1
|
Mosna MJ, Garde FJ, Stinson MG, Pastore CD, Carcagno AL. The chorioallantoic membrane (CAM) model: From its origins in developmental biology to its role in cancer research. Dev Biol 2025; 519:79-95. [PMID: 39694172 DOI: 10.1016/j.ydbio.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/24/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Over the past century, the chick embryo model, historically employed for research in developmental biology, has become a valuable tool for cancer research. The characteristics of the chick chorioallantoic membrane (CAM) make it a convenient model for the study of cancer, leading to the establishment of the CAM assay as an alternative to traditional in vivo cancer models. In this review we will explore the characteristics of the CAM that make it suitable for cancer research, as well as its consolidation as a versatile platform in this field. We will put particular emphasis on describing the key features that make this model an important asset for studying the hallmarks of cancer and for testing a wide variety of therapeutic strategies for its treatment, and which make it a suitable host for patient-derived xenografts (PDX). Additionally, we will examine the wide spectrum of methodological approaches available to study these subjects, highlighting some innovative cases. Finally, we will discuss the advantages and disadvantages of the chick CAM as a model for cancer research and how we can improve this model to its full potential.
Collapse
Affiliation(s)
- María Jimena Mosna
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Federico J Garde
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Marcelo G Stinson
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Candela D Pastore
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Abel L Carcagno
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; Departamento de Ecología, Genética y Evolución, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina.
| |
Collapse
|
2
|
Fermi V, Warta R, Wöllner A, Lotsch C, Jassowicz L, Rapp C, Knoll M, Jungwirth G, Jungk C, Dao Trong P, von Deimling A, Abdollahi A, Unterberg A, Herold-Mende C. Effective Reprogramming of Patient-Derived M2-Polarized Glioblastoma-Associated Microglia/Macrophages by Treatment with GW2580. Clin Cancer Res 2023; 29:4685-4697. [PMID: 37682326 DOI: 10.1158/1078-0432.ccr-23-0576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/26/2023] [Accepted: 09/05/2023] [Indexed: 09/09/2023]
Abstract
PURPOSE Targeting immunosuppressive and pro-tumorigenic glioblastoma (GBM)-associated macrophages and microglial cells (GAM) has great potential to improve patient outcomes. Colony-stimulating factor-1 receptor (CSF1R) has emerged as a promising target for reprograming anti-inflammatory M2-like GAMs. However, treatment data on patient-derived, tumor-educated GAMs and their influence on the adaptive immunity are lacking. EXPERIMENTAL DESIGN CD11b+-GAMs freshly isolated from patient tumors were treated with CSF1R-targeting drugs PLX3397, BLZ945, and GW2580. Phenotypical changes upon treatment were assessed using RNA sequencing, flow cytometry, and cytokine quantification. Functional analyses included inducible nitric oxide synthase activity, phagocytosis, transmigration, and autologous tumor cell killing assays. Antitumor effects and changes in GAM activation were confirmed in a complex patient-derived 3D tumor organoid model serving as a tumor avatar. RESULTS The most effective reprogramming of GAMs was observed upon GW2580 treatment, which led to the downregulation of M2-related markers, IL6, IL10, ERK1/2, and MAPK signaling pathways, while M1-like markers, gene set enrichment indicating activated MHC-II presentation, phagocytosis, and T-cell killing were substantially increased. Moreover, treatment of patient-derived GBM organoids with GW2580 confirmed successful reprogramming, resulting in impaired tumor cell proliferation. In line with its failure in clinical trials, PLX3397 was ineffective in our analysis. CONCLUSIONS This comparative analysis of CSF1R-targeting drugs on patient-derived GAMs and human GBM avatars identified GW2580 as the most powerful inhibitor with the ability to polarize immunosuppressive GAMs to a proinflammatory phenotype, supporting antitumor T-cell responses while also exerting a direct antitumor effect. These data indicate that GW2580 could be an important pillar in future therapies for GBM.
Collapse
Affiliation(s)
- Valentina Fermi
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Rolf Warta
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg, Germany
| | - Amélie Wöllner
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Catharina Lotsch
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Lena Jassowicz
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 522, Heidelberg, Germany
| | - Carmen Rapp
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Maximilian Knoll
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), University Hospital of Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Gerhard Jungwirth
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Christine Jungk
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Philip Dao Trong
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Andreas von Deimling
- Dept. of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Amir Abdollahi
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), University Hospital of Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
| | - Christel Herold-Mende
- Department of Neurosurgical Research, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany
- German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg, Germany
| |
Collapse
|
3
|
Zhou X, Jin G, Zhang J, Liu F. Recruitment mechanisms and therapeutic implications of tumor-associated macrophages in the glioma microenvironment. Front Immunol 2023; 14:1067641. [PMID: 37153567 PMCID: PMC10157099 DOI: 10.3389/fimmu.2023.1067641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
As one of the main components of the glioma immune microenvironment, glioma-associated macrophages (GAMs) have increasingly drawn research interest. Primarily comprised of resident microglias and peripherally derived mononuclear macrophages, GAMs are influential in a variety of activities such as tumor cell resistance to chemotherapy and radiotherapy as well as facilitation of glioma pathogenesis. In addition to in-depth research of GAM polarization, study of mechanisms relevant in tumor microenvironment recruitment has gradually increased. Suppression of GAMs at their source is likely to produce superior therapeutic outcomes. Here, we summarize the origin and recruitment mechanism of GAMs, as well as the therapeutic implications of GAM inhibition, to facilitate future glioma-related research and formulation of more effective treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Fusheng Liu
- *Correspondence: Junwen Zhang, ; Fusheng Liu,
| |
Collapse
|
4
|
Wang L, Li S, Fan H, Han M, Xie J, Du J, Peng F. Bifidobacterium lactis combined with Lactobacillus plantarum inhibit glioma growth in mice through modulating PI3K/AKT pathway and gut microbiota. Front Microbiol 2022; 13:986837. [PMID: 36147842 PMCID: PMC9486703 DOI: 10.3389/fmicb.2022.986837] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Glioma is a common primary aggressive tumor with limited clinical treatment. Recently, growing research suggests that gut microbiota is involved in tumor progression, and several probiotics can inhibit tumor growth. However, evidence for the effect of probiotics on glioma is lacking. Here, we found that Bifidobacterium (B.) lactis combined with Lactobacillus (L.) plantarum reduced tumor volume, prolonged survival time and repaired the intestinal barrier damage in an orthotopic mouse model of glioma. Experiments demonstrated that B. lactis combined with L. plantarum suppressed the PI3K/AKT pathway and down-regulated the expression of Ki-67 and N-cadherin. The glioma-inhibitory effect of probiotic combination is also related to the modulation of gut microbiota composition, which is characterized by an increase in relative abundance of Lactobacillus and a decrease in some potential pathogenic bacteria. Additionally, probiotic combination altered fecal metabolites represented by fatty acyls and organic oxygen compounds. Together, our results prove that B. lactis combined with L. plantarum can inhibit glioma growth by suppressing PI3K/AKT pathway and regulating gut microbiota composition and metabolites in mice, thus suggesting the potential benefits of B. lactis and L. plantarum against glioma.
Collapse
Affiliation(s)
- Li Wang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Sui Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Huali Fan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mingyu Han
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jie Xie
- College of Life Sciences, Sichuan Normal University, Chengdu, China
| | - Junrong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Junrong Du,
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- Fu Peng,
| |
Collapse
|
5
|
Wei J, Song R, Sabbagh A, Marisetty A, Shukla N, Fang D, Najem H, Ott M, Long J, Zhai L, Lesniak MS, James CD, Platanias L, Curran M, Heimberger AB. Cell-directed aptamer therapeutic targeting for cancers including those within the central nervous system. Oncoimmunology 2022; 11:2062827. [PMID: 35433114 PMCID: PMC9009928 DOI: 10.1080/2162402x.2022.2062827] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Osteopontin (OPN) is produced by tumor cells as well as by myeloid cells and is enriched in the tumor microenvironment (TME) of many cancers. Given the roles of OPN in tumor progression and immune suppression, we hypothesized that targeting OPN with aptamers that have high affinity and specificity could be a promising therapeutic strategy. Bi-specific aptamers targeting ligands for cellular internalization were conjugated to siRNAs to suppress OPN were created, and therapeutic leads were selected based on target engagement and in vivo activity. Aptamers as carriers for siRNA approaches were created including a cancer targeting nucleolin aptamer Ncl-OPN siRNA and a myeloid targeting CpG oligodeoxynucleotide (ODN)-OPN siRNA conjugate. These aptamers were selected as therapeutic leads based on 70–90% OPN inhibition in cancer (GL261, 344SQ, 4T1B2b) and myeloid (DC2.4) cells relative to scramble controls. In established immune competent 344SQ lung cancer and 4T1B2b breast cancer models, these aptamers, including in combination, demonstrate therapeutic activity by inhibiting tumor growth. The Ncl-OPN siRNA aptamer demonstrated efficacy in an immune competent orthotopic glioma model administered systemically secondary to the ability of the aptamer to access the glioma TME. Therapeutic activity was demonstrated using both aptamers in a breast cancer brain metastasis model. Targeted inhibition of OPN in tumor cells and myeloid cells using bifunctional aptamers that are internalized by specific cell types and suppress OPN expression once internalized may have clinical potential in cancer treatment.
Collapse
Affiliation(s)
- Jun Wei
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renduo Song
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aria Sabbagh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anantha Marisetty
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neal Shukla
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dexing Fang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Martina Ott
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lijie Zhai
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Charles David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Leonidas Platanias
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Michael Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| |
Collapse
|
6
|
Jafari SH, Rabiei N, Taghizadieh M, Mirazimi SMA, Kowsari H, Farzin MA, Razaghi Bahabadi Z, Rezaei S, Mohammadi AH, Alirezaei Z, Dashti F, Nejati M. Joint application of biochemical markers and imaging techniques in the accurate and early detection of glioblastoma. Pathol Res Pract 2021; 224:153528. [PMID: 34171601 DOI: 10.1016/j.prp.2021.153528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/28/2022]
Abstract
Glioblastoma is a primary brain tumor with the most metastatic effect in adults. Despite the wide range of multidimensional treatments, tumor heterogeneity is one of the main causes of tumor spread and gives great complexity to diagnostic and therapeutic methods. Therefore, featuring noble noninvasive prognostic methods that are focused on glioblastoma heterogeneity is perceived as an urgent need. Imaging neuro-oncological biomarkers including MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status, tumor grade along with other tumor characteristics and demographic features (e.g., age) are commonly referred to during diagnostic, therapeutic and prognostic processes. Therefore, the use of new noninvasive prognostic methods focused on glioblastoma heterogeneity is considered an urgent need. Some neuronal biomarkers, including the promoter methylation status of the promoter MGMT, the characteristics and grade of the tumor, along with the patient's demographics (such as age and sex) are involved in diagnosis, treatment, and prognosis. Among the wide array of imaging techniques, magnetic resonance imaging combined with the more physiologically detailed technique of H-magnetic resonance spectroscopy can be useful in diagnosing neurological cancer patients. In addition, intracranial tumor qualitative analysis and sometimes tumor biopsies help in accurate diagnosis. This review summarizes the evidence for biochemical biomarkers being a reliable biomarker in the early detection and disease management in GBM. Moreover, we highlight the correlation between Imaging techniques and biochemical biomarkers and ask whether they can be combined.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women's Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sayad Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Kowsari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Amin Farzin
- Department of Laboratory Medicine, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Razaghi Bahabadi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Mohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Alirezaei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Paramedical School, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
7
|
Cruz RGB, Madden SF, Richards CE, Vellanki SH, Jahns H, Hudson L, Fay J, O’Farrell N, Sheehan K, Jirström K, Brennan K, Hopkins AM. Human Epidermal Growth Factor Receptor-3 Expression Is Regulated at Transcriptional Level in Breast Cancer Settings by Junctional Adhesion Molecule-A via a Pathway Involving Beta-Catenin and FOXA1. Cancers (Basel) 2021; 13:cancers13040871. [PMID: 33669586 PMCID: PMC7922773 DOI: 10.3390/cancers13040871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary Signaling from the human epidermal growth factor receptor (HER) family of proteins increases in many cancers, including breast. HER2-high breast cancers are successfully treated with anti-HER2 therapies, but these drugs are limited by the fact that patients frequently develop resistance to them. One common mechanism by which resistance develops is when tumors acquire high levels of a family member called HER3. We had previously shown that a protein called JAM-A regulates the level of HER2 in breast cancer cells, and is associated with the development of resistance to HER2-targeted therapies. In this study we show for the first time that JAM-A levels also regulate those of HER3. Using breast cancer cell and tissue models and culminating in patient tissue material, we provide evidence that JAM-A regulates HER3 expression via a pathway involving the transcription factors β-catenin and FOXA1. We suggest that JAM-A merits future investigation as a novel drug target for its potential to reduce HER3 tumorigenic signaling and to offset the development of resistance to HER2-targeted therapies. Abstract The success of breast cancer therapies targeting the human epidermal growth factor receptor-2 (HER2) is limited by the development of drug resistance by mechanisms including upregulation of HER3. Having reported that HER2 expression and resistance to HER2-targeted therapies can be regulated by Junctional Adhesion Molecule-A (JAM-A), this study investigated if JAM-A regulates HER3 expression. Expressional alteration of JAM-A in breast cancer cells was used to test expressional effects on HER3 and its effectors, alongside associated functional behaviors, in vitro and semi-in vivo. HER3 transcription factors were identified and tested for regulation by JAM-A. Finally a patient tissue microarray was used to interrogate connections between putative pathway components connecting JAM-A and HER3. This study reveals for the first time that HER3 and its effectors are regulated at gene/protein expression level by JAM-A in breast cancer cell lines; with functional consequences in in vitro and semi-in vivo models. In bioinformatic, cellular and patient tissue models, this was associated with regulation of the HER3 transcription factor FOXA1 by JAM-A via a pathway involving β-catenin. Our data suggest a novel model whereby JAM-A expression regulates β-catenin localization, in turn regulating FOXA1 expression, which could drive HER3 gene transcription. JAM-A merits investigation as a novel target to prevent upregulation of HER3 during the development of resistance to HER2-targeted therapies, or to reduce HER3-dependent tumorigenic signaling.
Collapse
Affiliation(s)
- Rodrigo G. B. Cruz
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (R.G.B.C.); (C.E.R.); (S.H.V.); (L.H.); (K.B.)
| | - Stephen F. Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland;
| | - Cathy E. Richards
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (R.G.B.C.); (C.E.R.); (S.H.V.); (L.H.); (K.B.)
| | - Sri HariKrishna Vellanki
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (R.G.B.C.); (C.E.R.); (S.H.V.); (L.H.); (K.B.)
| | - Hanne Jahns
- Pathobiology Section, UCD School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland;
| | - Lance Hudson
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (R.G.B.C.); (C.E.R.); (S.H.V.); (L.H.); (K.B.)
| | - Joanna Fay
- Department of Pathology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (J.F.); (N.O.); (K.S.)
| | - Naoimh O’Farrell
- Department of Pathology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (J.F.); (N.O.); (K.S.)
| | - Katherine Sheehan
- Department of Pathology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (J.F.); (N.O.); (K.S.)
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, SE 221 85 Lund, Sweden;
| | - Kieran Brennan
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (R.G.B.C.); (C.E.R.); (S.H.V.); (L.H.); (K.B.)
| | - Ann M. Hopkins
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland; (R.G.B.C.); (C.E.R.); (S.H.V.); (L.H.); (K.B.)
- Correspondence: ; Tel.: +353-1-809-3858
| |
Collapse
|
8
|
Osteopontin: A Key Regulator of Tumor Progression and Immunomodulation. Cancers (Basel) 2020; 12:cancers12113379. [PMID: 33203146 PMCID: PMC7698217 DOI: 10.3390/cancers12113379] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Anti-PD-1/PD-L1 and anti-CTLA-4-based immune checkpoint blockade (ICB) immunotherapy have recently emerged as a breakthrough in human cancer treatment. Durable efficacy has been achieved in many types of human cancers. However, not all human cancers respond to current ICB immunotherapy and only a fraction of the responsive cancers exhibit efficacy. Osteopontin (OPN) expression is highly elevated in human cancers and functions as a tumor promoter. Emerging data suggest that OPN may also regulate immune cell function in the tumor microenvironment. This review aims at OPN function in human cancer progression and new findings of OPN as a new immune checkpoint. We propose that OPN compensates PD-L1 function to promote tumor immune evasion, which may underlie human cancer non-response to current ICB immunotherapy. Abstract OPN is a multifunctional phosphoglycoprotein expressed in a wide range of cells, including osteoclasts, osteoblasts, neurons, epithelial cells, T, B, NK, NK T, myeloid, and innate lymphoid cells. OPN plays an important role in diverse biological processes and is implicated in multiple diseases such as cardiovascular, diabetes, kidney, proinflammatory, fibrosis, nephrolithiasis, wound healing, and cancer. In cancer patients, overexpressed OPN is often detected in the tumor microenvironment and elevated serum OPN level is correlated with poor prognosis. Initially identified in activated T cells and termed as early T cell activation gene, OPN links innate cells to adaptive cells in immune response to infection and cancer. Recent single cell RNA sequencing revealed that OPN is primarily expressed in tumor cells and tumor-infiltrating myeloid cells in human cancer patients. Emerging experimental data reveal a key role of OPN is tumor immune evasion through regulating macrophage polarization, recruitment, and inhibition of T cell activation in the tumor microenvironment. Therefore, in addition to its well-established direct tumor cell promotion function, OPN also acts as an immune checkpoint to negatively regulate T cell activation. The OPN protein level is highly elevated in peripheral blood of human cancer patients. OPN blockade immunotherapy with OPN neutralization monoclonal antibodies (mAbs) thus represents an attractive approach in human cancer immunotherapy.
Collapse
|
9
|
Dumas AA, Pomella N, Rosser G, Guglielmi L, Vinel C, Millner TO, Rees J, Aley N, Sheer D, Wei J, Marisetty A, Heimberger AB, Bowman RL, Brandner S, Joyce JA, Marino S. Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J 2020; 39:e103790. [PMID: 32567735 PMCID: PMC7396846 DOI: 10.15252/embj.2019103790] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 05/03/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022] Open
Abstract
Tumour-associated microglia/macrophages (TAM) are the most numerous non-neoplastic populations in the tumour microenvironment in glioblastoma multiforme (GBM), the most common malignant brain tumour in adulthood. The mTOR pathway, an important regulator of cell survival/proliferation, is upregulated in GBM, but little is known about the potential role of this pathway in TAM. Here, we show that GBM-initiating cells induce mTOR signalling in the microglia but not bone marrow-derived macrophages in both in vitro and in vivo GBM mouse models. mTOR-dependent regulation of STAT3 and NF-κB activity promotes an immunosuppressive microglial phenotype. This hinders effector T-cell infiltration, proliferation and immune reactivity, thereby contributing to tumour immune evasion and promoting tumour growth in mouse models. The translational value of our results is demonstrated in whole transcriptome datasets of human GBM and in a novel in vitro model, whereby expanded-potential stem cells (EPSC)-derived microglia-like cells are conditioned by syngeneic patient-derived GBM-initiating cells. These results raise the possibility that microglia could be the primary target of mTOR inhibition, rather than the intrinsic tumour cells in GBM.
Collapse
Affiliation(s)
- Anaelle A Dumas
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Nicola Pomella
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Gabriel Rosser
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Loredana Guglielmi
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Claire Vinel
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Thomas O Millner
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Jeremy Rees
- National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Natasha Aley
- Division of NeuropathologyDepartment of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - Denise Sheer
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| | - Jun Wei
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Anantha Marisetty
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Amy B Heimberger
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Robert L Bowman
- Human Oncology and Pathogenesis ProgramMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Sebastian Brandner
- National Hospital for Neurology and NeurosurgeryUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Johanna A Joyce
- Department of OncologyLudwig Institute for Cancer ResearchUniversity of LausanneLausanneSwitzerland
| | - Silvia Marino
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University LondonLondonUK
| |
Collapse
|
10
|
Signaling Determinants of Glioma Cell Invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:129-149. [PMID: 32034712 DOI: 10.1007/978-3-030-30651-9_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
11
|
|
12
|
Szulzewsky F, Schwendinger N, Güneykaya D, Cimino PJ, Hambardzumyan D, Synowitz M, Holland EC, Kettenmann H. Loss of host-derived osteopontin creates a glioblastoma-promoting microenvironment. Neuro Oncol 2019; 20:355-366. [PMID: 29016864 DOI: 10.1093/neuonc/nox165] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background Microglia and periphery-derived monocytes infiltrate human and mouse glioblastoma and their density is positively correlated with malignancy. Using microarray and RNA sequencing, we have previously shown that glioblastoma-associated microglia/monocytes (GAMs) express osteopontin/SPP1. Methods We used quantitative reverse transcriptase PCR, immunofluorescence stainings, western blot, and flow cytometry to identify the various sources of osteopontin (OPN) expression in human and mouse glioblastoma. We implanted wild type GL261 glioblastoma cells, which do not express significant levels of OPN, into wild type and OPN-/- mice to investigate the role of microenvironment-derived OPN on glioblastoma progression. Results Our data indicate that GAMs are the predominant source of OPN in both human and mouse glioblastoma and express only the secreted form of OPN. Loss of microenvironment-derived OPN enhanced tumor progression. Staining by Ki67 and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling showed no difference in overall cell proliferation but a decreased apoptosis rate in tumors in OPN-/- mice. CD31 staining showed a significantly decreased number of microvessels in tumors in OPN-/- mice, accompanied by reduced coverage of vessels with platelet derived growth factor receptor β+ pericytes. Flow cytometry analysis revealed a significant increase of CD11b+/CD45low microglia but not of CD11b+/CD45high macrophages/monocytes in tumors in OPN-/- mice. Sorted CD11b+ cells from wild type and OPN-/- naïve brains and tumors did not show a significant difference in the expression pattern of activation marker genes. Conclusion Our results show that in tested human and mouse glioblastoma samples, OPN is predominantly expressed and secreted by GAMs and that, in contrast to OPN expression in the tumor cells per se, loss of stroma-derived OPN creates a glioblastoma-promoting microenvironment.
Collapse
Affiliation(s)
- Frank Szulzewsky
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association and Berlin Institute of Health, Berlin, Germany
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nina Schwendinger
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association and Berlin Institute of Health, Berlin, Germany
| | - Dilansu Güneykaya
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association and Berlin Institute of Health, Berlin, Germany
| | - Patrick J Cimino
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Dolores Hambardzumyan
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Eric C Holland
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Helmut Kettenmann
- Cellular Neurosciences, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
Knockdown of DSPP inhibits the migration and invasion of glioma cells. Pathol Res Pract 2018; 214:2025-2030. [DOI: 10.1016/j.prp.2018.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/07/2018] [Accepted: 09/28/2018] [Indexed: 11/17/2022]
|
14
|
Wei J, Marisetty A, Schrand B, Gabrusiewicz K, Hashimoto Y, Ott M, Grami Z, Kong LY, Ling X, Caruso H, Zhou S, Wang YA, Fuller GN, Huse J, Gilboa E, Kang N, Huang X, Verhaak R, Li S, Heimberger AB. Osteopontin mediates glioblastoma-associated macrophage infiltration and is a potential therapeutic target. J Clin Invest 2018; 129:137-149. [PMID: 30307407 DOI: 10.1172/jci121266] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/04/2018] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma is highly enriched with macrophages, and osteopontin (OPN) expression levels correlate with glioma grade and the degree of macrophage infiltration; thus, we studied whether OPN plays a crucial role in immune modulation. Quantitative PCR, immunoblotting, and ELISA were used to determine OPN expression. Knockdown of OPN was achieved using complementary siRNA, shRNA, and CRISPR/Cas9 techniques, followed by a series of in vitro functional migration and immunological assays. OPN gene-deficient mice were used to examine the roles of non-tumor-derived OPN on survival of mice harboring intracranial gliomas. Patients with mesenchymal glioblastoma multiforme (GBM) show high OPN expression, a negative survival prognosticator. OPN is a potent chemokine for macrophages, and its blockade significantly impaired the ability of glioma cells to recruit macrophages. Integrin αvβ5 (ITGαvβ5) is highly expressed on glioblastoma-infiltrating macrophages and constitutes a major OPN receptor. OPN maintains the M2 macrophage gene signature and phenotype. Both tumor-derived and host-derived OPN were critical for glioma development. OPN deficiency in either innate immune or glioma cells resulted in a marked reduction in M2 macrophages and elevated T cell effector activity infiltrating the glioma. Furthermore, OPN deficiency in the glioma cells sensitized them to direct CD8+ T cell cytotoxicity. Systemic administration in mice of 4-1BB-OPN bispecific aptamers was efficacious, increasing median survival time by 68% (P < 0.05). OPN is thus an important chemokine for recruiting macrophages to glioblastoma, mediates crosstalk between tumor cells and the innate immune system, and has the potential to be exploited as a therapeutic target.
Collapse
Affiliation(s)
- Jun Wei
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anantha Marisetty
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brett Schrand
- Department of Microbiology & Immunology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Konrad Gabrusiewicz
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuuri Hashimoto
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Martina Ott
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zacharia Grami
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiaoyang Ling
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hillary Caruso
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Gregory N Fuller
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason Huse
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eli Gilboa
- Department of Microbiology & Immunology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Nannan Kang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Roel Verhaak
- Jackson Laboratory of Genomic Medicine, Farmington, Connecticut, USA
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
15
|
Bahwini TM, Zhong Y, Gu C, Nasa Z, Oetomo D. Investigating the Mechanical Properties of Biological Brain Cells With Atomic Force Microscopy. J Med Device 2018. [DOI: 10.1115/1.4040995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Characterization of cell mechanical properties plays an important role in disease diagnoses and treatments. This paper uses advanced atomic force microscopy (AFM) to measure the geometrical and mechanical properties of two different human brain normal HNC-2 and cancer U87 MG cells. Based on experimental measurement, it measures the cell deformation and indentation force to characterize cell mechanical properties. A fitting algorithm is developed to generate the force-loading curves from experimental data. An inverse Hertzian method is also established to identify Young's moduli for HNC-2 and U87 MG cells. The results demonstrate that Young's modulus of cancer cells is different from that of normal cells, which can help us to differentiate normal and cancer cells from the biomechanical viewpoint.
Collapse
Affiliation(s)
| | - Yongmin Zhong
- School of Engineering, RMIT University, Melbourne 3083, Australia e-mail:
| | - Chengfan Gu
- Department of Mechanical Engineering, University of Melbourne, Parkville 3010, Australia e-mail:
| | - Zeyad Nasa
- Micro Nano Research Facility, College of Science, Engineering and Health, RMIT, Melbourne 3000, Australia e-mail:
| | - Denny Oetomo
- Department of Mechanical Engineering, University of Melbourne, Parkville 3010, Australia e-mail:
| |
Collapse
|
16
|
Henry A, Nokin MJ, Leroi N, Lallemand F, Lambert J, Goffart N, Roncarati P, Bianchi E, Peixoto P, Blomme A, Turtoi A, Peulen O, Habraken Y, Scholtes F, Martinive P, Delvenne P, Rogister B, Castronovo V, Bellahcène A. New role of osteopontin in DNA repair and impact on human glioblastoma radiosensitivity. Oncotarget 2018; 7:63708-63721. [PMID: 27563812 PMCID: PMC5325397 DOI: 10.18632/oncotarget.11483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/05/2016] [Indexed: 12/03/2022] Open
Abstract
Glioblastoma (GBM) represents the most aggressive and common solid human brain tumor. We have recently demonstrated the importance of osteopontin (OPN) in the acquisition/maintenance of stemness characters and tumorigenicity of glioma initiating cells. Consultation of publicly available TCGA database indicated that high OPN expression correlated with poor survival in GBM patients. In this study, we explored the role of OPN in GBM radioresistance using an OPN-depletion strategy in U87-MG, U87-MG vIII and U251-MG human GBM cell lines. Clonogenic experiments showed that OPN-depleted GBM cells were sensitized to irradiation. In comet assays, these cells displayed higher amounts of unrepaired DNA fragments post-irradiation when compared to control. We next evaluated the phosphorylation of key markers of DNA double-strand break repair pathway. Activating phosphorylation of H2AX, ATM and 53BP1 was significantly decreased in OPN-deficient cells. The addition of recombinant OPN prior to irradiation rescued phospho-H2AX foci formation thus establishing a new link between DNA repair and OPN expression in GBM cells. Finally, OPN knockdown improved mice survival and induced a significant reduction of heterotopic human GBM xenograft when combined with radiotherapy. This study reveals a new function of OPN in DNA damage repair process post-irradiation thus further confirming its major role in GBM aggressive disease.
Collapse
Affiliation(s)
- Aurélie Henry
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Marie-Julie Nokin
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Natacha Leroi
- Biology and Tumor Development Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - François Lallemand
- Biology and Tumor Development Laboratory, GIGA Cancer, University of Liège, Liège, Belgium.,Department of Radiology, University Hospital Liège, Liège, Belgium.,Cyclotron Research Center, University Hospital Liège, Liège, Belgium
| | | | - Nicolas Goffart
- GIGA Neurosciences, University of Liège, Liège, Belgium.,Department of Neurosurgery, Brain Center Rudolf Magnus Institute of Neurosciences and the T&P Bohnenn Laboratory for Neuro-Oncology University Medical Center, Utrecht, The Netherlands
| | | | - Elettra Bianchi
- Department of Pathology, University Hospital Liège, Liège, Belgium
| | - Paul Peixoto
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Yvette Habraken
- Virology and Immunology Laboratory, University of Liège, Liège, Belgium
| | - Félix Scholtes
- Department of Neurosurgery, University Hospital Liège, Liège, Belgium
| | | | | | | | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| |
Collapse
|
17
|
Functional assessment of glioma pathogenesis by in vivo multi-parametric magnetic resonance imaging and in vitro analyses. Sci Rep 2016; 6:26050. [PMID: 27198662 PMCID: PMC4873752 DOI: 10.1038/srep26050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 04/27/2016] [Indexed: 01/11/2023] Open
Abstract
Gliomas are aggressive brain tumors with poor prognosis. In this study, we report a novel approach combining both in vivo multi-parametric MRI and in vitro cell culture assessments to evaluate the pathogenic development of gliomas. Osteopontin (OPN), a pleiotropic factor, has been implicated in the formation and progression of various human cancers, including gliomas, through its functions in regulating cell proliferation, survival, angiogenesis, and migration. Using rat C6 glioma model, the combined approach successfully monitors the acquisition and decrease of cancer hallmarks. We show that knockdown of the expression of OPN reduces C6 cell proliferation, survival, viability and clonogenicity in vitro, and reduces tumor burden and prolongs animal survival in syngeneic rats. OPN depletion is associated with reduced tumor growth, decreased angiogenesis, and an increase of tumor-associated metabolites, as revealed by T2-weighted images, diffusion-weighted images, Ktrans maps, and 1H-MRS, respectively. These strategies allow us to define an important role of OPN in conferring cancer hallmarks, which can be further applied to assess the functional roles of other candidate genes in glioma. In particular, the non-invasive multi-parametric MRI measurement of cancer hallmarks related to proliferation, angiogenesis and altered metabolism may serve as a useful tool for diagnosis and for patient management.
Collapse
|
18
|
Lamour V, Henry A, Kroonen J, Nokin MJ, von Marschall Z, Fisher LW, Chau TL, Chariot A, Sanson M, Delattre JY, Turtoi A, Peulen O, Rogister B, Castronovo V, Bellahcène A. Targeting osteopontin suppresses glioblastoma stem-like cell character and tumorigenicityin vivo. Int J Cancer 2015; 137:1047-57. [DOI: 10.1002/ijc.29454] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/24/2014] [Accepted: 12/10/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Virginie Lamour
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| | - Aurélie Henry
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| | - Jérôme Kroonen
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liège; Belgium
| | - Marie-Julie Nokin
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| | | | - Larry W. Fisher
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, DHHS; Bethesda MD
| | - Tieu-Lan Chau
- Laboratory of Medical Chemistry, GIGA-Signal Transduction, University of Liège; Belgium
| | - Alain Chariot
- Laboratory of Medical Chemistry, GIGA-Signal Transduction, University of Liège; Belgium
| | - Marc Sanson
- UMR 975, INSERM-UPMC, GH Pitié-Salpêtrière; Paris
| | | | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| | - Bernard Rogister
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liège; Belgium
- Stem Cells and Regenerative Medicine, GIGA-Development, University of Liège; Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège; Belgium
| |
Collapse
|
19
|
Identification of therapeutic targets for glioblastoma by network analysis. Oncogene 2015; 35:608-20. [PMID: 25961929 DOI: 10.1038/onc.2015.119] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/27/2015] [Accepted: 02/17/2015] [Indexed: 01/30/2023]
Abstract
Glioblastoma can originate from terminally differentiated astrocytes and neurons, which can dedifferentiate to a stem cell-like state upon transformation. In this study, we confirmed that transformed dedifferentiated astrocytes and neurons acquired a stem/progenitor cell state, although they still retained gene expression memory from their parental cell. Transcriptional network analysis on these cells identified upregulated genes in three main pathways: Wnt signaling, cell cycle and focal adhesion with the gene Spp1, also known as osteopontin (OPN) serving as a key common node connecting these three pathways. Inhibition of OPN blocked the formation of neurospheres, affected the proliferative capacity of transformed neurons and reduced the expression levels of neural stem cell markers. Specific inhibition of OPN in both murine and human glioma tumors prolonged mice survival. We conclude that OPN is an important player in dedifferentiation of cells during tumor formation, hence its inhibition can be a therapeutic target for glioblastoma.
Collapse
|
20
|
The chick embryo chorioallantoic membrane as a model for tumor biology. Exp Cell Res 2014; 328:314-24. [PMID: 24972385 DOI: 10.1016/j.yexcr.2014.06.010] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 01/08/2023]
Abstract
Among the in vivo models, the chick embryo chorioallantoic membrane (CAM) has been used to implant several tumor types as well as malignant cell lines to study their growth rate, angiogenic potential and metastatic capability. This review article is focused on the major compelling literature data on the use of the CAM to investigate tumor growth and the metastatic process.
Collapse
|
21
|
Bournine L, Bensalem S, Peixoto P, Gonzalez A, Maiza-Benabdesselam F, Bedjou F, Wauters JN, Tits M, Frédérich M, Castronovo V, Bellahcène A. Revealing the anti-tumoral effect of Algerian Glaucium flavum roots against human cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:1211-1218. [PMID: 23860409 DOI: 10.1016/j.phymed.2013.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/19/2013] [Accepted: 06/02/2013] [Indexed: 06/02/2023]
Abstract
Glaucium flavum (G. flavum) is a plant from the Papaveraceae family native to Algeria where it is used in local traditional medicine to treat warts. G. flavum root crude alkaloid extract inhibited breast cancer cell proliferation and induced G2/M phase cycle arrest and apoptosis without affecting normal cells, which is a highly awaited feature of potential anti-cancer agents. G. flavum significantly reduced growth and vascularization of human glioma tumors on chicken chorioallantoic membrane (CAM) in vivo. The chromatographic profile of the dichloromethane extract of G. flavum root showed the presence of different constituents including the isoquinoline alkaloid protopine, as the major compound. We report for the first time that G. flavum extract may represent a new promising agent for cancer chemotherapy.
Collapse
Affiliation(s)
- Lamine Bournine
- Laboratory of Plant Biotechnology and Ethnobotany, Faculty of Natural Sciences and Life, University of Bejaia, Bejaia, Algeria; Metastasis Research Laboratory, GIGA-Cancer, University of Liege, Liege, Belgium; Laboratory of Pharmacognosy, CIRM, University of Liege, Liege, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Coniglio SJ, Segall JE. Review: molecular mechanism of microglia stimulated glioblastoma invasion. Matrix Biol 2013; 32:372-80. [PMID: 23933178 DOI: 10.1016/j.matbio.2013.07.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 07/28/2013] [Accepted: 07/28/2013] [Indexed: 01/01/2023]
Abstract
Glioblastoma multiforme is one of the deadliest human cancers and is characterized by a high degree of microglia and macrophage infiltration. The role of these glioma infiltrating macrophages (GIMs) in disease progression has been the subject of recent investigation. While initially thought to reflect an immune response to the tumor, the balance of evidence clearly suggests GIMs can have potent tumor-tropic functions and assist in glioma cell growth and infiltration into normal brain. In this review, we focus on the evidence for GIMs aiding mediating glioblastoma motility and invasion. We survey the literature for molecular pathways that are involved in paracrine interaction between glioma cells and GIMs and assess which of these might serve as attractive targets for therapeutic intervention.
Collapse
Affiliation(s)
- Salvatore J Coniglio
- Albert Einstein College of Medicine, Department of Anatomy and Structural Biology, Bronx, NY 10461, United States.
| | | |
Collapse
|
23
|
The anti-tumor effect of HDAC inhibition in a human pancreas cancer model is significantly improved by the simultaneous inhibition of cyclooxygenase 2. PLoS One 2013; 8:e75102. [PMID: 24040391 PMCID: PMC3770617 DOI: 10.1371/journal.pone.0075102] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/12/2013] [Indexed: 12/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is the fourth leading cause of cancer death worldwide, with no satisfactory treatment to date. In this study, we tested whether the combined inhibition of cyclooxygenase-2 (COX-2) and class I histone deacetylase (HDAC) may results in a better control of pancreatic ductal adenocarcinoma. The impact of the concomitant HDAC and COX-2 inhibition on cell growth, apoptosis and cell cycle was assessed first in vitro on human pancreas BxPC-3, PANC-1 or CFPAC-1 cells treated with chemical inhibitors (SAHA, MS-275 and celecoxib) or HDAC1/2/3/7 siRNA. To test the potential antitumoral activity of this combination in vivo, we have developed and characterized, a refined chick chorioallantoic membrane tumor model that histologically and proteomically mimics human pancreatic ductal adenocarcinoma. The combination of HDAC1/3 and COX-2 inhibition significantly impaired proliferation of BxPC-3 cells in vitro and stalled entirely the BxPC-3 cells tumor growth onto the chorioallantoic membrane in vivo. The combination was more effective than either drug used alone. Consistently, we showed that both HDAC1 and HDAC3 inhibition induced the expression of COX-2 via the NF-kB pathway. Our data demonstrate, for the first time in a Pancreatic Ductal Adenocarcinoma (PDAC) model, a significant action of HDAC and COX-2 inhibitors on cancer cell growth, which sets the basis for the development of potentially effective new combinatory therapies for pancreatic ductal adenocarcinoma patients.
Collapse
|
24
|
Warnock G, Turtoi A, Blomme A, Bretin F, Bahri MA, Lemaire C, Libert LC, Seret AE, Luxen A, Castronovo V, Plenevaux AR. In Vivo PET/CT in a Human Glioblastoma Chicken Chorioallantoic Membrane Model: A New Tool for Oncology and Radiotracer Development. J Nucl Med 2013; 54:1782-8. [DOI: 10.2967/jnumed.112.117150] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
25
|
Signaling determinants of glioma cell invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 986:121-41. [PMID: 22879067 DOI: 10.1007/978-94-007-4719-7_7] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
26
|
Li Z, Shigeoka D, Caulfield TR, Kawachi T, Qiu Y, Kamon T, Arai M, Tun HW, Yoshimitsu T. An integrated approach to the discovery of potent agelastatin A analogues for brain tumors: chemical synthesis and biological, physicochemical and CNS pharmacokinetic analyses. MEDCHEMCOMM 2013. [DOI: 10.1039/c3md00094j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
27
|
Yamaguchi Y, Shao Z, Sharif S, Du XY, Myles T, Merchant M, Harsh G, Glantz M, Recht L, Morser J, Leung LLK. Thrombin-cleaved fragments of osteopontin are overexpressed in malignant glial tumors and provide a molecular niche with survival advantage. J Biol Chem 2012. [PMID: 23204518 DOI: 10.1074/jbc.m112.362954] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Osteopontin (OPN), which is highly expressed in malignant glioblastoma (GBM), possesses inflammatory activity modulated by proteolytic cleavage by thrombin and plasma carboxypeptidase B2 (CPB2) at a highly conserved cleavage site. Full-length OPN (OPN-FL) was elevated in cerebrospinal fluid (CSF) samples from all cancer patients compared with noncancer patients. However, thrombin-cleaved OPN (OPN-R) and thrombin/CPB2-double-cleaved OPN (OPN-L) levels were markedly increased in GBM and non-GBM gliomas compared with systemic cancer and noncancer patients. Cleaved OPN constituted ∼23 and ∼31% of the total OPN in the GBM and non-GBM CSF samples, respectively. OPN-R was also elevated in GBM tissues. Thrombin-antithrombin levels were highly correlated with cleaved OPN, but not OPN-FL, suggesting that the cleaved OPN fragments resulted from increased thrombin and CPB2 in this extracellular compartment. Levels of VEGF and CCL4 were increased in CSF of GBM and correlated with the levels of cleaved OPN. GBM cell lines were more adherent to OPN-R and OPN-L than OPN-FL. Adhesion to OPN altered gene expression, in particular genes involved with cellular processes, cell cycle regulation, death, and inflammation. OPN and its cleaved forms promoted motility of U-87 MG cells and conferred resistance to apoptosis. Although functional mutation of the RGD motif in OPN largely abolished these functions, OPN(RAA)-R regained significant cell binding and signaling function, suggesting that the SVVYGLR motif in OPN-R may substitute for the RGD motif if the latter becomes inaccessible. OPN cleavage contributes to GBM development by allowing more cells to bind in niches where they acquire anti-apoptotic properties.
Collapse
Affiliation(s)
- Yasuto Yamaguchi
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Formolo CA, Williams R, Gordish-Dressman H, MacDonald TJ, Lee NH, Hathout Y. Secretome signature of invasive glioblastoma multiforme. J Proteome Res 2011; 10:3149-59. [PMID: 21574646 DOI: 10.1021/pr200210w] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The incurability of malignant glioblastomas is mainly attributed to their highly invasive nature coupled with resistance to chemo- and radiation therapy. Because invasiveness is partially dictated by the proteins these tumors secrete we used SILAC to characterize the secretomes of four glioblastoma cell lines (LN18, T98, U118 and U87). Although U87 and U118 cells both secreted high levels of well-known invasion promoting proteins, a Matrigel invasion assay showed U87 cells to be eight times more invasive than U118 cells, suggesting that additional proteins secreted by U87 cells may contribute to the highly invasive phenotype. Indeed, we identified a number of proteins highly or exclusively expressed by U87 cells as compared to the less invasive cell lines. The most striking of these include ADAM9, ADAM10, cathepsin B, cathepsin L1, osteopontin, neuropilin-1, semaphorin-7A, suprabasin, and chitinase-3-like protein 1. U87 cells also expressed significantly low levels of some cell adhesion proteins such as periostin and EMILIN-1. Correlation of secretome profiles with relative levels of invasiveness using Pavlidis template matching further indicated potential roles for these proteins in U87 glioblastoma invasion. Antibody inhibition of CH3L1 reduced U87 cell invasiveness by 30%.
Collapse
Affiliation(s)
- Catherine A Formolo
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Avenue NW, Washington, D.C. 20010, USA
| | | | | | | | | | | |
Collapse
|
29
|
Pang H, Cai L, Yang Y, Chen X, Sui G, Zhao C. Knockdown of Osteopontin Chemosensitizes MDA-MB-231 Cells to Cyclophosphamide by Enhancing Apoptosis Through Activating p38 MAPK Pathway. Cancer Biother Radiopharm 2011; 26:165-73. [PMID: 21539449 DOI: 10.1089/cbr.2010.0838] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hui Pang
- Department of Oncology Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Li Cai
- Department of Oncology Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yanmei Yang
- Cancer Research Institute of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xuesong Chen
- Department of Oncology Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guangjie Sui
- Department of Oncology Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Changhong Zhao
- Department of Oncology Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
30
|
Likui W, Hong W, Shuwen Z, Yuangang Y, Yan W. The potential of osteopontin as a therapeutic target for human colorectal cancer. J Gastrointest Surg 2011; 15:652-9. [PMID: 21318445 DOI: 10.1007/s11605-011-1445-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 01/28/2011] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Osteopontin (OPN), a phosphorylated glycoprotein, is involved in tumor progression and metastasis. Previously, we have reported that high OPN mRNA expression level possessed clinicopathological or prognostic significance in human colorectal cancer (CRC). The aim of this study is to investigate whether OPN can serve as a novel molecular target for CRC therapy. MATERIAL AND METHODS Western Blot assay was performed to detect the expression of OPN protein in 18 CRC and corresponding nontumor colon tissue samples. RNA interference (RNAi) was employed to knockdown endogenous OPN expression in CRC cell line (LoVo). MTT, colony formation, and tumorigenicity assays were performed to analyze the effect of OPN downregulation on the in vitro and in vivo proliferation of CRC cells. Wound healing and Matrigel invasion assays were performed to analyze the effect of OPN downregulation on migration and invasion of CRC cells. A clonogenic cell survival assay after radiation was performed to analyze the effect of OPN downregulation on the radiosensitivity of CRC cells. RESULTS The relative level of OPN protein expression in CRC tissues was significantly higher than that in corresponding nontumor colon tissues (P < 0.05). We found that RNAi-mediated OPN downregulation could inhibit not only in vitro proliferation but also in vivo tumorigenicity of CRC cells. In addition, OPN downregulation could suppress in vitro invasion capacity and enhance in vitro radiosensitivity of CRC cells, which might be associated with decreased levels of MMP-2 and -9 expression. CONCLUSION RNAi-targeting OPN could inhibit proliferation, invasion and enhance radiosensitivity of human CRC cells. Therefore, OPN could serve as a novel molecular target for gene therapy of CRC.
Collapse
Affiliation(s)
- Wang Likui
- Department of Infection, Beijing Friendship Hospital, Capital Medical University, 95 Yong-An Road, Beijing 100050, People's Republic of China.
| | | | | | | | | |
Collapse
|
31
|
Dentin matrix protein 1 induces membrane expression of VE-cadherin on endothelial cells and inhibits VEGF-induced angiogenesis by blocking VEGFR-2 phosphorylation. Blood 2010; 117:2515-26. [PMID: 21190990 DOI: 10.1182/blood-2010-08-298810] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dentin matrix protein 1 (DMP1) is a member of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family, a group of proteins initially described as mineralized extracellular matrices components. More recently, SIBLINGs have been implicated in several key steps of cancer progression, including angiogenesis. Although proangiogenic activities have been demonstrated for 2 SIBLINGs, the role of DMP1 in angiogenesis has not yet been addressed. We demonstrate that this extracellular matrix protein induced the expression of vascular endothelial cadherin (VE-cadherin), a key regulator of intercellular junctions and contact inhibition of growth of endothelial cells that is also known to modulate vascular endothelial growth factor receptor 2 (VEGFR-2) activity, the major high-affinity receptor for VEGF. DMP1 induced VE-cadherin and p27(Kip1) expression followed by cell-cycle arrest in human umbilical vein endothelial cells (HUVECs) in a CD44-dependent manner. VEGF-induced proliferation, migration, and tubulogenesis responses were specifically blocked on DMP1 pretreatment of HUVECs. Indeed, after VE-cadherin induction, DMP1 inhibited VEGFR-2 phosphorylation and Src-mediated signaling. However, DMP1 did not interfere with basic fibroblast growth factor-induced angiogenesis. In vivo, DMP1 significantly reduced laser-induced choroidal neovascularization lesions and tumor-associated angiogenesis. These data enable us to put DMP1 on the angiogenic chessboard for the first time and to identify this protein as a new specific inhibitor of VEGF-induced angiogenesis.
Collapse
|
32
|
Li J, Di C, Mattox AK, Wu L, Adamson DC. The future role of personalized medicine in the treatment of glioblastoma multiforme. Pharmgenomics Pers Med 2010; 3:111-27. [PMID: 23226047 PMCID: PMC3513213 DOI: 10.2147/pgpm.s6852] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) remains one of the most malignant primary central nervous system tumors. Personalized therapeutic approaches have not become standard of care for GBM, but science is fast approaching this goal. GBM's heterogeneous genomic landscape and resistance to radiotherapy and chemotherapy make this tumor one of the most challenging to treat. Recent advances in genome-wide studies and genetic profiling show that there is unlikely to be a single genetic or cellular event that can be effectively targeted in all patients. Instead, future therapies will likely require personalization for each patient's tumor genotype or proteomic profile. Over the past year, many investigations specifically focused simultaneously on strategies to target oncogenic pathways, angiogenesis, tumor immunology, epigenomic events, glioma stem cells (GSCs), and the highly migratory glioma cell population. Combination therapy targeting multiple pathways is becoming a fast growing area of research, and many studies put special attention on small molecule inhibitors. Because GBM is a highly vascular tumor, therapy that directs monoclonal antibodies or small molecule tyrosine kinase inhibitors toward angiogenic factors is also an area of focus for the development of new therapies. Passive, active, and adoptive immunotherapies have been explored by many studies recently, and epigenetic regulation of gene expression with microRNAs is also becoming an important area of study. GSCs can be useful targets to stop tumor recurrence and proliferation, and recent research has found key molecules that regulate GBM cell migration that can be targeted by therapy. Current standard of care for GBM remains nonspecific; however, pharmacogenomic studies are underway to pave the way for patient-specific therapies that are based on the unique aberrant pathways in individual patients. In conclusion, recent studies in GBM have found many diverse molecular targets possible for therapy. The next obstacle in treating this fatal tumor is ascertaining which molecules in each patient should be targeted and how best to target them, so that we can move our current nonspecific therapies toward the realm of personalized medicine.
Collapse
Affiliation(s)
- Jing Li
- Preston Robert Tisch Brain Tumor Center, Duke Medical Center, Durham, North Carolina, USA
- Department of Surgery (Neurosurgery), Duke Medical Center, Durham, North Carolina, USA
| | - Chunhui Di
- Preston Robert Tisch Brain Tumor Center, Duke Medical Center, Durham, North Carolina, USA
- Department of Surgery (Neurosurgery), Duke Medical Center, Durham, North Carolina, USA
| | - Austin K Mattox
- Preston Robert Tisch Brain Tumor Center, Duke Medical Center, Durham, North Carolina, USA
- Department of Surgery (Neurosurgery), Duke Medical Center, Durham, North Carolina, USA
| | - Linda Wu
- Preston Robert Tisch Brain Tumor Center, Duke Medical Center, Durham, North Carolina, USA
- Department of Surgery (Neurosurgery), Duke Medical Center, Durham, North Carolina, USA
| | - D Cory Adamson
- Preston Robert Tisch Brain Tumor Center, Duke Medical Center, Durham, North Carolina, USA
- Department of Surgery (Neurosurgery), Duke Medical Center, Durham, North Carolina, USA
- Department of Neurobiology, Duke Medical Center, Durham, North Carolina, USA
- Neurosurgery Section, Durham VA Medical Center, Durham, North Carolina, USA
| |
Collapse
|
33
|
Atai NA, Bansal M, Lo C, Bosman J, Tigchelaar W, Bosch KS, Jonker A, De Witt Hamer PC, Troost D, McCulloch CA, Everts V, Van Noorden CJF, Sodek J. Osteopontin is up-regulated and associated with neutrophil and macrophage infiltration in glioblastoma. Immunology 2010; 132:39-48. [PMID: 20722758 DOI: 10.1111/j.1365-2567.2010.03335.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteopontin (OPN) is a glycophosphoprotein with multiple intracellular and extracellular functions. In vitro, OPN enhances migration of mouse neutrophils and macrophages. In cancer, extracellular OPN facilitates migration of cancer cells via its RGD sequence. The present study was designed to investigate whether osteopontin is responsible for neutrophil and macrophage infiltration in human cancer and in particular in glioblastoma. We found that in vitro mouse neutrophil migration was RGD-dependent. In silico, we found that the OPN gene was one of the 5% most highly expressed genes in 20 out of 35 cancer microarray data sets in comparison with normal tissue in at least 30% of cancer patients. In some types of cancer, such as ovarian cancer, lung cancer and melanoma, the OPN gene was one of those with the highest expression levels in at least 90% of cancer patients. In glioblastoma, the most invasive type of brain tumours/glioma, but not in lower grades of glioma it was one of the 5% highest expressed genes in 90% of patients. In situ, we found increased protein levels of OPN in human glioblastoma versus normal human brain confirming in silico results. OPN protein expression was co-localized with neutrophils and macrophages. In conclusion, OPN in tumours not only induces migration of cancer cells but also of leucocytes.
Collapse
Affiliation(s)
- Nadia A Atai
- Department of Cell Biology and Histology, Academic Medical Centre, University of Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yin B, Li KH, An T, Chen T, Peng XZ. Nectin-like Molecule 1 Inhibits the Migration and Invasion of U251 Glioma Cells by Regulating the Expression of An Extracellular Matrix Protein Osteopontin. ACTA ACUST UNITED AC 2010; 25:100-4. [DOI: 10.1016/s1001-9294(10)60030-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
35
|
Yan W, Qian C, Zhao P, Zhang J, Shi L, Qian J, Liu N, Fu Z, Kang C, Pu P, You Y. Expression pattern of osteopontin splice variants and its functions on cell apoptosis and invasion in glioma cells. Neuro Oncol 2010; 12:765-75. [PMID: 20511184 DOI: 10.1093/neuonc/noq006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Osteopontin (OPN) is widely overexpressed in various cancers, including gliomas, and plays an important role in tumorigenesis. However, the expression pattern and functions of OPN splice variants expressed in gliomas remain unclear. The aims of our current study were to examine the expression pattern and functions of OPN splice variants in gliomas. In present study, the mRNA levels of OPN splice variants are markedly increased in gliomas tissues, and all OPN splice variants were also found in U251 and U87 cells. Furthermore, knock-down and regain of function experiments were designed to explore the functions of OPN splice variants in U251 and U87 cells. Lentiviral vectors of OPN small interference RNA (siRNA) targeting all three endogenous mRNAs of OPN and OPN splice variants synonymous mutant that were not silenced by OPN siRNA were constructed. Our results showed that all OPN splice variants synonymous mutant-protected glioma cells from apoptosis induced by OPN siRNA through alteration of the levels of Bcl-2 family proteins and OPN-b Mu elicted a significant effect. Both OPN-a Mu and -c Mu promoted glioma cell invasion through alteration of the levels of uPA, MMP-2, and MMP-9 expressions and the activities of MMP-2 and MMP-9 via activation PI-3K/AKT/NF-kappaB signaling pathway. Moreover, OPN-c Mu showed the strongest effect on glioma cell invasion, while OPN-b Mu showed no effect on the invasion of U251 and U87 cells. Thus, different splice variants of OPN have divergent functions in regulating apoptosis and invasion of glioma cells, which broadens their importance in glioma biotherapy.
Collapse
Affiliation(s)
- Wei Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|