1
|
Zhang J, Zhang J, Yang C. Autophagy in brain tumors: molecular mechanisms, challenges, and therapeutic opportunities. J Transl Med 2025; 23:52. [PMID: 39806481 PMCID: PMC11727735 DOI: 10.1186/s12967-024-06063-0] [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: 07/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Autophagy is responsible for maintaining cellular balance and ensuring survival. Autophagy plays a crucial role in the development of diseases, particularly human cancers, with actions that can either promote survival or induce cell death. However, brain tumors contribute to high levels of both mortality and morbidity globally, with resistance to treatments being acquired due to genetic mutations and dysregulation of molecular mechanisms, among other factors. Hence, having knowledge of the role of molecular processes in the advancement of brain tumors is enlightening, and the current review specifically examines the role of autophagy. The discussion would focus on the molecular pathways that control autophagy in brain tumors, and its dual role as a tumor suppressor and a supporter of tumor survival. Autophagy can control the advancement of different types of brain tumors like glioblastoma, glioma, and ependymoma, demonstrating its potential for treatment. Autophagy mechanisms can influence metastasis and drug resistance in glioblastoma, and there is a complex interplay between autophagy and cellular responses to stress like hypoxia and starvation. Autophagy can inhibit the growth of brain tumors by promoting apoptosis. Hence, focusing on autophagy could offer fresh perspectives on creating successful treatments.
Collapse
Affiliation(s)
- Jiarui Zhang
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinan Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| |
Collapse
|
2
|
Suh HN, Choi GE. Wnt signaling in the tumor microenvironment: A driver of brain tumor dynamics. Life Sci 2024; 358:123174. [PMID: 39471897 DOI: 10.1016/j.lfs.2024.123174] [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: 07/29/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
The Wnt signaling pathway is important for cell growth and development in the central nervous system and its associated vasculature. Thus, it is an interesting factor for establishing anti-brain cancer therapy. However, simply inhibiting the Wnt signaling pathway in patients with brain tumors is not an effective anti-cancer therapy. Due to their complex microenvironment, which comprises various cell types and signaling molecules, brain tumors pose significant challenges. It is important to understand the interplay between tumor cells and the microenvironment for developing effective therapeutic strategies for both benign and malignant brain tumors. Thus, this research focused on the role of the tumor microenvironment (TME) in brain tumor progression, particularly the involvement of Wnt-dependent signaling pathways. The brain parenchyma comprises neurons, glia, endothelial cells, and other extracellular matrix elements that can contribute to the TME. The TME components can secrete Wnt ligands or associated molecules, resulting in the aberrant activation of the Wnt signaling pathway, followed by tumor progression and therapeutic resistance. Therefore, it is essential to understand the intricate crosstalk between the Wnt signaling pathway and the TME in developing targeted therapies. This review aimed to elucidate the complexities of the brain TME and its interactions with the Wnt signaling pathways to improve treatment outcomes and our understanding of brain tumor biology.
Collapse
Affiliation(s)
- Han Na Suh
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeonbukdo 56212, Republic of Korea.
| | - Gee Euhn Choi
- Laboratory of Veterinary Biochemistry, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, South Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, South Korea.
| |
Collapse
|
3
|
Li L, Barash U, Ilan N, Farhoud M, Zhang X, Vlodavsky I, Li JP. A New Synthesized Dicarboxylated Oxy-Heparin Efficiently Attenuates Tumor Growth and Metastasis. Cells 2024; 13:211. [PMID: 38334603 PMCID: PMC10854774 DOI: 10.3390/cells13030211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Heparanase (Hpa1) is expressed by tumor cells and cells of the tumor microenvironment and functions to remodel the extracellular matrix (ECM) and regulate the bioavailability of ECM-bound factors that support tumor growth. Heparanase expression is upregulated in human carcinomas, sarcomas, and hematological malignancies, correlating with increased tumor metastasis, vascular density, and shorter postoperative survival of cancer patients, and encouraging the development of heparanase inhibitors as anti-cancer drugs. Among these are heparin/HS mimetics, the only heparanase-inhibiting compounds that are being evaluated in clinical trials. We have synthesized dicarboxylated oxy-heparins (DCoxHs) containing three carboxylate groups per split residue (DC-Hep). The resulting lead compound (termed XII) was upscaled, characterized, and examined for its effectiveness in tumor models. Potent anti-tumorigenic effects were obtained in models of pancreatic carcinoma, breast cancer, mesothelioma, and myeloma, yielding tumor growth inhibition (TGI) values ranging from 21 to 70% and extending the survival time of the mice. Of particular significance was the inhibition of spontaneous metastasis in an orthotopic model of breast carcinoma following resection of the primary tumor. It appears that apart from inhibition of heparanase enzymatic activity, compound XII reduces the levels of heparanase protein and inhibits its cellular uptake and activation. Heparanase-dependent and -independent effects of XII are being investigated. Collectively, our pre-clinical studies with compound XII strongly justify its examination in cancer patients.
Collapse
Affiliation(s)
- Li Li
- Shenzhen Hepalink Pharmaceutical Group Co., Ltd., Shenzhen 518057, China;
| | - Uri Barash
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 3525422, Israel; (U.B.); (N.I.); (M.F.)
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 3525422, Israel; (U.B.); (N.I.); (M.F.)
| | - Malik Farhoud
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 3525422, Israel; (U.B.); (N.I.); (M.F.)
| | - Xiao Zhang
- Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden;
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 3525422, Israel; (U.B.); (N.I.); (M.F.)
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, SciLifeLab, Uppsala University, 751 23 Uppsala, Sweden
| |
Collapse
|
4
|
Kong W, Zhang G, Wang Y, Zhang J, Ding T, Chen D, Pan Y, Yi R, Yin X, Wang X. Analysis of Expression Pattern and Prognostic Value of the Heparanase in Breast Cancer Through CD274/CTLA-4 Immune Checkpoint Proteins. Technol Cancer Res Treat 2024; 23:15330338241281285. [PMID: 39248214 PMCID: PMC11388313 DOI: 10.1177/15330338241281285] [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] [Indexed: 09/10/2024] Open
Abstract
Objectives: Heparanase (HPSE), an endoglycosidase that cleaves heparan sulfate, regulates various biological processes related to tumor progression. We explore the prognostic value of HPSE and its relationship with immunotherapy response in patients with breast cancer, to improve the effectiveness of immunotherapy and increase the survival outcomes. Methods: In the study, we explored the prognostic value of HPSE through the The Cancer Genome Atlas (TCGA) database. By using the single-sample gene set enrichment analysis (ssGSEA) method, we measured the infiltration levels of 24 immune cell types in the tumor microenvironment. Cancer Therapeutics Response Portal (CTRP) and PRISM datasets provide the area under the dose-response curve (AUC) to measure drug sensitivity. Using nomograms, we predicted overall survival ability. In vivo studies, we investigated the relationship between HPSE and immune checkpoint proteins and pro-inflammatory cytokines by immunohistochemistry of Triple-Negative Breast Cancer tumors in mice. Results: Our model demonstrated that the integrating of HPSE with the clinical stage effectively predicts patients' survival time, highlighting high HPSE expression as a prognostic risk factor for breast cancer. Then the Receiver Operating Characteristic (ROC) curve [AUC of 1 year = 0.747, AUC of 3 years = 0.731] and Decision Curve Analysis (DCA) curve illustrated the satisfactory discriminative capacity of our model, emphasizing its valuable clinical applicability. Immune-related results showed that HPSE correlates strongly with immune infiltrating cells, immune-related genes, and the anti-cancer immunity cycle. In vivo studies have demonstrated that HPSE in breast cancer is associated with increased expression of immune checkpoint proteins CD274 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and is positively correlated with the pro-inflammatory cytokine TNF-α. Meanwhile, we analyzed the 11 types of drugs that are sensitive to the HPSE gene. Conclusion: Our results show that HPSE can serve as an effective biomarker to predict the prognosis of breast cancer patients and reflect the impact of immunotherapy.
Collapse
Affiliation(s)
- Weijia Kong
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ganlin Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yue Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jiahui Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Tongjing Ding
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Dong Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuancan Pan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Runxi Yi
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohui Yin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Wang C, Huang Y, Jia B, Huang Y, Chen J. Heparanase promotes malignant phenotypes of human oral squamous carcinoma cells by regulating the epithelial-mesenchymal transition-related molecules and infiltrated levels of natural killer cells. Arch Oral Biol 2023; 154:105775. [PMID: 37481997 DOI: 10.1016/j.archoralbio.2023.105775] [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: 04/19/2023] [Revised: 07/11/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVES The aim of the present study was to explore the functional role of heparanase (HPSE) and investigate the effect of HPSE on epithelial-mesenchymal transition (EMT) and Tumor-infiltrating activated natural killer cells in oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS human oral squamous carcinoma (SCC-25) cells were transfected with HPSE-specific small interfering RNA. Cell Counting Kit-8 assay was performed to examine cell proliferation, while flow cytometry was performed to analyze the cell cycle. Scratch assay was conducted to analyze cell migration, followed by Transwell assay to determine cell invasion. Real-Time Polymerase Chain Reaction and Western-blot assays were performed to measure epithelial-mesenchymal transition protein expression. RNA Sequencing analysis and tumor-infiltrating immune cells estimation were performed to elucidate the effect of HPSE on OSCC. RESULTS Knockdown of HPSE expression decreased the proliferation rate of SCC-25 cells resulting in a significant elevation in cell percentage at the Gap phase 0/Gap phase 1 phase by suppressed cell migration and invasion. The E-cadherin messenger RNA and protein expression increased while Snail and Vimentin expression decreased. RNA Sequencing analysis performed between small interfering RNA and negative control groups identified 42 differentially expressed genes, such as syndecan binding protein, RAB11A, member RAS oncogene family, and DDB1 and CUL4 associated factor 15. CONCLUSIONS These results indicated that knockdown of HPSE suppressed SCC-25 cell proliferation, invasion, migration, and epithelial-mesenchymal transition, possibly via syndecan binding protein and RAB11A, member RAS oncogene family. Moreover, HPSE regulates the infiltrated levels of natural killer cells activated, possibly via DDB1 and CUL4 associated factor 15.
Collapse
Affiliation(s)
- Changlin Wang
- Department of Stomatology, Yancheng Third People's Hospital,The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng 224001 China
| | - Yisheng Huang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280 China
| | - Bo Jia
- Stomatological Hospital, Southern Medical University, Guangzhou 510280 China
| | - Yuhua Huang
- Department of Stomatology, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou 510120, China.
| | - Jun Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510280 China.
| |
Collapse
|
6
|
Vlodavsky I, Kayal Y, Hilwi M, Soboh S, Sanderson RD, Ilan N. Heparanase-A single protein with multiple enzymatic and nonenzymatic functions. PROTEOGLYCAN RESEARCH 2023; 1:e6. [PMID: 37547889 PMCID: PMC10398610 DOI: 10.1002/pgr2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 08/08/2023]
Abstract
Heparanase (Hpa1) is expressed by tumor cells and cells of the tumor microenvironment and functions extracellularly to remodel the extracellular matrix (ECM) and regulate the bioavailability of ECM-bound factors, augmenting, among other effects, gene transcription, autophagy, exosome formation, and heparan sulfate (HS) turnover. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth, metastasis, and chemoresistance. The enzyme appears to fulfill some normal functions associated, for example, with vesicular traffic, lysosomal-based secretion, autophagy, HS turnover, and gene transcription. It activates cells of the innate immune system, promotes the formation of exosomes and autophagosomes, and stimulates signal transduction pathways via enzymatic and nonenzymatic activities. These effects dynamically impact multiple regulatory pathways that together drive tumor growth, dissemination, and drug resistance as well as inflammatory responses. The emerging premise is that heparanase expressed by tumor cells, immune cells, endothelial cells, and other cells of the tumor microenvironment is a key regulator of the aggressive phenotype of cancer, an important contributor to the poor outcome of cancer patients and a valid target for therapy. So far, however, antiheparanase-based therapy has not been implemented in the clinic. Unlike heparanase, heparanase-2 (Hpa2), a close homolog of heparanase (Hpa1), does not undergo proteolytic processing and hence lacks intrinsic HS-degrading activity, the hallmark of heparanase. Hpa2 retains the capacity to bind heparin/HS and exhibits an even higher affinity towards HS than heparanase, thus competing for HS binding and inhibiting heparanase enzymatic activity. It appears that Hpa2 functions as a natural inhibitor of Hpa1 regulates the expression of selected genes that maintain tissue hemostasis and normal function, and plays a protective role against cancer and inflammation, together emphasizing the significance of maintaining a proper balance between Hpa1 and Hpa2.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Yasmin Kayal
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Maram Hilwi
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Soaad Soboh
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Ralph D. Sanderson
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Neta Ilan
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| |
Collapse
|
7
|
Behrooz AB, Latifi-Navid H, Nezhadi A, Świat M, Los M, Jamalpoor Z, Ghavami S. Molecular mechanisms of microRNAs in glioblastoma pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119482. [PMID: 37146725 DOI: 10.1016/j.bbamcr.2023.119482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Glioblastoma (GBM) is human's most prevalent and severe brain cancer. Epigenetic regulators, micro(mi)RNAs, significantly impact cellular health and disease because of their wide range of targets and functions. The "epigenetic symphony" in which miRNAs perform is responsible for orchestrating the transcription of genetic information. The discovery of regulatory miRNA activities in GBM biology has shown that various miRNAs play a vital role in disease onset and development. Here, we summarize our current understanding of the current state-of-the-art and latest findings regarding the interactions between miRNAs and molecular mechanisms commonly associated with GBM pathogenesis. Moreover, by literature review and reconstruction of the GBM gene regulatory network, we uncovered the connection between miRNAs and critical signaling pathways such as cell proliferation, invasion, and cell death, which provides promising hints for identifying potential therapeutic targets for the treatment of GBM. In addition, the role of miRNAs in GBM patient survival was investigated. The present review, which contains new analyses of the previous literature, may lead to new avenues to explore in the future for the development of multitargeted miRNA-based therapies for GBM.
Collapse
Affiliation(s)
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Akram Nezhadi
- Cognitive Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Maciej Świat
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Marek Los
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
8
|
Jiménez-Morales JM, Hernández-Cuenca YE, Reyes-Abrahantes A, Ruiz-García H, Barajas-Olmos F, García-Ortiz H, Orozco L, Quiñones-Hinojosa A, Reyes-González J, Del Carmen Abrahantes-Pérez M. MicroRNA delivery systems in glioma therapy and perspectives: A systematic review. J Control Release 2022; 349:712-730. [PMID: 35905783 DOI: 10.1016/j.jconrel.2022.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
Abstract
Gliomas are the deadliest of all primary brain tumors, and they constitute a serious global health problem. MicroRNAs (miRNAs) are gene expression regulators associated with glioma pathogenesis. Thus, miRNAs represent potential therapeutic agents for treating gliomas. However, miRNAs have not been established as part of the regular clinical armamentarium. This systemic review evaluates current molecular and pre-clinical studies with the aim of defining the most appealing supramolecular platform for administering therapeutic miRNA to patients with gliomas. An integrated analysis suggested that cationic lipid nanoparticles, functionalized with octa-arginine peptides, represent a potentially specific, practical, non-invasive intervention for treating gliomas. This supramolecular platform allows loading both hydrophilic (miRNA) and hydrophobic (anti-tumor drugs, like temozolomide) molecules. This systemic review is the first to describe miRNA delivery systems targeted to gliomas that integrate several types of molecules as active ingredients. Further experimental validation is warranted to confirm the practical value of miRNA delivery systems.
Collapse
Affiliation(s)
- José Marcos Jiménez-Morales
- Precision Translational Oncology Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico
| | - Yanet Elisa Hernández-Cuenca
- Precision Translational Oncology Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico
| | - Ander Reyes-Abrahantes
- Precision Translational Oncology Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico
| | - Henry Ruiz-García
- Department of Neurosurgery, Mayo Clinic, Jacksonville, United States; Brain Tumor Stem Cell Research Laboratory, Mayo Clinic, Jacksonville, United States
| | - Francisco Barajas-Olmos
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico
| | - Humberto García-Ortiz
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico
| | - Lorena Orozco
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Mayo Clinic, Jacksonville, United States; Brain Tumor Stem Cell Research Laboratory, Mayo Clinic, Jacksonville, United States
| | - Jesús Reyes-González
- Precision Translational Oncology Laboratory, National Institute of Genomic Medicine (INMEGEN), 14610 Mexico City, Mexico.
| | | |
Collapse
|
9
|
Thakur A, Faujdar C, Sharma R, Sharma S, Malik B, Nepali K, Liou JP. Glioblastoma: Current Status, Emerging Targets, and Recent Advances. J Med Chem 2022; 65:8596-8685. [PMID: 35786935 PMCID: PMC9297300 DOI: 10.1021/acs.jmedchem.1c01946] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Glioblastoma (GBM) is a highly malignant
brain tumor characterized
by a heterogeneous population of genetically unstable and highly infiltrative
cells that are resistant to chemotherapy. Although substantial efforts
have been invested in the field of anti-GBM drug discovery in the
past decade, success has primarily been confined to the preclinical
level, and clinical studies have often been hampered due to efficacy-,
selectivity-, or physicochemical property-related issues. Thus, expansion
of the list of molecular targets coupled with a pragmatic design of
new small-molecule inhibitors with central nervous system (CNS)-penetrating
ability is required to steer the wheels of anti-GBM drug discovery
endeavors. This Perspective presents various aspects of drug discovery
(challenges in GBM drug discovery and delivery, therapeutic targets,
and agents under clinical investigation). The comprehensively covered
sections include the recent medicinal chemistry campaigns embarked
upon to validate the potential of numerous enzymes/proteins/receptors
as therapeutic targets in GBM.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Chetna Faujdar
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida 201307, India
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Basant Malik
- Department of Sterile Product Development, Research and Development-Unit 2, Jubiliant Generics Ltd., Noida 201301, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| |
Collapse
|
10
|
Oishi T, Koizumi S, Kurozumi K. Molecular Mechanisms and Clinical Challenges of Glioma Invasion. Brain Sci 2022; 12:brainsci12020291. [PMID: 35204054 PMCID: PMC8870089 DOI: 10.3390/brainsci12020291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/17/2022] Open
Abstract
Glioma is the most common primary brain tumor, and its prognosis is poor. Glioma cells are highly invasive to the brain parenchyma. It is difficult to achieve complete resection due to the nature of the brain tissue, and tumors that invade the parenchyma often recur. The invasiveness of tumor cells has been studied from various aspects, and the related molecular mechanisms are gradually becoming clear. Cell adhesion factors and extracellular matrix factors have a strong influence on glioma invasion. The molecular mechanisms that enhance the invasiveness of glioma stem cells, which have been investigated in recent years, have also been clarified. In addition, it has been discussed from both basic and clinical perspectives that current therapies can alter the invasiveness of tumors, and there is a need to develop therapeutic approaches to glioma invasion in the future. In this review, we will summarize the factors that influence the invasiveness of glioma based on the environment of tumor cells and tissues, and describe the impact of the treatment of glioma on invasion in terms of molecular biology, and the novel therapies for invasion that are currently being developed.
Collapse
|
11
|
Multiple Irradiation Affects Cellular and Extracellular Components of the Mouse Brain Tissue and Adhesion and Proliferation of Glioblastoma Cells in Experimental System In Vivo. Int J Mol Sci 2021; 22:ijms222413350. [PMID: 34948147 PMCID: PMC8703639 DOI: 10.3390/ijms222413350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022] Open
Abstract
Intensive adjuvant radiotherapy (RT) is a standard treatment for glioblastoma multiforme (GBM) patients; however, its effect on the normal brain tissue remains unclear. Here, we investigated the short-term effects of multiple irradiation on the cellular and extracellular glycosylated components of normal brain tissue and their functional significance. Triple irradiation (7 Gy*3 days) of C57Bl/6 mouse brain inhibited the viability, proliferation and biosynthetic activity of normal glial cells, resulting in a fast brain-zone-dependent deregulation of the expression of proteoglycans (PGs) (decorin, biglycan, versican, brevican and CD44). Complex time-point-specific (24–72 h) changes in decorin and brevican protein and chondroitin sulfate (CS) and heparan sulfate (HS) content suggested deterioration of the PGs glycosylation in irradiated brain tissue, while the transcriptional activity of HS-biosynthetic system remained unchanged. The primary glial cultures and organotypic slices from triple-irradiated brain tissue were more susceptible to GBM U87 cells’ adhesion and proliferation in co-culture systems in vitro and ex vivo. In summary, multiple irradiation affects glycosylated components of normal brain extracellular matrix (ECM) through inhibition of the functional activity of normal glial cells. The changed content and pattern of PGs and GAGs in irradiated brain tissues are accompanied by the increased adhesion and proliferation of GBM cells, suggesting a novel molecular mechanism of negative side-effects of anti-GBM radiotherapy.
Collapse
|
12
|
Martinez-Morales P, Morán Cruz I, Roa-de la Cruz L, Maycotte P, Reyes Salinas JS, Vazquez Zamora VJ, Gutierrez Quiroz CT, Montiel-Jarquin AJ, Vallejo-Ruiz V. Hallmarks of glycogene expression and glycosylation pathways in squamous and adenocarcinoma cervical cancer. PeerJ 2021; 9:e12081. [PMID: 34540372 PMCID: PMC8415283 DOI: 10.7717/peerj.12081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background Dysregulation of glycogene expression in cancer can lead to aberrant glycan expression, which can promote tumorigenesis. Cervical cancer (CC) displays an increased expression of glycogenes involved in sialylation and sialylated glycans. Here, we show a comprehensive analysis of glycogene expression in CC to identify glycogene expression signatures and the possible glycosylation pathways altered. Methods First, we performed a microarray expression assay to compare glycogene expression changes between normal and cervical cancer tissues. Second, we used 401 glycogenes to analyze glycogene expression in adenocarcinoma and squamous carcinoma from RNA-seq data at the cBioPortal for Cancer Genomics. Results The analysis of the microarray expression assay indicated that CC displayed an increase in glycogenes related to GPI-anchored biosynthesis and a decrease in genes associated with chondroitin and dermatan sulfate with respect to normal tissue. Also, the glycogene analysis of CC samples by the RNA-seq showed that the glycogenes involved in the chondroitin and dermatan sulfate pathway were downregulated. Interestingly the adenocarcinoma tumors displayed a unique glycogene expression signature compared to squamous cancer that shows heterogeneous glycogene expression divided into six types. Squamous carcinoma type 5 (SCC-5) showed increased expression of genes implicated in keratan and heparan sulfate synthesis, glycosaminoglycan degradation, ganglio, and globo glycosphingolipid synthesis was related to poorly differentiated tumors and poor survival. Squamous carcinoma type 6 (SCC-6) displayed an increased expression of genes involved in chondroitin/dermatan sulfate synthesis and lacto and neolacto glycosphingolipid synthesis and was associated with nonkeratinizing squamous cancer and good survival. In summary, our study showed that CC tumors are not a uniform entity, and their glycome signatures could be related to different clinicopathological characteristics.
Collapse
Affiliation(s)
- Patricia Martinez-Morales
- CONACYT-Centro de Investigación Biomédica de Oriente, Mexican Institute of Social Security, Metepec, Puebla, México
| | - Irene Morán Cruz
- Centro de Investigación Biomédica de Oriente, Laboratory of Molecular Biology, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| | - Lorena Roa-de la Cruz
- Department of Biological Chemical Sciences, Universidad de las Américas-Puebla, San Andrés Cholula, Puebla, Mexico
| | - Paola Maycotte
- Centro de Investigación Biomédica de Oriente, Laboratory of Cell Biology, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| | - Juan Salvador Reyes Salinas
- Hospital de especialidades, General Manuel Ávila Camacho, Instituto Mexicano del Seguro Social, Puebla, Puebla, México
| | - Victor Javier Vazquez Zamora
- Hospital de especialidades, General Manuel Ávila Camacho, Instituto Mexicano del Seguro Social, Puebla, Puebla, México
| | | | - Alvaro Jose Montiel-Jarquin
- Hospital de especialidades, General Manuel Ávila Camacho, Instituto Mexicano del Seguro Social, Puebla, Puebla, México
| | - Verónica Vallejo-Ruiz
- Centro de Investigación Biomédica de Oriente, Laboratory of Molecular Biology, Instituto Mexicano del Seguro Social, Metepec, Puebla, México
| |
Collapse
|
13
|
Si J, Li W, Li X, Cao L, Chen Z, Jiang Z. Heparanase confers temozolomide resistance by regulation of exosome secretion and circular RNA composition in glioma. Cancer Sci 2021; 112:3491-3506. [PMID: 34036683 PMCID: PMC8409313 DOI: 10.1111/cas.14984] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Temozolomide (TMZ) resistance is the main challenge in the management of glioma patients. Heparanase can mediate the secretion and function of exosomes, which are considered to be a promising molecular delivery system for cancer therapy. Therefore, this study aimed to investigate whether heparanase‐mediated delivery of exosomes was related to TMZ resistance of glioma. Heparanase was upregulated in TMZ‐resistant glioma cells, and overexpression of heparanase led to increased resistance of U87 cells to TMZ. Knockdown of heparanase led to increased sensitivity of TMZ‐resistant U251 cells (U251R) cells to TMZ. Heparanase promoted the secretion of exosomes from glioma cells, and coculture with exosomes derived from heparanase knockdown U251R cells partly restored the sensitivity of U251 cells to TMZ compared with exosomes derived from si‐control transfected U251R cells. It was identified by circular RNA microarrays that hsa_circ_0042003 was upregulated in exosomes derived from U251R, which could be positively regulated by heparanase. U251R cell‐derived exosomal hsa_circ_0042003 conferred the resistance of U251 cells to TMZ. In vivo studies also showed that U251R cell‐derived exosomes induced resistance of U251 cells to TMZ, and the combination of tail‐injected exosomal si‐heparanase or exosomal si‐hsa_circ_0042003 and intraperitoneal TMZ applied to nude mice abolished TMZ resistance. Heparanase mediated the transfer of exosomal hsa_circ_0042003 from TMZ‐resistant glioma cells to drug‐sensitive cells, which contributed to the chemoresistance of glioma to TMZ.
Collapse
Affiliation(s)
- Jinchao Si
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Li
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xin Li
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lixing Cao
- Department of Perioperative Research Centre of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiqiang Chen
- Department of Perioperative Research Centre of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhi Jiang
- Department of Perioperative Research Centre of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
14
|
Barash U, Rangappa S, Mohan CD, Vishwanath D, Boyango I, Basappa B, Vlodavsky I, Rangappa KS. New Heparanase-Inhibiting Triazolo-Thiadiazoles Attenuate Primary Tumor Growth and Metastasis. Cancers (Basel) 2021; 13:cancers13122959. [PMID: 34199150 PMCID: PMC8231572 DOI: 10.3390/cancers13122959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Heparanase is an endoglycosidase that plays a critical role in tumor progression and metastasis. The expression of heparanase in the tumor microenvironment is positively correlated with the aggressiveness of the tumor and is associated with poor prognosis. In this study, we have demonstrated that a new triazole–thiadiazole-bearing small molecule showed good heparanase inhibition along with attenuation of tumor growth and metastasis. To the best of our knowledge, this is the first report showing a marked decrease in primary tumor growth in mice treated with a small molecule that inhibits heparanase enzymatic activity. Given these encouraging results, studies are underway to better elucidate the mode of action and clinical significance of triazolo–thiadiazoles. Abstract Compelling evidence ties heparanase, an endoglycosidase that cleaves heparan sulfate side (HS) chains of proteoglycans, with all steps of tumor development, including tumor initiation, angiogenesis, growth, metastasis, and chemoresistance. Moreover, heparanase levels correlate with shorter postoperative survival of cancer patients, encouraging the development of heparanase inhibitors as anti-cancer drugs. Heparanase-inhibiting heparin/heparan sulfate-mimicking compounds and neutralizing antibodies are highly effective in animal models of cancer progression, yet none of the compounds reached the stage of approval for clinical use. The present study focused on newly synthesized triazolo–thiadiazoles, of which compound 4-iodo-2-(3-(p-tolyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)phenol (4-MMI) was identified as a potent inhibitor of heparanase enzymatic activity, cell invasion, experimental metastasis, and tumor growth in mouse models. To the best of our knowledge, this is the first report showing a marked decrease in primary tumor growth in mice treated with small molecules that inhibit heparanase enzymatic activity. This result encourages the optimization of 4-MMI for preclinical and clinical studies primarily in cancer but also other indications (i.e., colitis, pancreatitis, diabetic nephropathy, tissue fibrosis) involving heparanase, including viral infection and COVID-19.
Collapse
Affiliation(s)
- Uri Barash
- Technion Integrated Cancer Center (TICC), the Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (U.B.); (I.B.)
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, BG Nagara, Nagamangala Taluk 571448, India;
| | | | - Divakar Vishwanath
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India; (D.V.); (B.B.)
| | - Ilanit Boyango
- Technion Integrated Cancer Center (TICC), the Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (U.B.); (I.B.)
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India; (D.V.); (B.B.)
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), the Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (U.B.); (I.B.)
- Correspondence: (I.V.); (K.S.R.)
| | - Kanchugarakoppal S. Rangappa
- Institution of Excellence, Vijnana Bhavan, University of Mysore, Manasagangotri, Mysore 570006, India
- Correspondence: (I.V.); (K.S.R.)
| |
Collapse
|
15
|
Jandrey EHF, Bezerra M, Inoue LT, Furnari FB, Camargo AA, Costa ÉT. A Key Pathway to Cancer Resilience: The Role of Autophagy in Glioblastomas. Front Oncol 2021; 11:652133. [PMID: 34178638 PMCID: PMC8222785 DOI: 10.3389/fonc.2021.652133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
There are no effective strategies for the successful treatment of glioblastomas (GBM). Current therapeutic modalities effectively target bulk tumor cells but leave behind marginal GBM cells that escape from the surgical margins and radiotherapy field, exhibiting high migratory phenotype and resistance to all available anti-glioma therapies. Drug resistance is mostly driven by tumor cell plasticity: a concept associated with reactivating transcriptional programs in response to adverse and dynamic conditions from the tumor microenvironment. Autophagy, or "self-eating", pathway is an emerging target for cancer therapy and has been regarded as one of the key drivers of cell plasticity in response to energy demanding stress conditions. Many studies shed light on the importance of autophagy as an adaptive mechanism, protecting GBM cells from unfavorable conditions, while others recognize that autophagy can kill those cells by triggering a non-apoptotic cell death program, called 'autophagy cell death' (ACD). In this review, we carefully analyzed literature data and conclude that there is no clear evidence indicating the presence of ACD under pathophysiological settings in GBM disease. It seems to be exclusively induced by excessive (supra-physiological) stress signals, mostly from in vitro cell culture studies. Instead, pre-clinical and clinical data indicate that autophagy is an emblematic example of the 'dark-side' of a rescue pathway that contributes profoundly to a pro-tumoral adaptive response. From a standpoint of treating the real human disease, only combinatorial therapy targeting autophagy with cytotoxic drugs in the adjuvant setting for GBM patients, associated with the development of less toxic and more specific autophagy inhibitors, may inhibit adaptive response and enhance the sensibility of glioma cells to conventional therapies.
Collapse
Affiliation(s)
| | - Marcelle Bezerra
- Molecular Oncology Center, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Frank B. Furnari
- Ludwig Institute for Cancer Research, University of California San Diego (UCSD), San Diego, CA, United States
| | | | | |
Collapse
|
16
|
Brain-invasive meningiomas: molecular mechanisms and potential therapeutic options. Brain Tumor Pathol 2021; 38:156-172. [PMID: 33903981 DOI: 10.1007/s10014-021-00399-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Meningiomas are the most commonly diagnosed benign intracranial adult tumors. Subsets of meningiomas that present with extensive invasion into surrounding brain areas have high recurrence rates, resulting in difficulties for complete resection, substantially increased mortality of patients, and are therapeutically challenging for neurosurgeons. Exciting new data have provided insights into the understanding of the molecular machinery of invasion. Moreover, clinical trials for several novel approaches have been launched. Here, we will highlight the mechanisms which govern brain invasion and new promising therapeutic approaches for brain-invasive meningiomas, including pharmacological approaches targeting three major aspects of tumor cell invasion: extracellular matrix degradation, cell adhesion, and growth factors, as well as other innovative treatments such as immunotherapy, hormone therapy, Tumor Treating Fields, and biodegradable copolymers (wafers), impregnated chemotherapy. Those ongoing studies can offer more diversified possibilities of potential treatments for brain-invasive meningiomas, and help to increase the survival benefits for patients.
Collapse
|
17
|
Yang C, Zhang S, Chang X, Huang Y, Cui D, Liu Z. MicroRNA-219a-2-3p modulates the proliferation of thyroid cancer cells via the HPSE/cyclin D1 pathway. Exp Ther Med 2021; 21:659. [PMID: 33968189 DOI: 10.3892/etm.2021.10091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/25/2021] [Indexed: 12/18/2022] Open
Abstract
Heparanase (HPSE) is an endo-β-D-glucuronidase overexpressed in different types of human cancer, and a predicted target of microRNA (miRNA/miR)-219a-2-3p in thyroid cancer. The present study aimed to investigate the potential role of HPSE and miR-219a-2-3p in thyroid cancer, and the molecular mechanism of miR-219a-2-3p regulating the proliferation of thyroid cancer cells via HPSE was confirmed. Immunohistochemistry analysis was performed to detect HPSE expression in thyroid cancer sections. In addition, reverse transcription-quantitative PCR analysis was performed to detect mRNA and miR-219a-2-3p expression levels in thyroid cancer samples and cell lines. miR-219-2-3p mimic or HPSE plasmid were transfected into B-CPAP and TPC-1 thyroid cancer cells. Furthermore, western blot analysis was performed to detect the protein expression levels of HPSE and cyclin D1. Cell cycle analysis was performed using propidium iodide staining and flow cytometry, and EdU incorporation was performed to detect cell proliferation. The results demonstrated that high HPSE expression was significantly associated with tumor size, extracapsular invasion and lymph node metastasis. Notably, a statistically negative correlation was observed between HPSE mRNA expression and miR-219a-2-3p expression in thyroid cancer tumors, as well as in thyroid cancer cell lines. When exogenously expressed in B-CPAP and TPC-1 cells, miR-219a-2-3p induced cell cycle arrest at the G0/G1 phase and decreased the percentage of proliferating cells. Furthermore, HPSE and cyclin D1 protein expression decreased following transfection with miR-219a-2-3p. Notably, when HPSE was ectopically expressed in miR-219a-2-3p transfected cells, cyclin D1 expression and the number of proliferative cells increased. Taken together, these results suggest that HPSE contributes to the proliferation of thyroid cancer cells. In addition, miR-219a-2-3p was confirmed to target HPSE and inhibit cell proliferation, which was associated with cyclin D1 suppression-mediated cell cycle arrest.
Collapse
Affiliation(s)
- Chuanjia Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Siyang Zhang
- Science and Experiment Center, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Xiaoying Chang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yonglian Huang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dongxu Cui
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
18
|
Agelidis A, Turturice BA, Suryawanshi RK, Yadavalli T, Jaishankar D, Ames J, Hopkins J, Koujah L, Patil CD, Hadigal SR, Kyzar EJ, Campeau A, Wozniak JM, Gonzalez DJ, Vlodavsky I, Li JP, Perkins DL, Finn PW, Shukla D. Disruption of innate defense responses by endoglycosidase HPSE promotes cell survival. JCI Insight 2021; 6:144255. [PMID: 33621216 PMCID: PMC8119219 DOI: 10.1172/jci.insight.144255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/18/2021] [Indexed: 01/03/2023] Open
Abstract
The drive to withstand environmental stresses and defend against invasion is a universal trait extant in all forms of life. While numerous canonical signaling cascades have been characterized in detail, it remains unclear how these pathways interface to generate coordinated responses to diverse stimuli. To dissect these connections, we followed heparanase (HPSE), a protein best known for its endoglycosidic activity at the extracellular matrix but recently recognized to drive various forms of late-stage disease through unknown mechanisms. Using herpes simplex virus-1 (HSV-1) infection as a model cellular perturbation, we demonstrate that HPSE acts beyond its established enzymatic role to restrict multiple forms of cell-intrinsic defense and facilitate host cell reprogramming by the invading pathogen. We reveal that cells devoid of HPSE are innately resistant to infection and counteract viral takeover through multiple amplified defense mechanisms. With a unique grasp of the fundamental processes of transcriptional regulation and cell death, HPSE represents a potent cellular intersection with broad therapeutic potential.
Collapse
Affiliation(s)
- Alex Agelidis
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | - Benjamin A. Turturice
- Department of Microbiology and Immunology
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | | - Dinesh Jaishankar
- Department of Ophthalmology and Visual Sciences, and
- Department of Dermatology, Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Joshua Ames
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | - James Hopkins
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | - Lulia Koujah
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| | | | | | - Evan J. Kyzar
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Anaamika Campeau
- Department of Pharmacology and
- Skaggs School of Pharmacy, UCSD, San Diego, La Jolla, California, USA
| | - Jacob M. Wozniak
- Department of Pharmacology and
- Skaggs School of Pharmacy, UCSD, San Diego, La Jolla, California, USA
| | - David J. Gonzalez
- Department of Pharmacology and
- Skaggs School of Pharmacy, UCSD, San Diego, La Jolla, California, USA
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Jin-ping Li
- Department of Medical Biochemistry and Microbiology, University of Uppsala, Uppsala, Sweden
| | - David L. Perkins
- Division of Nephrology, Department of Medicine, and
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Patricia W. Finn
- Department of Microbiology and Immunology
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology
- Department of Ophthalmology and Visual Sciences, and
| |
Collapse
|
19
|
Agelidis A, Suryawanshi RK, Patil CD, Campeau A, Gonzalez DJ, Shukla D. Dissociation of DNA damage sensing by endoglycosidase HPSE. iScience 2021; 24:102242. [PMID: 33748723 PMCID: PMC7957091 DOI: 10.1016/j.isci.2021.102242] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/25/2022] Open
Abstract
Balance between cell proliferation and elimination is critical in handling threats both exogenous and of internal dysfunction. Recent work has implicated a conserved but poorly understood endoglycosidase heparanase (HPSE) in the restriction of innate defense responses, yet biochemical mediators of these key functions remained unclear. Here, an unbiased immunopurification proteomics strategy is employed to identify and rank uncharacterized interactions between HPSE and mediators of canonical signaling pathways linking cell cycle and stress responses. We demonstrate with models of genotoxic stress including herpes simplex virus infection and chemotherapeutic treatment that HPSE dampens innate responses to double-stranded DNA breakage by interfering with signal transduction between initial sensors and downstream mediators. Given the long-standing recognition of HPSE in driving late-stage inflammatory disease exemplified by tissue destruction and cancer metastasis, modulation of this protein with control over the DNA damage response imparts a unique strategy in the development of unconventional multivalent therapy. HPSE binds key proteins at interface of DNA damage signaling and IFN responses Nuclear translocation of DNA damage transducer ATM is enhanced in absence of HPSE Cells lacking HPSE display enhanced sensitivity to DNA damage-induced death HPSE interfaces with regulators of DNA damage response to influence cell fate
Collapse
Affiliation(s)
- Alex Agelidis
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rahul K. Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chandrashekhar D. Patil
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Anaamika Campeau
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy, University of California, San Diego, La Jolla, CA 92093, USA
| | - David J. Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Skaggs School of Pharmacy, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Corresponding author
| |
Collapse
|
20
|
Wang Z, Shi Y, Ying C, Jiang Y, Hu J. Hypoxia-induced PLOD1 overexpression contributes to the malignant phenotype of glioblastoma via NF-κB signaling. Oncogene 2021; 40:1458-1475. [PMID: 33420370 PMCID: PMC7906902 DOI: 10.1038/s41388-020-01635-y] [Citation(s) in RCA: 32] [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: 09/12/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
Procollagen lysyl hydroxylase 1 (PLOD1) is highly expressed in malignant tumors such as esophageal squamous cell carcinoma, gastric cancer, and colorectal cancer. Bioinformatics analysis revealed that PLOD1 is associated with the progression of GBM, particularly the most malignant mesenchymal subtype (MES). Moreover, in the TCGA and CGGA datasets, the mean survival time of patients with high PLOD1 expression was significantly shorter than that of patients with low expression. The clinical samples confirmed this result. Therefore, we aimed to investigate the effect of PLOD1 on the development of mesenchymal GBM in vitro and in vivo and its possible mechanisms. Molecular experiments were conducted on the patient-derived glioma stem cells and found that PLOD1 expressed higher in tumor tissues and cancer cell lines of patients with GBM, especially in the MES. PLOD1 also enhanced tumor viability, proliferation, migration, and promoted MES transition while inhibited apoptosis. Tumor xenograft results also indicated that PLOD1 overexpression significantly promotes malignant behavior of tumors. Mechanistically, bioinformatics analysis further revealed that PLOD1 expression was closely associated with the NF-κB signaling pathway. Besides, we also found that hypoxic environments also enhanced the tumor-promoting effects of PLOD1. In conclusion, overexpression of PLOD1 may be an important factor in the enhanced invasiveness and MES transition of GBM. Thus, PLOD1 is a potential treatment target for mesenchymal GBM or even all GBM.
Collapse
Affiliation(s)
- Zhenlin Wang
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, China
| | - Yuping Shi
- Department of Nephrology, Shanghai TongRen Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, China
| | - Chenting Ying
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, China
| | - Yang Jiang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Jiangfeng Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, China.
| |
Collapse
|
21
|
Patel D, Wairkar S, Yergeri MC. Current Developments in Targeted Drug Delivery Systems for Glioma. Curr Pharm Des 2021; 26:3973-3984. [PMID: 32329681 DOI: 10.2174/1381612826666200424161929] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/01/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Glioma is one of the most commonly observed tumours, representing about 75% of brain tumours in the adult population. Generally, glioma treatment includes surgical resection followed by radiotherapy and chemotherapy. The current chemotherapy for glioma involves the use of temozolomide, doxorubicin, monoclonal antibodies, etc. however, the clinical outcomes in patients are not satisfactory. Primarily, the blood-brain barrier hinders these drugs from reaching the target leading to the recurrence of glioma post-surgery. In addition, these drugs are not target-specific and affect the healthy cells of the body. Therefore, glioma-targeted drug delivery is essential to reduce the rate of recurrence and treat the condition with more reliable alternatives. METHODS A literature search was conducted to understand glioma pathophysiology, its current therapeutic approaches for targeted delivery using databases like Pub Med, Web of Science, Scopus, and Google Scholar, etc. Results: This review gives an insight to challenges associated with current treatments, factors influencing drug delivery in glioma, and recent advancements in targeted drug delivery. CONCLUSION The promising results could be seen with nanotechnology-based approaches, like polymeric, lipidbased, and hybrid nanoparticles in the treatment of glioma. Biotechnological developments, such as carrier peptides and gene therapy, are future prospects in glioma therapy. Therefore, these targeted delivery systems will be beneficial in clinical practices for glioma treatment.
Collapse
Affiliation(s)
- Dhrumi Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400056, India
| | - Mayur C Yergeri
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400056, India
| |
Collapse
|
22
|
Pasqualini C, Kozaki T, Bruschi M, Nguyen THH, Minard-Colin V, Castel D, Grill J, Ginhoux F. Modeling the Interaction between the Microenvironment and Tumor Cells in Brain Tumors. Neuron 2020; 108:1025-1044. [PMID: 33065047 DOI: 10.1016/j.neuron.2020.09.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/26/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Despite considerable recent advances in understanding and treating many other cancers, malignant brain tumors remain associated with low survival or severe long-term sequelae. Limited progress, including development of immunotherapies, relates in part to difficulties in accurately reproducing brain microenvironment with current preclinical models. The cellular interactions among resident microglia, recruited tumor-associated macrophages, stromal cells, glial cells, neurons, and cancer cells and how they affect tumor growth or behavior are emerging, yet many questions remain. The role of the blood-brain barrier, extracellular matrix components, and heterogeneity among tumor types and within different regions of a single tumor further complicate the matter. Here, we focus on brain microenvironment features impacted by tumor biology. We also discuss limits of current preclinical models and how complementary models, such as humanized animals and organoids, will allow deeper mechanistic insights on cancer biology, allowing for more efficient testing of therapeutic strategies, including immunotherapy, for brain cancers.
Collapse
Affiliation(s)
- Claudia Pasqualini
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Marco Bruschi
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Thi Hai Hoa Nguyen
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Véronique Minard-Colin
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France; INSERM U1015, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - David Castel
- Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Jacques Grill
- Children and Adolescent Oncology Department, Gustave Roussy, Paris-Saclay University, Villejuif, France; Genomics & Oncogenesis of Pediatric Brain Tumors, INSERM U981, Gustave Roussy, Paris-Saclay University, Villejuif, France.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore; Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
23
|
Chhabra M, Ferro V. PI-88 and Related Heparan Sulfate Mimetics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:473-491. [PMID: 32274723 DOI: 10.1007/978-3-030-34521-1_19] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The heparan sulfate mimetic PI-88 (muparfostat) is a complex mixture of sulfated oligosaccharides that was identified in the late 1990s as a potent inhibitor of heparanase. In preclinical animal models it was shown to block angiogenesis, metastasis and tumor growth, and subsequently became the first heparanase inhibitor to enter clinical trials for cancer. It progressed to Phase III trials but ultimately was not approved for use. Herein we summarize the preparation, physicochemical and biological properties of PI-88, and discuss preclinical/clinical and structure-activity relationship studies. In addition, we discuss the PI-88-inspired development of related HS mimetic heparanase inhibitors with improved properties, ultimately leading to the discovery of PG545 (pixatimod) which is currently in clinical trials.
Collapse
Affiliation(s)
- Mohit Chhabra
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia. .,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
24
|
Pinhal MAS, Melo CM, Nader HB. The Good and Bad Sides of Heparanase-1 and Heparanase-2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:821-845. [PMID: 32274740 DOI: 10.1007/978-3-030-34521-1_36] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
In this chapter, we will emphasize the importance of heparan sulfate proteoglycans (HSPG) in controlling various physiological and pathological molecular mechanisms and discuss how the heparanase enzyme can modulate the effects triggered by HSPG. Additionally, we will also navigate about the existing knowledge of the possible role of heparanase-2 in biological events. Heparan sulfate is widely distributed and evolutionarily conserved, evidencing its vital importance in cell development and functions such as cell proliferation, migration, adhesion, differentiation, and angiogenesis. During remodeling of the extracellular matrix, the breakdown of heparan sulfate by heparanase results in the release of molecules containing anchored glycosaminoglycan chains of great interest in heparanase-mediated cell signaling pathways in various physiological states, tumor development, inflammation, and other diseases. Taken together, it appears that heparanase plays a key role in the maintenance of the pathology of cancer and inflammatory diseases and is a potential target for anti-cancer therapies. Therefore, heparanase inhibitors are currently being examined in clinical trials as novel cancer therapeutics. Heparanase-2 has no enzymatic activity, displays higher affinity for heparan sulfate and the coding region alignment shows 40% identity with the heparanase gene. Heparanase-2 plays an important role in embryogenic development however its mode of action and biological function remain to be elucidated. Heparanase-2 functions as an inhibitor of the heparanase-1 enzyme and also inhibits neovascularization mediated by VEGF. The HPSE2 gene is repressed by the Polycomb complex, together suggesting a role as a tumor suppressor.
Collapse
Affiliation(s)
| | - Carina Mucciolo Melo
- Biochemistry Department, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Helena Bonciani Nader
- Biochemistry Department, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
25
|
Heparanase-The Message Comes in Different Flavors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:253-283. [DOI: 10.1007/978-3-030-34521-1_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Fu K, Bai Z, Chen L, Ye W, Wang M, Hu J, Liu C, Zhou W. Antitumor activity and structure-activity relationship of heparanase inhibitors: Recent advances. Eur J Med Chem 2020; 193:112221. [PMID: 32222663 DOI: 10.1016/j.ejmech.2020.112221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/08/2020] [Accepted: 03/08/2020] [Indexed: 12/26/2022]
Abstract
Heparanase (HPSE)-directed tumor progression plays a crucial role in mediating tumor-host crosstalk and priming the tumor microenvironment, leading to tumor growth, metastasis and chemo-resistance. HPSE-mediated breakdown of structural heparan sulfate (HS) networks in the extracellular matrix (ECM) and basement membranes (BM) directly facilitates tumor growth and metastasis. Lysosome HPSE also induces multi-drug resistance via enhanced autophagy. Therefore, HPSE inhibitors development has become an attractive topic to block tumor growth and metastasis or eliminate drug resistance. In this review, we summarize HPSE inhibitors applied experimentally and clinically according to interaction with the binding sites of HPSE and participation of growth factors. The antitumor activity and structure-activity relationship (SAR) are also emphasized.
Collapse
Affiliation(s)
- Kaishuo Fu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China
| | - Zhifeng Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China
| | - Lanlan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China
| | - Wenchong Ye
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China
| | - Meizhu Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China
| | - Jiliang Hu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China
| | - Chunhui Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, PR China.
| | - Wen Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, China.
| |
Collapse
|
27
|
Suhovskih AV, Kazanskaya GM, Volkov AM, Tsidulko AY, Aidagulova SV, Grigorieva EV. Chemoradiotherapy Increases Intratumor Heterogeneity of HPSE Expression in the Relapsed Glioblastoma Tumors. Int J Mol Sci 2020; 21:ijms21041301. [PMID: 32075104 PMCID: PMC7073003 DOI: 10.3390/ijms21041301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 01/22/2023] Open
Abstract
Adjuvant chemoradiotherapy is a standard treatment option for glioblastoma multiforme (GBM). Despite intensive care, recurrent tumors developed during the first year are fatal for the patients. Possibly contributing to this effect, among other causes, is that therapy induces changes of polysaccharide heparan sulfate (HS) chains in the cancer cells and/or tumor microenvironment. The aim of this study was to perform a comparative analysis of heparanase (HPSE) expression and HS content in different normal and GBM brain tissues. Immunohistochemical analysis revealed a significant decrease of HPSE protein content in the tumor (12-15-fold) and paratumorous (2.5-3-fold) GBM tissues compared with normal brain tissue, both in cellular and extracellular compartments. The relapsed GBM tumors demonstrated significantly higher intertumor and/or intratumor heterogeneity of HPSE and HS content and distribution compared with the matched primary ones (from the same patient) (n = 8), although overall expression levels did not show significant differences, suggesting local deterioration of HPSE expression with reference to the control system or by the treatment. Double immunofluorescence staining of various glioblastoma cell lines (U87, U343, LN18, LN71, T406) demonstrated a complex pattern of HPSE expression and HS content with a tendency towards a negative association of these parameters. Taken together, the results demonstrate the increase of intratumor heterogeneity of HPSE protein in relapsed GBM tumors and suggest misbalance of HPSE expression regulation by the adjuvant anti-GBM chemoradiotherapy.
Collapse
Affiliation(s)
- Anastasia V. Suhovskih
- Institute of Molecular Biology and Biophysics FRC FTM, 2/12, Timakova str., 630117 Novosibirsk, Russia; (A.Y.T.); (E.V.G.)
- Novosibirsk State University, 1, Pirogova str., 630090 Novosibirsk, Russia
- Meshalkin National Medical Research Centre, 15, Rechkunovskaya str., 630055 Novosibirsk, Russia; (G.M.K.); (A.M.V.)
- Correspondence: ; Tel.: +7-383-333-5011
| | - Galina M. Kazanskaya
- Meshalkin National Medical Research Centre, 15, Rechkunovskaya str., 630055 Novosibirsk, Russia; (G.M.K.); (A.M.V.)
| | - Alexander M. Volkov
- Meshalkin National Medical Research Centre, 15, Rechkunovskaya str., 630055 Novosibirsk, Russia; (G.M.K.); (A.M.V.)
| | - Alexandra Y. Tsidulko
- Institute of Molecular Biology and Biophysics FRC FTM, 2/12, Timakova str., 630117 Novosibirsk, Russia; (A.Y.T.); (E.V.G.)
| | | | - Elvira V. Grigorieva
- Institute of Molecular Biology and Biophysics FRC FTM, 2/12, Timakova str., 630117 Novosibirsk, Russia; (A.Y.T.); (E.V.G.)
- Novosibirsk State University, 1, Pirogova str., 630090 Novosibirsk, Russia
| |
Collapse
|
28
|
Xiong A, Spyrou A, Forsberg-Nilsson K. Involvement of Heparan Sulfate and Heparanase in Neural Development and Pathogenesis of Brain Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:365-403. [PMID: 32274718 DOI: 10.1007/978-3-030-34521-1_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain tumors are aggressive and devastating diseases. The most common type of brain tumor, glioblastoma (GBM), is incurable and has one of the worst five-year survival rates of all human cancers. GBMs are invasive and infiltrate healthy brain tissue, which is one main reason they remain fatal despite resection, since cells that have already migrated away lead to rapid regrowth of the tumor. Curative therapy for medulloblastoma (MB), the most common pediatric brain tumor, has improved, but the outcome is still poor for many patients, and treatment causes long-term complications. Recent advances in the classification of pediatric brain tumors reveal distinct subgroups, allowing more targeted therapy for the most aggressive forms, and sparing children with less malignant tumors the side-effects of massive treatment. Heparan sulfate proteoglycans (HSPGs), main components of the neurogenic niche, interact specifically with a large number of physiologically important molecules and vital roles for HS biosynthesis and degradation in neural stem cell differentiation have been presented. HSPGs are composed of a core protein with attached highly charged, sulfated disaccharide chains. The major enzyme that degrades HS is heparanase (HPSE), an important regulator of extracellular matrix (ECM) remodeling which has been suggested to promote the growth and invasion of other types of tumors. This is of clinical interest because GBM are highly invasive and children with metastatic MB at the time of diagnosis exhibit a worse outcome. Here we review the involvement of HS and HPSE in development of the nervous system and some of its most malignant brain tumors, glioblastoma and medulloblastoma.
Collapse
Affiliation(s)
- Anqi Xiong
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Insitutet, Stockholm, Sweden
| | - Argyris Spyrou
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
29
|
Heparanase Inhibition by Pixatimod (PG545): Basic Aspects and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:539-565. [PMID: 32274726 DOI: 10.1007/978-3-030-34521-1_22] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pixatimod is an inhibitor of heparanase, a protein which promotes cancer via its regulation of the extracellular environment by enzymatic cleavage of heparan sulfate (HS) and non-enzymatic signaling. Through its inhibition of heparanase and other HS-binding signaling proteins, pixatimod blocks a number of pro-cancerous processes including cell proliferation, invasion, metastasis, angiogenesis and epithelial-mesenchymal transition. Several laboratories have found that these activities have translated into potent activity using a range of different mouse cancer models, including approximately 30 xenograft and 20 syngeneic models. Analyses of biological samples from these studies have confirmed the heparanase targeting of this agent in vivo and the broad spectrum of anti-cancer effects that heparanase blockade achieves. Pixatimod has been tested in combination with a number of approved anti-cancer drugs demonstrating its clinical potential, including with gemcitabine, paclitaxel, sorafenib, platinum agents and an anti-PD-1 antibody. Clinical testing has shown pixatimod to be well tolerated as a monotherapy, and it is currently being investigated in combination with the anti-PD-1 drug nivolumab in a pancreatic cancer phase I trial.
Collapse
|
30
|
Rauschenbach L. Spinal Cord Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1226:97-109. [PMID: 32030679 DOI: 10.1007/978-3-030-36214-0_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intramedullary spinal cord tumors (IMSCT) are rare entities for which there currently exist no standardized treatment paradigms. Consequently, patients usually receive treatment modalities that were established for intracerebral tumors; these approaches, however, typically result in functional impairment, recurrent tumor growth, and short overall survival. There is a distinct lack of promising research efforts in this field, which raises questions about whether spinal cord tumor microenvironment (TME) might promote the development, progression, and treatment resistance of IMSCT. In this review, we aim to examine spinal cord biology, compare spinal cord and brain microenvironments, and discuss mutual interactions between IMSCT and TME. Manipulating these pathways may provide new treatment approaches for future patient groups.
Collapse
Affiliation(s)
- Laurèl Rauschenbach
- Department of Neurosurgery, University Hospital Essen, Essen, Germany. .,DKFZ Division of Translational Neuro-Oncology at the West German Cancer Center (WTZ), German Cancer Consortium (DKTK) Partner Site, University Hospital Essen, Essen, Germany.
| |
Collapse
|
31
|
Barash U, Spyrou A, Liu P, Vlodavsky E, Zhu C, Luo J, Su D, Ilan N, Forsberg-Nilsson K, Vlodavsky I, Yang X. Heparanase promotes glioma progression via enhancing CD24 expression. Int J Cancer 2019; 145:1596-1608. [PMID: 31032901 DOI: 10.1002/ijc.32375] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 02/05/2023]
Abstract
Heparanase is an endo-β-d-glucuronidase that cleaves heparan sulfate (HS) side chains of heparan sulfate proteoglycans. Compelling evidence tie heparanase levels with all steps of tumor formation including tumor initiation, growth, metastasis and chemo-resistance, likely involving augmentation of signaling pathways and gene transcription. In order to reveal the molecular mechanism(s) underlying the protumorigenic properties of heparanase, we established an inducible (Tet-on) system in U87 human glioma cells and applied gene array methodology in order to identify genes associated with heparanase induction. We found that CD24, a mucin-like cell adhesion protein, is consistently upregulated by heparanase and by heparanase splice variant devoid of enzymatic activity, whereas heparanase gene silencing was associated with decreased CD24 expression. This finding was further substantiated by a similar pattern of heparanase and CD24 immunostaining in glioma patients (Pearson's correlation; R = 0.66, p = 0.00001). Noteworthy, overexpression of CD24 stimulated glioma cell migration, invasion, colony formation in soft agar and tumor growth in mice suggesting that CD24 functions promote tumor growth. Likewise, anti-CD24 neutralizing monoclonal antibody attenuated glioma tumor growth, and a similar inhibition was observed in mice treated with a neutralizing mAb directed against L1 cell adhesion molecule (L1CAM), a ligand for CD24. Importantly, significant shorter patient survival was found in heparanase-high/CD24-high tumors vs. heparanase-high/CD24-low tumors for both high-grade and low-grade glioma (p = 0.02). Our results thus uncover a novel heparanase-CD24-L1CAM axis that plays a significant role in glioma tumorigenesis.
Collapse
Affiliation(s)
- Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Haifa, Israel
| | - Argyris Spyrou
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Pei Liu
- Shantou University Medical College, Shantou, China
| | | | - Chenchen Zhu
- Shantou University Medical College, Shantou, China
| | - Juanjuan Luo
- Shantou University Medical College, Shantou, China
| | - Dongsheng Su
- Shantou University Medical College, Shantou, China
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Haifa, Israel
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Haifa, Israel
| | - Xiaojun Yang
- Shantou University Medical College, Shantou, China
| |
Collapse
|
32
|
Masola V, Zaza G, Gambaro G, Franchi M, Onisto M. Role of heparanase in tumor progression: Molecular aspects and therapeutic options. Semin Cancer Biol 2019; 62:86-98. [PMID: 31348993 DOI: 10.1016/j.semcancer.2019.07.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/11/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023]
Abstract
Heparanase (HPSE) is an endoglycosidase that catalyses the cutting of the side chains of heparan-sulphate proteoglycans (HS), thus determining the remodelling of the extracellular matrix and basement membranes, as well as promoting the release of different HS-related molecules as growth factors, cytokines and enzymes. Ever since the HPSE was identified in the late 1980s, several experimental studies have shown that its overexpression was instrumental in increasing tumor growth, metastatic dissemination, angiogenesis and inflammation. More recently, HPSE involvment has also been demonstrated in mediating tumor-host crosstalk, in inducing gene transcription, in the activation of signaling pathways and in the formation of exosomes and in autophagy. All of these activities (enzymatic and non-enzymatic) together make heparanase a multifunctional molecule that increases the aggressiveness and chemo-resistance of tumor cells. Conversely, heparanase gene-silencing or tumor treatment with compounds that inhibit heparanase activity have been shown to significantly attenuate tumor progression in different animal models of tumorigenesis, further emphasizing the therapeutic potential of anti-heparanase therapy for several types of neoplasms. This review focuses on present knowledge and recent development in the study of heparanase in cancer progression as well as on novel mechanisms by which heparanase regulates tumor metastasis and chemo-resistance. Moreover, recent advances in strategies for its inhibition as a potential therapeutic option will be discussed.
Collapse
Affiliation(s)
- Valentina Masola
- Dept. of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy; Dept. of Medicine, University of Verona, 37134, Verona, Italy
| | - Gianluigi Zaza
- Dept. of Medicine, University of Verona, 37134, Verona, Italy
| | | | - Marco Franchi
- Dept. of Life Quality Sciences, University of Bologna, Corso D'Augusto 237, 47921, Rimini, Italy
| | - Maurizio Onisto
- Dept. of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy.
| |
Collapse
|
33
|
Jimenez-Pascual A, Siebzehnrubl FA. Fibroblast Growth Factor Receptor Functions in Glioblastoma. Cells 2019; 8:E715. [PMID: 31337028 PMCID: PMC6678715 DOI: 10.3390/cells8070715] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma is the most lethal brain cancer in adults, with no known cure. This cancer is characterized by a pronounced genetic heterogeneity, but aberrant activation of receptor tyrosine kinase signaling is among the most frequent molecular alterations in glioblastoma. Somatic mutations of fibroblast growth factor receptors (FGFRs) are rare in these cancers, but many studies have documented that signaling through FGFRs impacts glioblastoma progression and patient survival. Small-molecule inhibitors of FGFR tyrosine kinases are currently being trialed, underlining the therapeutic potential of blocking this signaling pathway. Nevertheless, a comprehensive overview of the state of the art of the literature on FGFRs in glioblastoma is lacking. Here, we review the evidence for the biological functions of FGFRs in glioblastoma, as well as pharmacological approaches to targeting these receptors.
Collapse
MESH Headings
- Brain Neoplasms/metabolism
- Disease Progression
- Glioblastoma/metabolism
- Humans
- Receptor, Fibroblast Growth Factor, Type 1/chemistry
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- Receptor, Fibroblast Growth Factor, Type 2/chemistry
- Receptor, Fibroblast Growth Factor, Type 2/physiology
- Receptor, Fibroblast Growth Factor, Type 3/chemistry
- Receptor, Fibroblast Growth Factor, Type 3/physiology
- Receptor, Fibroblast Growth Factor, Type 4/chemistry
- Receptor, Fibroblast Growth Factor, Type 4/physiology
Collapse
Affiliation(s)
- Ana Jimenez-Pascual
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK.
| |
Collapse
|
34
|
Mayfosh AJ, Baschuk N, Hulett MD. Leukocyte Heparanase: A Double-Edged Sword in Tumor Progression. Front Oncol 2019; 9:331. [PMID: 31110966 PMCID: PMC6501466 DOI: 10.3389/fonc.2019.00331] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/11/2019] [Indexed: 12/19/2022] Open
Abstract
Heparanase is a β-D-endoglucuronidase that cleaves heparan sulfate, a complex glycosaminoglycan found ubiquitously throughout mammalian cells and tissues. Heparanase has been strongly associated with important pathological processes including inflammatory disease and tumor metastasis, through its ability to promote various cellular functions such as cell migration, invasion, adhesion, and cytokine release. A number of cell types express heparanase including leukocytes, cells of the vasculature as well as tumor cells. However, the relative contribution of heparanase from these different cell sources to these processes is poorly defined. It is now well-established that the immune system plays a critical role in shaping tumor progression. Intriguingly, leukocyte-derived heparanase has been shown to either assist or impede tumor progression, depending on the setting. This review covers our current knowledge of heparanase in immune regulation of tumor progression, as well as the potential applications and implications of exploiting or inhibiting heparanase in cancer therapy.
Collapse
Affiliation(s)
- Alyce J Mayfosh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Nikola Baschuk
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
35
|
Liu X, Zhou ZH, Li W, Zhang SK, Li J, Zhou MJ, Song JW. Heparanase Promotes Tumor Growth and Liver Metastasis of Colorectal Cancer Cells by Activating the p38/MMP1 Axis. Front Oncol 2019; 9:216. [PMID: 31001480 PMCID: PMC6454005 DOI: 10.3389/fonc.2019.00216] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/12/2019] [Indexed: 01/04/2023] Open
Abstract
Heparanase (HPSE), the only known mammalian endoglycosidase responsible for heparan sulfate cleavage, is a multi-faceted protein affecting multiple malignant behaviors in cancer cells. In this study, we examined the expression of HPSE in different colorectal cancer (CRC) cell lines. Gene manipulation was applied to reveal the effect of HPSE on proliferation, invasion, and metastasis of CRC. Knockdown of HPSE resulted in decreased cell proliferation in vitro, whereas overexpression of HPSE resulted in the opposite phenomenon. Consistently, in vivo data showed that knockdown of HPSE suppressed tumor growth of CRC. Furthermore, knockdown of HPSE inhibited invasion and liver metastasis in vitro and in vivo. RNA-sequencing analysis was performed upon knockdown of HPSE, and several pathways were identified that are closely associated with invasion and metastasis. In addition, HPSE is positively correlated with MMP1 expression in CRC, and HPSE regulates MMP1 expression via p38 MAPK signaling pathway. In conclusion, our data demonstrate that HPSE knockdown attenuated tumor growth and liver metastasis in CRC, implying that HPSE might serve as a potential therapeutic target in the treatment of CRC.
Collapse
Affiliation(s)
- Xue Liu
- Department of Pathology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Zhi-Hang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen Li
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Shi-Kun Zhang
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Jing Li
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Ming-Ju Zhou
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| | - Jin-Wen Song
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China.,Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing, China
| |
Collapse
|
36
|
Zhou L, Deng ZZ, Li HY, Jiang N, Wei ZS, Hong MF, Chen XD, Wang JH, Zhang MX, Shi YH, Lu ZQ, Huang XM. TRIM31 promotes glioma proliferation and invasion through activating NF-κB pathway. Onco Targets Ther 2019; 12:2289-2297. [PMID: 30988633 PMCID: PMC6441556 DOI: 10.2147/ott.s183625] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Glioma is the most lethal primary brain tumor, the survival rate still isn't improved in the past decades. It's essential to study the regulatory mechanism of glioma progression, hoping to find new therapy targets or methods. The family of tripartite motif (TRIM) containing proteins are E3 ubiquitination ligases, which play critical role in various tumor progression. METHODS Cell proliferation and invasion were analyzed by colony formation assay, soft agar growth assay, BrdU incorporation assay and transwell invasion assay. Luciferase reporter analysis was used to analyze NF-κB pathway activity. RESULTS We found TRIM31 was upregulated in glioma cells and tissues, its overexpression significantly promoted glioma cell proliferation and invasion, while its knockdown significantly inhibited glioma cell proliferation and invasion. Mechanism analysis found TRIM31 promoted NF-κB pathway activity and increased its targets expression. NF-κB inhibition reversed the phenotype caused by TRIM31, confirming TRIM31 promoted glioma progression through activating NF-κB pathway. Using clinical specimens found TRIM31 expression was positively correlative with NF-κB activity. CONCLUSION This study found TRIM31 promoted glioma proliferation and invasion through activating NF-κB activity.
Collapse
Affiliation(s)
- Li Zhou
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China, ,
| | - Zhe-Zhi Deng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China,
| | - Hai-Yan Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China,
| | - Nan Jiang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Zhi-Sheng Wei
- Department of Neurology, The First Affiliated Hospital of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Ming-Fan Hong
- Department of Neurology, The First Affiliated Hospital of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Xiao-Dang Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China,
| | - Ji-Hui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China,
| | - Ming-Xing Zhang
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China, ,
| | - Yi-Hua Shi
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China, ,
| | - Zheng-Qi Lu
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China, ,
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China,
| | - Xu-Ming Huang
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China, ,
| |
Collapse
|
37
|
Indira Chandran V, Welinder C, Månsson AS, Offer S, Freyhult E, Pernemalm M, Lund SM, Pedersen S, Lehtiö J, Marko-Varga G, Johansson MC, Englund E, Sundgren PC, Belting M. Ultrasensitive Immunoprofiling of Plasma Extracellular Vesicles Identifies Syndecan-1 as a Potential Tool for Minimally Invasive Diagnosis of Glioma. Clin Cancer Res 2019; 25:3115-3127. [PMID: 30679164 DOI: 10.1158/1078-0432.ccr-18-2946] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/16/2018] [Accepted: 01/16/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Liquid biopsy has great potential to improve the management of brain tumor patients at high risk of surgery-associated complications. Here, the aim was to explore plasma extracellular vesicle (plEV) immunoprofiling as a tool for noninvasive diagnosis of glioma. EXPERIMENTAL DESIGN PlEV isolation and analysis were optimized using advanced mass spectrometry, nanoparticle tracking analysis, and electron microscopy. We then established a new procedure that combines size exclusion chromatography isolation and proximity extension assay-based ultrasensitive immunoprofiling of plEV proteins that was applied on a well-defined glioma study cohort (n = 82). RESULTS Among potential candidates, we for the first time identify syndecan-1 (SDC1) as a plEV constituent that can discriminate between high-grade glioblastoma multiforme (GBM, WHO grade IV) and low-grade glioma [LGG, WHO grade II; area under the ROC curve (AUC): 0.81; sensitivity: 71%; specificity: 91%]. These findings were independently validated by ELISA. Tumor SDC1 mRNA expression similarly discriminated between GBM and LGG in an independent glioma patient population from The Cancer Genome Atlas cohort (AUC: 0.91; sensitivity: 79%; specificity: 91%). In experimental studies with GBM cells, we show that SDC1 is efficiently sorted to secreted EVs. Importantly, we found strong support of plEVSDC1 originating from GBM tumors, as plEVSDC1 correlated with SDC1 protein expression in matched patient tumors, and plEVSDC1 was decreased postoperatively depending on the extent of surgery. CONCLUSIONS Our studies support the concept of circulating plEVs as a tool for noninvasive diagnosis and monitoring of gliomas and should move this field closer to the goal of improving the management of cancer patients.
Collapse
Affiliation(s)
- Vineesh Indira Chandran
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - Charlotte Welinder
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden.,Center of Excellence in Biological and Medical Mass Spectrometry (CEBMMS), Lund University, Lund, Sweden
| | - Ann-Sofie Månsson
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - Svenja Offer
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - Eva Freyhult
- National Bioinformatics Infrastructure, SciLife Lab, Uppsala, Sweden
| | - Maria Pernemalm
- Department of Oncology and Pathology, Karolinska Institute, Solna, Sweden
| | - Sigrid M Lund
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.,Faculty of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Janne Lehtiö
- Department of Oncology and Pathology, Karolinska Institute, Solna, Sweden
| | - Gyorgy Marko-Varga
- Center of Excellence in Biological and Medical Mass Spectrometry (CEBMMS), Lund University, Lund, Sweden.,Clinical Protein Science and Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Maria C Johansson
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - Elisabet Englund
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden
| | - Pia C Sundgren
- Department of Clinical Sciences, Lund, Section of Diagnostic Radiology, Lund University, Lund, Sweden.,Lund BioImaging Centre, Lund University, Lund, Sweden.,Department of Medical Imaging and Function, Skåne University Hospital, Lund, Lund, Sweden
| | - Mattias Belting
- Department of Clinical Sciences, Lund, Section of Oncology and Pathology, Lund University, Lund, Sweden. .,Department of Hematology, Oncology and Radiophysics, Skåne University Hospital, Lund, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
38
|
Harnessing the immune system in glioblastoma. Br J Cancer 2018; 119:1171-1181. [PMID: 30393372 PMCID: PMC6251037 DOI: 10.1038/s41416-018-0258-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common primary malignant brain tumour. Survival is poor and improved treatment options are urgently needed. Although immunotherapies have emerged as effective treatments for a number of cancers, translation of these through to brain tumours is a distinct challenge, particularly due to the blood-brain barrier and the unique immune tumour microenvironment afforded by CNS-specific cells. This review discusses the immune system within the CNS, mechanisms of immune escape employed by glioblastoma, and the immunological effects of conventional glioblastoma treatments. Novel therapies for glioblastoma that harness the immune system and their current clinical progress are outlined, including cancer vaccines, T-cell therapies and immune checkpoint modulators.
Collapse
|
39
|
Hammond E, Haynes NM, Cullinane C, Brennan TV, Bampton D, Handley P, Karoli T, Lanksheer F, Lin L, Yang Y, Dredge K. Immunomodulatory activities of pixatimod: emerging nonclinical and clinical data, and its potential utility in combination with PD-1 inhibitors. J Immunother Cancer 2018; 6:54. [PMID: 29898788 PMCID: PMC6000956 DOI: 10.1186/s40425-018-0363-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/21/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Pixatimod (PG545) is a novel clinical-stage immunomodulatory agent capable of inhibiting the infiltration of tumor-associated macrophages (TAMs) yet also stimulate dendritic cells (DCs), leading to activation of natural killer (NK) cells. Preclinically, pixatimod inhibits heparanase (HPSE) which may be associated with its inhibitory effect on TAMs whereas its immunostimulatory activity on DCs is through the MyD88-dependent TLR9 pathway. Pixatimod recently completed a Phase Ia monotherapy trial in advanced cancer patients. METHODS To characterize the safety of pixatimod administered by intravenous (IV) infusion, a one month toxicology study was conducted to support a Phase Ia monotherapy clinical trial. The relative exposure (AUC) of pixatimod across relevant species was determined and the influence of route of administration on the immunomodulatory activity was also evaluated. Finally, the potential utility of pixatimod in combination with PD-1 inhibition was also investigated using the syngeneic 4T1.2 breast cancer model. RESULTS The nonclinical safety profile revealed that the main toxicities associated with pixatimod are elevated cholesterol, triglycerides, APTT, decreased platelets and other changes symptomatic of modulating the immune system such as pyrexia, changes in WBC subsets, inflammatory changes in liver, spleen and kidney. Though adverse events such as fever, elevated cholesterol and triglycerides were reported in the Phase Ia trial, none were considered dose limiting toxicities and the compound was well tolerated up to 100 mg via IV infusion. Exposure (AUC) up to 100 mg was considered proportional with some accumulation upon repeated dosing, a phenomenon also noted in the toxicology study. The immunomodulatory activity of pixatimod was independent of the route of administration and it enhanced the effectiveness of PD-1 inhibition in a poorly immunogenic tumor model. CONCLUSIONS Pixatimod modulates innate immune cells but also enhances T cell infiltration in combination with anti-PD-1 therapy. The safety and PK profile of the compound supports its ongoing development in a Phase Ib study for advanced cancer/pancreatic adenocarcinoma with the checkpoint inhibitor nivolumab (Opdivo®). TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02042781 . First posted: 23 January, 2014 - Retrospectively registered.
Collapse
Affiliation(s)
| | - Nicole M Haynes
- 0000000403978434grid.1055.1Division of Cancer ResearchPeter MacCallum Cancer Centre 3000 Melbourne VIC Australia
- 0000 0001 2179 088Xgrid.1008.9Sir Peter MacCallum Department of OncologyUniversity of Melbourne 3052 Parkville VIC Australia
| | - Carleen Cullinane
- 0000000403978434grid.1055.1Division of Cancer ResearchPeter MacCallum Cancer Centre 3000 Melbourne VIC Australia
- 0000 0001 2179 088Xgrid.1008.9Sir Peter MacCallum Department of OncologyUniversity of Melbourne 3052 Parkville VIC Australia
| | - Todd V Brennan
- 0000000100241216grid.189509.cDepartment of SurgeryDuke University Medical Center 27710 Durham North Carolina USA
| | | | | | - Tomislav Karoli
- Zucero Therapeutics 4076 Brisbane QLD Australia
- Present address: Novasep Kalkstrasse 218 51377 Leverkusen Germany
| | - Fleur Lanksheer
- Progen Pharmaceuticals 4076 Brisbane QLD Australia
- 0000 0000 8831 109Xgrid.266842.cPresent address: School of Humanities and Social ScienceThe University of Newcastle Newcastle NSW Australia
| | - Liwen Lin
- 0000000100241216grid.189509.cDepartment of SurgeryDuke University Medical Center 27710 Durham North Carolina USA
| | - Yiping Yang
- 0000000100241216grid.189509.cDepartments of Medicine and ImmunologyDuke University Medical Center 27710 Durham North Carolina USA
| | | |
Collapse
|
40
|
Patient derived xenografts (PDX) predict an effective heparanase-based therapy for lung cancer. Oncotarget 2018; 9:19294-19306. [PMID: 29721203 PMCID: PMC5922397 DOI: 10.18632/oncotarget.25022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/17/2018] [Indexed: 01/04/2023] Open
Abstract
Heparanase, the sole heparan sulfate (HS) degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, metastasis, angiogenesis, and inflammation. Heparanase accomplishes this by degrading HS and thereby facilitating cell invasion and regulating the bioavailability of heparin-binding proteins. HS mimicking compounds that inhibit heparanase enzymatic activity were examined in numerous preclinical cancer models. While these studies utilized established tumor cell lines, the current study utilized, for the first time, patient-derived xenografts (PDX) which better resemble the behavior and drug responsiveness of a given cancer patient. We have previously shown that heparanase levels are substantially elevated in lung cancer, correlating with reduced patients survival. Applying patient-derived lung cancer xenografts and a potent inhibitor of heparanase enzymatic activity (PG545) we investigated the significance of heparanase in the pathogenesis of lung cancer. PG545 was highly effective in lung cancer PDX, inhibiting tumor growth in >85% of the cases. Importantly, we show that PG545 was highly effective in PDX that did not respond to conventional chemotherapy (cisplatin) and vice versa. Moreover, we show that spontaneous metastasis to lymph nodes is markedly inhibited by PG545 but not by cisplatin. These results reflect the variability among patients and strongly imply that PG545 can be applied for lung cancer therapy in a personalized manner where conventional chemotherapy fails, thus highlighting the potential benefits of developing anti-heparanase treatment modalities for oncology.
Collapse
|
41
|
Elevated heparanase expression is associated with poor prognosis in breast cancer: a study based on systematic review and TCGA data. Oncotarget 2018; 8:43521-43535. [PMID: 28388549 PMCID: PMC5522166 DOI: 10.18632/oncotarget.16575] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 03/10/2017] [Indexed: 01/01/2023] Open
Abstract
Heparanase promotes tumorigenesis, angiogenesis, and metastasis. Here, we conducted a study based on systematic review and the Cancer Genome Atlas (TCGA) data that examined heparanase expression in clinical samples to determine its prognostic value. According to the meta-analysis and TCGA data, we found that heparanase expression was up-regulated in most breast cancer specimens, and elevated heparanase expression was associated with increased lymph node metastasis, larger tumor size, higher histological grade, and poor survival. These results suggest that targeting heparanase might improve treatments for breast cancer patients.
Collapse
|
42
|
Correction: Heparanase Promotes Glioma Progression and Is Inversely Correlated with Patient Survival. Mol Cancer Res 2018. [PMID: 29523758 DOI: 10.1158/1541-7786.mcr-18-0186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
44
|
Vlodavsky I, Gross-Cohen M, Weissmann M, Ilan N, Sanderson RD. Opposing Functions of Heparanase-1 and Heparanase-2 in Cancer Progression. Trends Biochem Sci 2017; 43:18-31. [PMID: 29162390 DOI: 10.1016/j.tibs.2017.10.007] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/24/2017] [Accepted: 10/27/2017] [Indexed: 12/24/2022]
Abstract
Heparanase, the sole heparan sulfate (HS)-degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, metastasis, angiogenesis, and inflammation. Heparanase accomplishes this by degrading HS and thereby regulating the bioavailability of heparin-binding proteins; priming the tumor microenvironment; mediating tumor-host crosstalk; and inducing gene transcription, signaling pathways, exosome formation, and autophagy that together promote tumor cell performance and chemoresistance. By contrast, heparanase-2, a close homolog of heparanase, lacks enzymatic activity, inhibits heparanase activity, and regulates selected genes that promote normal differentiation, endoplasmic reticulum stress, tumor fibrosis, and apoptosis, together resulting in tumor suppression. The emerging premise is that heparanase is a master regulator of the aggressive phenotype of cancer, while heparanase-2 functions as a tumor suppressor.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel.
| | - Miriam Gross-Cohen
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Marina Weissmann
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Ralph D Sanderson
- University of Alabama at Birmingham, Department of Pathology, Birmingham, AL 35294, USA
| |
Collapse
|
45
|
Ushakov VS, Tsidulko AY, de La Bourdonnaye G, Kazanskaya GM, Volkov AM, Kiselev RS, Kobozev VV, Kostromskaya DV, Gaytan AS, Krivoshapkin AL, Aidagulova SV, Grigorieva EV. Heparan Sulfate Biosynthetic System Is Inhibited in Human Glioma Due to EXT1/2 and HS6ST1/2 Down-Regulation. Int J Mol Sci 2017; 18:ijms18112301. [PMID: 29104277 PMCID: PMC5713271 DOI: 10.3390/ijms18112301] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 10/23/2017] [Accepted: 10/28/2017] [Indexed: 01/14/2023] Open
Abstract
Heparan sulfate (HS) is an important component of the extracellular matrix and cell surface, which plays a key role in cell–cell and cell–matrix interactions. Functional activity of HS directly depends on its structure, which determined by a complex system of HS biosynthetic enzymes. During malignant transformation, the system can undergo significant changes, but for glioma, HS biosynthesis has not been studied in detail. In this study, we performed a comparative analysis of the HS biosynthetic system in human gliomas of different grades. RT-PCR analysis showed that the overall transcriptional activity of the main HS biosynthesis-involved genes (EXT1, EXT2, NDST1, NDST2, GLCE, HS2ST1, HS3ST1, HS3ST2, HS6ST1, HS6ST2, SULF1, SULF2, HPSE) was decreased by 1.5–2-fold in Grade II-III glioma (p < 0.01) and by 3-fold in Grade IV glioma (glioblastoma multiforme, GBM) (p < 0.05), as compared with the para-tumourous tissue. The inhibition was mainly due to the elongation (a decrease in EXT1/2 expression by 3–4-fold) and 6-O-sulfation steps (a decrease in 6OST1/2 expression by 2–5-fold) of the HS biosynthesis. Heparanase (HPSE) expression was identified in 50% of GBM tumours by immunostaining, and was characterised by a high intratumoural heterogeneity of the presence of the HPSE protein. The detected disorganisation of the HS biosynthetic system in gliomas might be a potential molecular mechanism for the changes of HS structure and content in tumour microenvironments, contributing to the invasion of glioma cells and the development of the disease.
Collapse
Affiliation(s)
- Victor S Ushakov
- Institute of Molecular Biology and Biophysics, Novosibirsk 630117, Russia.
- Novosibirsk State University, Novosibirsk 630090, Russia.
| | | | - Gabin de La Bourdonnaye
- Novosibirsk State University, Novosibirsk 630090, Russia.
- National Institute of Applied Sciences, 31400 Toulouse, France.
| | - Galina M Kazanskaya
- Institute of Molecular Biology and Biophysics, Novosibirsk 630117, Russia.
- Meshalkin National Medical Research Centre, 630055 Novosibirsk, Russia.
| | | | - Roman S Kiselev
- Meshalkin National Medical Research Centre, 630055 Novosibirsk, Russia.
- Novosibirsk State Medical University, 630090 Novosibirsk, Russia.
| | | | | | | | - Alexei L Krivoshapkin
- Meshalkin National Medical Research Centre, 630055 Novosibirsk, Russia.
- Novosibirsk State Medical University, 630090 Novosibirsk, Russia.
- European Medical Centre, 129110 Moscow, Russia.
| | | | - Elvira V Grigorieva
- Institute of Molecular Biology and Biophysics, Novosibirsk 630117, Russia.
- Novosibirsk State University, Novosibirsk 630090, Russia.
| |
Collapse
|
46
|
Li G, Qin Z, Chen Z, Xie L, Wang R, Zhao H. Tumor Microenvironment in Treatment of Glioma. Open Med (Wars) 2017; 12:247-251. [PMID: 28828406 PMCID: PMC5558106 DOI: 10.1515/med-2017-0035] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 04/15/2017] [Indexed: 12/14/2022] Open
Abstract
Glioma is one of the most malignant and fatal tumors in adults. Researchers and physicians endeavor to improve clinical efficacy towards it but made little achievement. In recent years, people have made advances in understanding characteristics and functions of tumor microenvironment and its role in different processes of tumor. In this paper, we describe the effects of tumor microenvironment on glioma proliferation, invasion and treatments. By explaining underlying mechanisms and enumerating new therapy strategies employing tumor microenvironment, we aim to provide novel ideas to improve clinical outcomes of glioma.
Collapse
Affiliation(s)
- Guijie Li
- Department of Otorhinolaryngology Head and Neck Surgery, China-Japan Union hospital of Jilin University, Changchun130033, China
| | - Zhigang Qin
- Department of Neurosurgery, China-Japan Union hospital of Jilin University, Changchun130033, China
| | - Zhuo Chen
- Department of Neurosurgery, China-Japan Union hospital of Jilin University, Changchun130033, China
| | - Lijuan Xie
- Department of Vascular Surgery, China-Japan Union hospital of Jilin University, Changchun130033, China
| | - Ren Wang
- Department of Neurosurgery, The People's Hospital of Fusong County of Jilin Province, Fusong134500, China
| | - Hang Zhao
- Department of Neurosurgery, China-Japan Union hospital of Jilin University, Changchun130033, China
| |
Collapse
|
47
|
Tran VM, Wade A, McKinney A, Chen K, Lindberg OR, Engler JR, Persson AI, Phillips JJ. Heparan Sulfate Glycosaminoglycans in Glioblastoma Promote Tumor Invasion. Mol Cancer Res 2017; 15:1623-1633. [PMID: 28778876 DOI: 10.1158/1541-7786.mcr-17-0352] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 01/18/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor of adults and confers a poor prognosis due, in part, to diffuse invasion of tumor cells. Heparan sulfate (HS) glycosaminoglycans, present on the cell surface and in the extracellular matrix, regulate cell signaling pathways and cell-microenvironment interactions. In GBM, the expression of HS glycosaminoglycans and the enzymes that regulate their function are altered, but the actual HS content and structure are unknown. However, inhibition of HS glycosaminoglycan function is emerging as a promising therapeutic strategy for some cancers. In this study, we use liquid chromatography-mass spectrometry analysis to demonstrate differences in HS disaccharide content and structure across four patient-derived tumorsphere lines (GBM1, 5, 6, 43) and between two murine tumorsphere lines derived from murine GBM with enrichment of mesenchymal and proneural gene expression (mMES and mPN, respectively) markers. In GBM, the heterogeneous HS content and structure across patient-derived tumorsphere lines suggested diverse functions in the GBM tumor microenvironment. In GBM5 and mPN, elevated expression of sulfatase 2 (SULF2), an extracellular enzyme that alters ligand binding to HS, was associated with low trisulfated HS disaccharides, a substrate of SULF2. In contrast, other primary tumorsphere lines had elevated expression of the HS-modifying enzyme heparanase (HPSE). Using gene editing strategies to inhibit HPSE, a role for HPSE in promoting tumor cell adhesion and invasion was identified. These studies characterize the heterogeneity in HS glycosaminoglycan content and structure across GBM and reveal their role in tumor cell invasion.Implications: HS-interacting factors promote GBM invasion and are potential therapeutic targets. Mol Cancer Res; 15(11); 1623-33. ©2017 AACR.
Collapse
Affiliation(s)
- Vy M Tran
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Anna Wade
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Andrew McKinney
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Katharine Chen
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Olle R Lindberg
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Jane R Engler
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Anders I Persson
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.,Sandler Neurosciences Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California. .,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.,Division of Neuropathology, Department of Pathology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
48
|
Spyrou A, Kundu S, Haseeb L, Yu D, Olofsson T, Dredge K, Hammond E, Barash U, Vlodavsky I, Forsberg-Nilsson K. Inhibition of Heparanase in Pediatric Brain Tumor Cells Attenuates their Proliferation, Invasive Capacity, and In Vivo Tumor Growth. Mol Cancer Ther 2017; 16:1705-1716. [PMID: 28716813 DOI: 10.1158/1535-7163.mct-16-0900] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/30/2017] [Accepted: 05/22/2017] [Indexed: 11/16/2022]
Abstract
Curative therapy for medulloblastoma and other pediatric embryonal brain tumors has improved, but the outcome still remains poor and current treatment causes long-term complications. Malignant brain tumors infiltrate the healthy brain tissue and, thus despite resection, cells that have already migrated cause rapid tumor regrowth. Heparan sulfate proteoglycans (HSPG), major components of the extracellular matrix (ECM), modulate the activities of a variety of proteins. The major enzyme that degrades HS, heparanase (HPSE), is an important regulator of the ECM. Here, we report that the levels of HPSE in pediatric brain tumors are higher than in healthy brain tissue and that treatment of pediatric brain tumor cells with HPSE stimulated their growth. In addition, the latent, 65 kDa form of HPSE (that requires intracellular enzymatic processing for activation) enhanced cell viability and rapidly activated the ERK and AKT signaling pathways, before enzymatically active HPSE was detected. The HPSE inhibitor PG545 efficiently killed pediatric brain tumor cells, but not normal human astrocytes, and this compound also reduced tumor cell invasion in vitro and potently reduced the size of flank tumors in vivo Our findings indicate that HPSE in malignant brain tumors affects both the tumor cells themselves and their ECM. In conclusion, HPSE plays a substantial role in childhood brain tumors, by contributing to tumor aggressiveness and thereby represents a potential therapeutic target. Mol Cancer Ther; 16(8); 1705-16. ©2017 AACR.
Collapse
Affiliation(s)
- Argyris Spyrou
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Soumi Kundu
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lulu Haseeb
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Di Yu
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tommie Olofsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Keith Dredge
- Zucero Therapeutics Pty Ltd., Darra, Brisbane, Queensland, Australia
| | - Edward Hammond
- Zucero Therapeutics Pty Ltd., Darra, Brisbane, Queensland, Australia
| | - Uri Barash
- Cancer and Vascular Biology Research Center, Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
49
|
Changyaleket B, Deliu Z, Chignalia AZ, Feinstein DL. Heparanase: Potential roles in multiple sclerosis. J Neuroimmunol 2017; 310:72-81. [PMID: 28778449 DOI: 10.1016/j.jneuroim.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 06/22/2017] [Accepted: 07/01/2017] [Indexed: 12/14/2022]
Abstract
Heparanase is a heparan sulfate degrading enzyme that cleaves heparan sulfate (HS) chains present on HS proteoglycans (HSPGs), and has been well characterized for its roles in tumor metastasis and inflammation. However, heparanase is emerging as a contributing factor in the genesis and severity of a variety of neurodegenerative diseases and conditions. This is in part due to the wide variety of HSPGs on which the presence or absence of HS moieties dictates protein function. This includes growth factors, chemokines, cytokines, as well as components of the extracellular matrix (ECM) which in turn regulate leukocyte infiltration into the CNS. Roles for heparanase in stroke, Alzheimer's disease, and glioma growth have been described; roles for heparanase in other disease such as multiple sclerosis (MS) are less well established. However, given its known roles in inflammation and leukocyte infiltration, it is likely that heparanase also contributes to MS pathology. In this review, we will briefly summarize what is known about heparanase roles in the CNS, and speculate as to its potential role in regulating disease progression in MS and its animal model EAE (experimental autoimmune encephalitis), which may justify testing of heparanase inhibitors for MS treatment.
Collapse
Affiliation(s)
| | - Zane Deliu
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
| | - Andreia Z Chignalia
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA; Jesse Brown Veteran Affairs Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
50
|
Epigenetic Regulation of the Biosynthesis & Enzymatic Modification of Heparan Sulfate Proteoglycans: Implications for Tumorigenesis and Cancer Biomarkers. Int J Mol Sci 2017; 18:ijms18071361. [PMID: 28672878 PMCID: PMC5535854 DOI: 10.3390/ijms18071361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/05/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence suggests that the enzymes in the biosynthetic pathway for the synthesis of heparan sulfate moieties of heparan sulfate proteoglycans (HSPGs) are epigenetically regulated at many levels. As the exact composition of the heparan sulfate portion of the resulting HSPG molecules is critical to the broad spectrum of biological processes involved in oncogenesis, the epigenetic regulation of heparan sulfate biosynthesis has far-reaching effects on many cellular activities related to cancer progression. Given the current focus on developing new anti-cancer therapeutics focused on epigenetic targets, it is important to understand the effects that these emerging therapeutics may have on the synthesis of HSPGs as alterations in HSPG composition may have profound and unanticipated effects. As an introduction, this review will briefly summarize the variety of important roles which HSPGs play in a wide-spectrum of cancer-related cellular and physiological functions and then describe the biosynthesis of the heparan sulfate chains of HSPGs, including how alterations observed in cancer cells serve as potential biomarkers. This review will then focus on detailing the multiple levels of epigenetic regulation of the enzymes in the heparan sulfate synthesis pathway with a particular focus on regulation by miRNA and effects of epigenetic therapies on HSPGs. We will also explore the use of lectins to detect differences in heparan sulfate composition and preview their potential diagnostic and prognostic use in the clinic.
Collapse
|