could contribute to the chemoresistance phenotype in brain cancer cells.
Collapse
|
27
|
Akla N, Pratt J, Annabi B. Concanavalin-A triggers inflammatory response through JAK/STAT3 signalling and modulates MT1-MMP regulation of COX-2 in mesenchymal stromal cells. Exp Cell Res 2012; 318:2498-506. [PMID: 22971618 DOI: 10.1016/j.yexcr.2012.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/02/2012] [Accepted: 08/20/2012] [Indexed: 12/22/2022]
Abstract
Pharmacological targeting of inflammation through STAT3 and NF-κB signaling pathways is, among other inflammatory biomarkers, associated with cyclooxygenase (COX)-2 inhibition and is believed to play a crucial role in prevention and therapy of cancer. Recently, inflammatory factors were found to impact on mesenchymal stromal cells (MSC) contribution to tumor angiogenesis. Given MSC chemotaxis and cell survival are regulated, in part, by the membrane type-1 matrix metalloproteinase (MT1-MMP), an MMP also involved in transducing NF-κB intracellular signaling pathways, we tested whether STAT3 regulation by MT1-MMP may also contribute to the expression balance of COX-2 in MSC. We demonstrate that STAT3 phosphorylation was triggered in MSC treated with the MT1-MMP inducer lectin Concanavalin-A (ConA), and that this phosphorylation was abrogated by the JAK2 inhibitor AG490. MT1-MMP gene silencing significantly inhibited ConA-induced STAT3 phosphorylation and this was correlated with reduced proMMP-2 activation and COX-2 expression. On the other hand, STAT3 gene silencing potentiated ConA-induced COX-2 expression, providing evidence for a new MT1-MMP/JAK/STAT3 signaling axis that may, in part, explain how MT1-MMP contributes to proinflammatory intracellular signaling. Given that MSC are avidly recruited within inflammatory microenvironments and within experimental vascularizing tumors, these mechanistic observations support a possible dual control of cell adaptation to inflammation by MT1-MMP and that may enable MSC to be active participants within inflamed tissues.
Collapse
Affiliation(s)
- Naoufal Akla
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, Quebec, Canada H3C 3P8
| | | | | |
Collapse
|
28
|
KUNG CHANGI, CHEN CHENGYI, YANG CHIHCHANG, LIN CHENGYU, CHEN TIENHUA, WANG HWAISHI. Enhanced membrane-type 1 matrix metalloproteinase expression by hyaluronan oligosaccharides in breast cancer cells facilitates CD44 cleavage and tumor cell migration. Oncol Rep 2012; 28:1808-14. [DOI: 10.3892/or.2012.1993] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/03/2012] [Indexed: 11/06/2022] Open
|
29
|
MT1-MMP expression level status dictates the in vitro action of lupeol on inflammatory biomarkers MMP-9 and COX-2 in medulloblastoma cells. Inflammopharmacology 2012; 21:91-9. [PMID: 22707305 DOI: 10.1007/s10787-012-0142-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 05/28/2012] [Indexed: 10/28/2022]
|
30
|
Pratt J, Roy R, Annabi B. Concanavalin-A-induced autophagy biomarkers requires membrane type-1 matrix metalloproteinase intracellular signaling in glioblastoma cells. Glycobiology 2012; 22:1245-55. [PMID: 22692046 DOI: 10.1093/glycob/cws093] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pre-clinical trials for cancer therapeutics support the anti-neoplastic properties of the lectin from Canavalia ensiformis (Concanavalin-A, ConA) in targeting apoptosis and autophagy in a variety of cancer cells. Given that membrane type-1 matrix metalloproteinase (MT1-MMP), a plasma membrane-anchored matrix metalloproteinase, is a glycoprotein strongly expressed in radioresistant and chemoresistant glioblastoma that mediates pro-apoptotic signalling in brain cancer cells, we investigated whether MT1-MMP could also signal autophagy. Among the four lectins tested, we found that the mannopyranoside/glucopyranoside-binding ConA, which is also well documented to trigger MT1-MMP expression, increases autophagic acidic vacuoles formation as demonstrated by Acridine Orange cell staining. Although siRNA-mediated MT1-MMP gene silencing effectively reversed ConA-induced autophagy, inhibition of the MT1-MMP extracellular catalytic function with Actinonin or Ilomastat did not. Conversely, direct overexpression of the recombinant Wt-MT1-MMP protein triggered proMMP-2 activation and green fluorescent protein-microtubule-associated protein light chain 3 puncta indicative of autophagosomes formation, while deletion of MT1-MMP's cytoplasmic domain disabled such autophagy induction. ConA-treated U87 cells also showed an upregulation of BNIP3 and of autophagy-related gene members autophagy-related protein 3, autophagy-related protein 12 and autophagy-related protein 16-like 1, where respective inductions were reversed when MT1-MMP gene expression was silenced. Altogether, we provide molecular evidence supporting the pro-autophagic mechanism of action of ConA in glioblastoma cells. We also highlight new signal transduction functions of MT1-MMP within apoptotic and autophagic pathways that often characterize cancer cell responses to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jonathan Pratt
- Laboratoire d'Oncologie Moléculaire, Centre de Recherche BioMED, Québec, Canada
| | | | | |
Collapse
|
31
|
Sina A, Lord-Dufour S, Roy R, Annabi B. Ciblage pharmacologique de la MT1-MMP dans les cellules tumorales cérébrales par l’actinonine, un inhibiteur de l’aminopeptidase N/CD13. BIO TRIBUNE MAGAZINE 2011; 38:39-45. [DOI: 10.1007/s11834-011-0042-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
|
32
|
Kim W, Seong KM, Youn B. Phenylpropanoids in radioregulation: double edged sword. Exp Mol Med 2011; 43:323-33. [PMID: 21483230 PMCID: PMC3128910 DOI: 10.3858/emm.2011.43.6.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2011] [Indexed: 11/04/2022] Open
Abstract
Radiotherapy, frequently used for treatment of solid tumors, carries two main obstacles including acquired radioresistance in cancer cells during radiotherapy and normal tissue injury. Phenylpropanoids, which are naturally occurring phytochemicals found in plants, have been identified as potential radiotherapeutic agents due to their anti-cancer activity and relatively safe levels of cytotoxicity. Various studies have proposed that these compounds could not only sensitize cancer cells to radiation resulting in inhibition of growth and cell death but also protect normal cells against radiation-induced damage. This review is intended to provide an overview of recent investigations on the usage of phenylpropanoids in combination with radiotherapy in cancer treatment.
Collapse
Affiliation(s)
- Wanyeon Kim
- College of Natural Sciences Department of Biological Sciences Pusan National University Busan 609-735, Korea
| | | | | |
Collapse
|
33
|
Proulx-Bonneau S, Guezguez A, Annabi B. A concerted HIF-1α/MT1-MMP signalling axis regulates the expression of the 3BP2 adaptor protein in hypoxic mesenchymal stromal cells. PLoS One 2011; 6:e21511. [PMID: 21738685 PMCID: PMC3124525 DOI: 10.1371/journal.pone.0021511] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/31/2011] [Indexed: 01/01/2023] Open
Abstract
Increased plasticity, migratory and immunosuppressive abilities characterize mesenchymal stromal cells (MSC) which enable them to be active participants in the development of hypoxic solid tumours. Our understanding of the oncogenic adaptation of MSC to hypoxia however lacks the identification and characterization of specific biomarkers. In this study, we assessed the hypoxic regulation of 3BP2/SH3BP2 (Abl SH3-binding protein 2), an immune response adaptor/scaffold protein which regulates leukocyte differentiation and motility. Gene silencing of 3BP2 abrogated MSC migration in response to hypoxic cues and generation of MSC stably expressing the transcription factor hypoxia inducible factor 1alpha (HIF-1α) resulted in increased endogenous 3BP2 expression as well as cell migration. Analysis of the 3BP2 promoter sequence revealed only one potential HIF-1α binding site within the human but none in the murine sequence. An alternate early signalling cascade that regulated 3BP2 expression was found to involve membrane type-1 matrix metalloproteinase (MT1-MMP) transcriptional regulation which gene silencing abrogated 3BP2 expression in response to hypoxia. Collectively, we provide evidence for a concerted HIF-1α/MT1-MMP signalling axis that explains the induction of adaptor protein 3BP2 and which may link protein binding partners together and stimulate oncogenic MSC migration. These mechanistic observations support the potential for malignant transformation of MSC within hypoxic tumour stroma and may contribute to evasion of the immune system by a tumour.
Collapse
Affiliation(s)
- Sébastien Proulx-Bonneau
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, Quebec, Canada
| | - Amel Guezguez
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, Quebec, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Centre de recherche BIOMED, Département de Chimie, Université du Québec à Montréal, Quebec, Canada
- * E-mail:
| |
Collapse
|
34
|
Fluid shear stress regulates the invasive potential of glioma cells via modulation of migratory activity and matrix metalloproteinase expression. PLoS One 2011; 6:e20348. [PMID: 21637818 PMCID: PMC3102715 DOI: 10.1371/journal.pone.0020348] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 04/30/2011] [Indexed: 01/15/2023] Open
Abstract
Background Glioma cells are exposed to elevated interstitial fluid flow during the onset of angiogenesis, at the tumor periphery while invading normal parenchyma, within white matter tracts, and during vascular normalization therapy. Glioma cell lines that have been exposed to fluid flow forces in vivo have much lower invasive potentials than in vitro cell motility assays without flow would indicate. Methodology/Principal Findings A 3D Modified Boyden chamber (Darcy flow through collagen/cell suspension) model was designed to mimic the fluid dynamic microenvironment to study the effects of fluid shear stress on the migratory activity of glioma cells. Novel methods for gel compaction and isolation of chemotactic migration from flow stimulation were utilized for three glioma cell lines: U87, CNS-1, and U251. All physiologic levels of fluid shear stress suppressed the migratory activity of U87 and CNS-1 cell lines. U251 motility remained unaltered within the 3D interstitial flow model. Matrix Metalloproteinase (MMP) inhibition experiments and assays demonstrated that the glioma cells depended on MMP activity to invade, and suppression in motility correlated with downregulation of MMP-1 and MMP-2 levels. This was confirmed by RT-PCR and with the aid of MMP-1 and MMP-2 shRNA constructs. Conclusions/Significance Fluid shear stress in the tumor microenvironment may explain reduced glioma invasion through modulation of cell motility and MMP levels. The flow-induced migration trends were consistent with reported invasive potentials of implanted gliomas. The models developed for this study imply that flow-modulated motility involves mechanotransduction of fluid shear stress affecting MMP activation and expression. These models should be useful for the continued study of interstitial flow effects on processes that affect tumor progression.
Collapse
|
35
|
Chen Y, Lee YD, Vedala H, Allen BL, Star A. Exploring the chemical sensitivity of a carbon nanotube/green tea composite. ACS NANO 2010; 4:6854-62. [PMID: 21043457 PMCID: PMC3026703 DOI: 10.1021/nn100988t] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Single-walled carbon nanotubes (SWNTs) possess unique electronic and physical properties, which make them very attractive for a wide range of applications. In particular, SWNTs and their composites have shown a great potential for chemical and biological sensing. Green tea, or more specifically its main antioxidant component, epigallocatechin gallate (EGCG), has been found to disperse SWNTs in water. However, the chemical sensitivity of this SWNT/green tea (SWNT/EGCG) composite remained unexplored. With EGCG present, this SWNT composite should have strong antioxidant properties and thus respond to reactive oxygen species (ROS). Here we report on fabrication and characterization of SWNT/EGCG thin films and the measurement of their relative conductance as a function of H(2)O(2) concentrations. We further investigated the sensing mechanism by Fourier transform infrared (FTIR) spectroscopy and field-effect transistor measurements (FET). We propose here that the response to H(2)O(2) arises from the oxidation of EGCG in the composite. These findings suggest that SWNT/green tea composite has a great potential for developing simple resistivity-based sensors.
Collapse
Affiliation(s)
| | | | | | | | - Alexander Star
- Department of Chemistry, University of Pittsburgh and the National Energy Technology Laboratory, Pittsburgh, PA 15260 (USA), Fax: (+1) 412-624-4027
| |
Collapse
|
36
|
A role for MT1-MMP as a cell death sensor/effector through the regulation of endoplasmic reticulum stress in U87 glioblastoma cells. J Neurooncol 2010; 104:33-43. [PMID: 21088866 DOI: 10.1007/s11060-010-0468-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 11/08/2010] [Indexed: 12/30/2022]
|
37
|
Wiranowska M, Ladd S, Moscinski LC, Hill B, Haller E, Mikecz K, Plaas A. Modulation of hyaluronan production by CD44 positive glioma cells. Int J Cancer 2010; 127:532-42. [PMID: 19957333 PMCID: PMC3962665 DOI: 10.1002/ijc.25085] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study examines the functional relationship between glioma cell production of hyaluronan (HA), known to play a role in glioma invasion, expression of its CD44 receptor, and glioma cell viability. Production of HA by CD44 positive mouse G26 and human U373 glioma cell lines was evaluated and compared to that of a CD44 positive mouse fibroblast-like L929 cell line. We found that both G26 and U373 MG glioma cells, but not L929 fibroblast-like cells, synthesized HA. The synthesis of HA by glioma cells was found during the proliferative phase as well as post-confluency, as detected by fluorophore-assisted carbohydrate electrophoresis. Eighty to ninety percent of the HA synthesized was secreted into the medium and 10-20% remained associated with the cells. To examine a possible mechanistic link between the CD44-HA interaction and endogenous HA production, glioma cells were treated with either anti-CD44 antibodies (clones KM201 or IM7) or HA oligosaccharides (hexamer oligoHA-6 or decamer oligoHA-10). We found that oligoHA-10, which was previously shown to compete effectively with the CD44-HA interaction, enhanced glioma HA synthesis by approximately 1.5-fold, without affecting cell viability. IM7 treatment of human U373 glioma cells resulted in over 50% decrease of HA production, which was associated with changes in cell size and apoptosis. Taken together, these data show that CD44 specific ligands, such as the IM7 antibody or oligoHA-10 could down-regulate or up-regulate glioma HA production, respectively. Our results suggest that interference with CD44/HA may lead to the discovery and development of new treatment modalities for glioma.
Collapse
Affiliation(s)
- Marzenna Wiranowska
- Department of Pathology and Cell Biology, University of South Florida, College of Medicine, Tampa, FL 33612-4799, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Annabi B, Lachambre MP, Plouffe K, Sartelet H, Béliveau R. Modulation of invasive properties of CD133(+) glioblastoma stem cells: A role for MT1-MMP in bioactive lysophospholipid signaling. Mol Carcinog 2009; 48:910-9. [PMID: 19326372 DOI: 10.1002/mc.20541] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
39
|
Sina A, Lord-Dufour S, Annabi B. Cell-based evidence for aminopeptidase N/CD13 inhibitor actinonin targeting of MT1-MMP-mediated proMMP-2 activation. Cancer Lett 2009; 279:171-6. [PMID: 19264392 DOI: 10.1016/j.canlet.2009.01.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 12/19/2008] [Accepted: 01/22/2009] [Indexed: 11/16/2022]
|
40
|
Kuo YC, Su CH, Liu CY, Chen TH, Chen CP, Wang HS. Transforming growth factor-beta induces CD44 cleavage that promotes migration of MDA-MB-435s cells through the up-regulation of membrane type 1-matrix metalloproteinase. Int J Cancer 2009; 124:2568-76. [PMID: 19243022 DOI: 10.1002/ijc.24263] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
CD44, a transmembrane receptor for hyaluronic acid, is implicated in various adhesion-dependent cellular processes, including cell migration, tumor cell metastasis and invasion. Recent studies demonstrated that CD44 expressed in cancer cells can be proteolytically cleaved at the ectodomain by membrane type 1-matrix metalloproteinase (MT1-MMP) to form soluble CD44 and that CD44 cleavage plays a critical role in cancer cell migration. Here, we show that transforming growth factor-beta (TGF-beta), a multifunctional cytokine involved in cell proliferation, differentiation, migration and pathological processes, induces MT1-MMP expression in MDA-MB-435s cells. TGF-beta-induced MT1-MMP expression was blocked by the specific extracellular regulated kinase-1/2 (ERK1/2) inhibitor PD98059 and the specific phosphoinositide 3-OH kinase (PI3K) inhibitor LY294002. In addition, treatment with SP600125, an inhibitor for c-Jun NH(2)-terminal kinase (JNK), resulted in a significant inhibition of MT1-MMP production. These data suggest that ERK1/2, PI3K, and JNK likely play a role in TGF-beta-induced MT1-MMP expression. Interestingly, treatment of MDA-MB-435s cells with TGF-beta resulted in a colocalization of MT1-MMP and CD44 in the cell membrane and in an increased level of soluble CD44. Using an electric cell-substrate impedance sensing cell-electrode system, we demonstrated that TGF-beta treatment promotes MDA-MB-435s cell migration, involving MT1-MMP-mediated CD44 cleavage. MT1-MMP siRNA transfection-inhibited TGF-beta-induced cancer cell transendothelial migration. Thus, this study contributes to our understanding of molecular mechanisms that play a critical role in tumor cell invasion and metastasis.
Collapse
Affiliation(s)
- Yi-Chih Kuo
- Institute of Anatomy and Cell Biology, School of Medicine, Yang Ming University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Annabi B, Rojas-Sutterlin S, Laflamme C, Lachambre MP, Rolland Y, Sartelet H, Béliveau R. Tumor environment dictates medulloblastoma cancer stem cell expression and invasive phenotype. Mol Cancer Res 2008; 6:907-16. [PMID: 18567795 DOI: 10.1158/1541-7786.mcr-07-2184] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neural precursor surface marker CD133 is thought to be enriched in brain cancer stem cells and in radioresistant DAOY medulloblastoma-derived tumor cells. Given that membrane type-1 matrix metalloproteinase (MT1-MMP) expression is a hallmark of highly invasive, radioresistant, and hypoxic brain tumor cells, we sought to determine whether MT1-MMP and other MMPs could regulate the invasive phenotype of CD133(+) DAOY cells. We found that when DAOY medulloblastoma or U87 glioblastoma cells were implanted in nude mice, only those cells specifically implanted in the brain environment generated CD133(+) brain tumors. Vascular endothelial growth factor and basic fibroblast growth factor gene expression increases in correlation with CD133 expression in those tumors. When DAOY cultures were induced to generate in vitro neurosphere-like cells, gene expression of CD133, MT1-MMP, MMP-9, and MDR-1 was induced and correlated with an increase in neurosphere invasiveness. Specific small interfering RNA gene silencing of either MT1-MMP or MMP-9 reduced the capacity of the DAOY monolayers to generate neurospheres and concomitantly abrogated their invasive capacity. On the other hand, overexpression of MT1-MMP in DAOY triggered neurosphere-like formation which was further amplified when cells were cultured in neurosphere medium. Collectively, we show that both MT1-MMP and MMP-9 contribute to the invasive phenotype during CD133(+) neurosphere-like formation in medulloblastoma cells. Increases in MMP-9 may contribute to the opening of the blood-brain barrier, whereas increased MT1-MMP would promote brain tumor infiltration. Our study suggests that MMP-9 or MT1-MMP targeting may reduce the formation of brain tumor stem cells.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire de Médecine Moléculaire, Université du Québec à Montréal, CP 8888, Succursale Centre-ville, Montreal, Quebec, Canada H3C 3P8
| | | | | | | | | | | | | |
Collapse
|
42
|
Fortier S, Labelle D, Sina A, Moreau R, Annabi B. Silencing of the MT1-MMP/ G6PT axis suppresses calcium mobilization by sphingosine-1-phosphate in glioblastoma cells. FEBS Lett 2008; 582:799-804. [PMID: 18267120 DOI: 10.1016/j.febslet.2008.01.061] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 01/30/2008] [Accepted: 01/31/2008] [Indexed: 01/25/2023]
Abstract
The contributions of membrane type-1 matrix metalloproteinase (MT1-MMP) and of the glucose-6-phosphate transporter (G6PT) in sphingosine-1-phosphate (S1P)-mediated Ca(2+) mobilization were assessed in glioblastoma cells. We show that gene silencing of MT1-MMP or G6PT decreased the extent of S1P-induced Ca(2+) mobilization, chemotaxis, and extracellular signal-related kinase phosphorylation. Chlorogenic acid and (-)-epigallocatechin-3-gallate, two diet-derived inhibitors of G6PT and of MT1-MMP, respectively, reduced S1P-mediated Ca(2+) mobilization. An intact MT1-MMP/G6PT signaling axis is thus required for efficient Ca(2+) mobilization in response to bioactive lipids such as S1P. Targeted inhibition of either MT1-MMP or G6PT may lead to reduced infiltrative and invasive properties of brain tumor cells.
Collapse
Affiliation(s)
- Simon Fortier
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre BioMed, Université du Québec à Montréal, Succ Centre-ville, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
43
|
Chi A, Norden AD, Wen PY. Inhibition of angiogenesis and invasion in malignant gliomas. Expert Rev Anticancer Ther 2008; 7:1537-60. [PMID: 18020923 DOI: 10.1586/14737140.7.11.1537] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Malignant gliomas confer a dismal prognosis. As the molecular events that underlie tumor angiogenesis are elucidated, angiogenesis inhibition is emerging as a promising therapy for recurrent and newly diagnosed tumors. Data from animal studies suggest that angiogenesis inhibition may promote an invasive phenotype in tumor cells. This may represent an important mechanism of resistance to antiangiogenic therapies. Recent studies have begun to clarify the mechanisms by which glioma cells detach from the tumor mass, remodel the extracellular matrix and infiltrate normal brain. An array of potential therapeutic targets exists. Combination therapy with antiangiogenic and novel anti-invasion agents is a promising approach that may produce a synergistic antitumor effect and a survival benefit for patients with these devastating tumors.
Collapse
Affiliation(s)
- Andrew Chi
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Division of Neuro-Oncology, Department of Neurology, Brigham & Women's Hospital, SW430D, 44 Binney Street, Boston, MA 02115, USA.
| | | | | |
Collapse
|
44
|
Silva WI, Maldonado HM, Velázquez G, García JO, González FA. Caveolins in glial cell model systems: from detection to significance. J Neurochem 2008; 103 Suppl 1:101-12. [PMID: 17986145 DOI: 10.1111/j.1471-4159.2007.04712.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Glial cells prevail in number and in diversity of cellular phenotypes in the nervous system. They have also gained prominence due to their multiple physiological and pathophysiological roles. Our current knowledge of the asymmetry and heterogeneity of the plasma membrane demands an in depth analysis of the diverse array of membrane microdomains postulated to exist in the context of glial cells. This review focuses and analyzes the studies reported to date on the detection of caveolae membrane rafts and the caveolin family members in glial cell model systems, the conditions leading to changes in their level of expression, and their functional and clinical significance. Outstanding in this work emerge the ubiquitous expression of caveolins, including the typically regarded 'muscle-specific' cav3, in diverse glial cell model systems, their participation in reactive astrogliosis, cancer, and their key relevance to calcium signaling. The knowledge obtained to date demands incorporation of the caveolins and caveolae membrane rafts in our current models on the role of glial cells in heath and neurological disease.
Collapse
Affiliation(s)
- W I Silva
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | | | | | | | | |
Collapse
|
45
|
Tabu K, Ohba Y, Suzuki T, Makino Y, Kimura T, Ohnishi A, Sakai M, Watanabe T, Tanaka S, Sawa H. Oligodendrocyte lineage transcription factor 2 inhibits the motility of a human glial tumor cell line by activating RhoA. Mol Cancer Res 2007; 5:1099-109. [PMID: 17951409 DOI: 10.1158/1541-7786.mcr-07-0096] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The basic helix-loop-helix transcription factor, oligodendrocyte lineage transcription factor 2 (OLIG2), is specifically expressed in the developing and mature central nervous system and plays an important role in oligodendrogenesis from neural progenitors. It is also expressed in various types of glial tumors, but rarely in glioblastoma. Although we previously showed that OLIG2 expression inhibits glioma cell growth, its role in tumorigenesis remains incompletely understood. Here, we investigated the effect of OLIG2 expression on the migration of the human glioblastoma cell line U12-1. In these cells, OLIG2 expression is controlled by the Tet-off system. Induction of OLIG2 expression inhibited both the migration and invasiveness of U12-1 cells. OLIG2 expression also increased the activity of the GTPase RhoA as well as inducing the cells to form stress fibers and focal adhesions. Experiments using short interfering RNA against p27(Kip1) revealed that up-regulation of the p27(Kip1) protein was not essential for RhoA activation, rather it contributed independently to the decreased motility of OLIG2-expressing U12-1 cells. Alternatively, semiquantitative reverse transcription-PCR analysis revealed that mRNA expression of RhoGAP8, which regulates cell migration, was decreased by OLIG2 expression. Furthermore, expression of C3 transferase, which inhibits Rho via ADP ribosylation, attenuated the OLIG2-induced inhibition of cell motility. Imaging by fluorescence resonance energy transfer revealed that in U12-1 cells lacking OLIG2, the active form of RhoA was localized to protrusions of the cell membrane. In contrast, in OLIG2-expressing cells, it lined almost the entire plasma membrane. Thus, OLIG2 suppresses the motile phenotype of glioblastoma cells by activating RhoA.
Collapse
Affiliation(s)
- Kouichi Tabu
- Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Differentially expressed genes are marked by histone 3 lysine 9 trimethylation in human cancer cells. Oncogene 2007; 27:2412-21. [DOI: 10.1038/sj.onc.1210895] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Kasper G, Reule M, Tschirschmann M, Dankert N, Stout-Weider K, Lauster R, Schrock E, Mennerich D, Duda GN, Lehmann KE. Stromelysin-3 over-expression enhances tumourigenesis in MCF-7 and MDA-MB-231 breast cancer cell lines: involvement of the IGF-1 signalling pathway. BMC Cancer 2007; 7:12. [PMID: 17233884 PMCID: PMC1783860 DOI: 10.1186/1471-2407-7-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Accepted: 01/17/2007] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Stromelysin-3 (ST-3) is over-expressed in the majority of human carcinomas including breast carcinoma. Due to its known effect in promoting tumour formation, but its impeding effect on metastasis, a dual role of ST-3 in tumour progression, depending on the cellular grade of dedifferentiation, was hypothesized. METHODS The present study was designed to investigate the influence of ST-3 in vivo and in vitro on the oestrogen-dependent, non-invasive MCF-7 breast carcinoma cell line as well as on the oestrogen-independent, invasive MDA-MB-231 breast carcinoma cell line. Therefore an orthotopic human xenograft tumour model in nude mice, as well as a 3D matrigel cell culture system, were employed. RESULTS Using both in vitro and in vivo techniques, we have demonstrated that over-expression of ST-3 in MCF-7 and MDA-MB-231 cells leads to both increased cell numbers and tumour volumes. This observation was dependent upon the presence of growth factors. In particular, the enhanced proliferative capacity was in MCF-7/ST-3 completely and in MDA-MB-231/ST-3 cells partially dependent on the IGF-1 signalling pathway. Microarray analysis of ST-3 over-expressing cells revealed that in addition to cell proliferation, further biological processes seemed to be affected, such as cell motility and stress response. The MAPK-pathway as well as the Wnt and PI3-kinase pathways, appear to also play a potential role. Furthermore, we have demonstrated that breast cancer cell lines of different differentiation status, as well as the non-tumourigenic cell line MCF-10A, have a comparable capability to induce endogenous ST-3 expression in fibroblasts. CONCLUSION These data reveal that ST-3 is capable of enhancing tumourigenesis in highly differentiated "early stage" breast cancer cell lines as well as in further progressed breast cancer cell lines that have already undergone epithelial-mesenchymal transition. We propose that ST-3 induction in tumour fibroblasts leads to the stimulation of the IGF-1R pathway in carcinoma cells, thus enhancing their proliferative capacity. In addition, further different cellular processes seem to be activated by ST-3, possibly accounting for the dual role of ST-3 in tumour progression and metastasis.
Collapse
Affiliation(s)
- Grit Kasper
- Musculoskeletal Research Center Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Niels Dankert
- Musculoskeletal Research Center Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Stout-Weider
- Institute of Medical Genetics, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Evelin Schrock
- Institute of Clinical Genetics, Technische Universität, Dresden, Germany
| | | | - Georg N Duda
- Musculoskeletal Research Center Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Kerstin E Lehmann
- Center for Cardiovascular Research, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
48
|
Ngameni B, Touaibia M, Patnam R, Belkaid A, Sonna P, Ngadjui BT, Annabi B, Roy R. Inhibition of MMP-2 secretion from brain tumor cells suggests chemopreventive properties of a furanocoumarin glycoside and of chalcones isolated from the twigs of Dorstenia turbinata. PHYTOCHEMISTRY 2006; 67:2573-9. [PMID: 17070879 DOI: 10.1016/j.phytochem.2006.09.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Revised: 08/22/2006] [Accepted: 09/18/2006] [Indexed: 05/12/2023]
Abstract
A furanocoumarin glycoside new named turbinatocoumarin (1) was isolated from the twigs of Dorstenia turbinata. The structure of turbinatocoumarin (1) was assigned as 5-methoxy-3-[3-(beta-glucopyranosyloxy)-2-hydroxy-3-methylbutyl]psoralen by means of spectroscopic analysis. Known compounds have also been isolated from this genus and identified as (2'S, 3'R)-3'-hydroxymarmesin (2), 5-methoxy-3-(3-methyl-2,3-dihydroxybutyl)psoralen (3), psoralen (4), kanzonol C (5) which was isolated for the first time from this genus, 4-hydroxylonchocarpin (6), umbelliferone, 4-hydroxy-3-methoxybenzaldehyde and 4-methoxyphenol. As part of our continuing search for potential naturally-occurring antitumor drug candidates, the inhibition of matrix metalloproteinase (MMP)-2 secretion from brain tumor-derived glioblastoma cells by the isolated compounds 1, 3, 5, and 6 was evaluated by zymography and compared to the documented naturally-occurring MMP secretion inhibitors chlorogenic acid (CHL) and epigallocatechin-3-gallate (EGCg). Among the compounds tested, the inhibiting MMP secretion concentrations ranged from 0.025 to 250 microM with up to 80% inhibition. The inhibitory activities of compounds 5 and 6 were found comparable to the common reference compounds CHL and EGCg. This suggests that alternate sources can be explored and exploited for the availability of chemopreventive molecules.
Collapse
Affiliation(s)
- Bathelemy Ngameni
- Département de Chimie, Université du Québec à Montréal, Montreal, 2101 Jeanne-Mance, CP 8888, Succ., Centre-ville, Quebec, Canada H3C 3P8
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Salhia B, Tran NL, Symons M, Winkles JA, Rutka JT, Berens ME. Molecular pathways triggering glioma cell invasion. Expert Rev Mol Diagn 2006; 6:613-26. [PMID: 16824034 DOI: 10.1586/14737159.6.4.613] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The efficacy of treating malignant gliomas with adjuvant therapies remains largely unsuccessful due to the inability to effectively target invading cells. Although our understanding of glioma oncogenesis has steadily improved, the molecular mechanisms that mediate glioma invasion are still poorly understood. It is clear that genetic alterations in malignant gliomas affect cell proliferation and cell cycle control, which are the targets of most chemotherapeutic agents. However, effective therapy against cell invasion has been less successful. Future treatment protocols must incorporate pharmacotherapeutic strategies that target resistant infiltrative glioma cells as well as proliferating ones. Thus, delineating the point of convergence of signaling pathways, which mediate glioma invasion, proliferation and apoptosis, may identify novel targets that can serve as possible points of therapeutic intervention. The optimization of novel strategies will require reliable preclinical testing using an in vivo animal model of brain invasion. Current applications of existing animal models are not currently optimized or characterized for use in glioma invasion research. As such, the development of a bona fide brain invasion model in vivo must be established. Progress in understanding molecular mechanisms driving glioma invasion will be critical to the success of managing and improving the outcome of patients with this grave disease.
Collapse
Affiliation(s)
- Bodour Salhia
- The Arthur & Sonia Labatt Brain Tumour Research Center, The Hospital for Sick Children, The University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
50
|
Meriane M, Duhamel S, Lejeune L, Galipeau J, Annabi B. Cooperation of Matrix Metalloproteinases with the RhoA/Rho Kinase and Mitogen-Activated Protein Kinase Kinase-1/Extracellular Signal-Regulated Kinase Signaling Pathways Is Required for the Sphingosine-1-Phosphate-Induced Mobilization of Marrow-Derived Stromal Cells. Stem Cells 2006; 24:2557-65. [PMID: 16931773 DOI: 10.1634/stemcells.2006-0209] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The ease of isolation and ex vivo culture of marrow-derived stromal cells (MSCs) from adult bone marrow renders them a very promising source of adult stem cells for gene transfer and cell therapy. However, little is known about the signaling pathways that control their in vivo mobilization and subsequent biodistribution. Platelet-derived sphingosine-1-phosphate (S1P), a bioactive lipid that acts via G-protein-coupled-receptors, exerts strong chemoattraction upon MSCs through yet-uncharacterized signaling pathways. We show that the S1P-induced migration and morphological changes of MSCs in vitro require the activities of extracellular signal-regulated kinase (ERK), Rho kinase (ROCK), and matrix metalloproteinase (MMP) signaling molecules. Specifically, S1P-induced remodeling of the MSC cytoskeleton led to the rapid (<1 minute) formation of actin stress fibers via activation of the RhoA/ROCK pathway and required the catalytic activity of MMPs. S1P-induced activation of the mitogen-activated protein kinase kinase-1 (MEK1)/ERK pathway also contributed to the induction of the actin stress fibers and to the redistribution of paxillin at the focal adhesions through tyrosine phosphorylation of focal adhesion kinase in an MMP-dependent manner. Moreover, MMP- and ROCK-dependent molecular events are implicated in the regulation of the S1P-induced activation of ERK. Our results demonstrate that MSC mobilization in response to S1P requires cooperation between MMP-mediated signaling events and the RhoA/ROCK and MEK1/ERK intracellular pathways. Therefore, the characterization of the cellular factors and the intracellular signaling pathways underlying MSC mobilization is crucial to achieve high efficacy in therapeutic use.
Collapse
Affiliation(s)
- Mayya Meriane
- Laboratoire d'Oncologie Moléculaire, Département de Chimie, Centre BioMed, Université du Québec à Montréal, Canada
| | | | | | | | | |
Collapse
|