1
|
Zhang H, Chen C, Han J, Wang S, Jia Q, Ling P, Li S, Fang J. Hyaluronan and Glucose Dual-targeting Probe: Synthesis and Application. Bioorg Chem 2024; 153:107816. [PMID: 39276493 DOI: 10.1016/j.bioorg.2024.107816] [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: 07/02/2024] [Revised: 08/31/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
In this work, we developed a dual-targeting probe consisted of well-defined hyaluronan (HA) oligosaccharide and glucose (Glc) labeled with Rhodamine B (HGR). The probe was designed to enhance tumor targeting both in vitro and in vivo, by simultaneously targeting CD44 and Glc transporter 1 (GLUT1). The HA oligosaccharide component was crucial for accurately assessing the impact of sugar chain structure on targeting efficacy, while its unoccupied carboxyl groups could minimize interference with HA's binding affinity to CD44. In vitro studies demonstrated that HGR possessed remarkable cytocompatibility and superior targeting abilities compared to single-targeting probes. It displayed a marked preference for CD44high/GLUT1high cells rather than CD44low/GLUT1low cells. In vivo studies using murine models further confirmed the significantly enhanced targeting efficacy and excellent biocompatibility of HGR. Therefore, this designed dual-targeting probe holds potential for clinical tumor detection.
Collapse
Affiliation(s)
- Henan Zhang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China
| | - Changsheng Chen
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China
| | - Jingjun Han
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China
| | - Shuaishuai Wang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China
| | - Qingwen Jia
- Shandong Freda Pharmaceutical Group Co. Ltd, Jinan, Shandong 250101, China
| | - Peixue Ling
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China
| | - Shuang Li
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China.
| | - Junqiang Fang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao 266237, China
| |
Collapse
|
2
|
Borsdorf S, Zeug A, Wu Y, Mitroshina E, Vedunova M, Gaitonde SA, Bouvier M, Wehr MC, Labus J, Ponimaskin E. The cell adhesion molecule CD44 acts as a modulator of 5-HT7 receptor functions. Cell Commun Signal 2024; 22:563. [PMID: 39580460 PMCID: PMC11585102 DOI: 10.1186/s12964-024-01931-0] [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: 07/18/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Homo- and heteromerization of G protein-coupled receptors (GPCRs) plays an important role in the regulation of receptor functions. Recently, we demonstrated an interaction between the serotonin receptor 7 (5-HT7R), a class A GPCR, and the cell adhesion molecule CD44. However, the functional consequences of this interaction on 5-HT7R-mediated signaling remained enigmatic. METHODS Using a quantitative FRET (Förster resonance energy transfer) approach, we determined the affinities for the formation of homo- and heteromeric complexes of 5-HT7R and CD44. The impact of heteromerization on 5-HT7R-mediated cAMP signaling was assessed using a cAMP responsive luciferase assay and a FRET-based cAMP biosensor under basal conditions as well as upon pharmacological modulation of the 5-HT7R and/or CD44 with specific ligands. We also investigated receptor-mediated G protein activation using BRET (bioluminescence resonance energy transfer)-based biosensors in both, homo- and heteromeric conditions. Finally, we analyzed expression profiles for 5-HT7R and CD44 in the brain during development. RESULTS We found that homo- and heteromerization of the 5-HT7R and CD44 occur at similar extent. Functionally, heteromerization increased 5-HT7R-mediated cAMP production under basal conditions. In contrast, agonist-mediated cAMP production was decreased in the presence of CD44. Mechanistically, this might be explained by increased Gαs and decreased GαoB activation by 5-HT7R/CD44 heteromers. Unexpectedly, treatment of the heteromeric complex with the CD44 ligand hyaluronic acid boosted constitutive 5-HT7R-mediated cAMP signaling and receptor-mediated transcription, suggesting the existence of a transactivation mechanism. CONCLUSIONS Interaction with the hyaluronan receptor CD44 modulates both the constitutive activity of 5-HT7R as well as its agonist-mediated signaling. Heteromerization also results in the transactivation of 5-HT7R-mediated signaling via CD44 ligand.
Collapse
Affiliation(s)
- Saskia Borsdorf
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Yuxin Wu
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elena Mitroshina
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria Vedunova
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Supriya A Gaitonde
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Michael C Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Systasy Bioscience GmbH, Planegg-Martinsried, Germany
| | - Josephine Labus
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
3
|
Elhashani S, Glenn M, Raymant M, Schmid MC, Mielgo A. Expression of versican isoforms V0/V1 by pancreatic cancer associated fibroblasts increases fibroblast proliferation. Pancreatology 2024; 24:719-731. [PMID: 38719756 DOI: 10.1016/j.pan.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Versican is a large extracellular matrix (ECM) proteoglycan with four isoforms V0-3. Elevated V0/V1 levels in breast cancer and glioma regulate cell migration and proliferation, but the role of versican in pancreatic ductal adenocarcinoma (PDAC) remains unclear. METHODS In this study, we evaluated the expression levels of versican isoforms, as well as their cellular source and interacting partners, in vivo, in human and mouse primary and metastatic PDAC tumours and in vitro, in pancreatic tumour cells and fibroblasts using immunostaining, confocal microscopy and qPCR techniques. We also investigated the effect of versican expression on fibroblast proliferation and migration using genetic and pharmacological approaches. RESULTS We found that versican V0/V1 is highly expressed by cancer-associated fibroblasts (CAFs) in mouse and human primary and metastatic PDAC tumours. Our data also show that exposing fibroblasts to tumour-conditioned media upregulates V0 and V1 expressions, while Verbascoside (a CD44 inhibitor) downregulates V0/V1 expression. Importantly, V0/V1 knockdown significantly inhibits fibroblast proliferation. Mechanistically, we found that inhibiting hyaluronan synthesis does not affect versican co-localisation with CD44 in fibroblasts. CONCLUSION CAFs express high levels of versican V0/V1 in primary and liver metastatic PDAC tumours and versican V0/V1 supports fibroblast proliferation.
Collapse
Affiliation(s)
- Sufyan Elhashani
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Mark Glenn
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
4
|
Xu Y, Bai Z, Lan T, Fu C, Cheng P. CD44 and its implication in neoplastic diseases. MedComm (Beijing) 2024; 5:e554. [PMID: 38783892 PMCID: PMC11112461 DOI: 10.1002/mco2.554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 05/25/2024] Open
Abstract
CD44, a nonkinase single span transmembrane glycoprotein, is a major cell surface receptor for many other extracellular matrix components as well as classic markers of cancer stem cells and immune cells. Through alternative splicing of CD44 gene, CD44 is divided into two isoforms, the standard isoform of CD44 (CD44s) and the variant isoform of CD44 (CD44v). Different isoforms of CD44 participate in regulating various signaling pathways, modulating cancer proliferation, invasion, metastasis, and drug resistance, with its aberrant expression and dysregulation contributing to tumor initiation and progression. However, CD44s and CD44v play overlapping or contradictory roles in tumor initiation and progression, which is not fully understood. Herein, we discuss the present understanding of the functional and structural roles of CD44 in the pathogenic mechanism of multiple cancers. The regulation functions of CD44 in cancers-associated signaling pathways is summarized. Moreover, we provide an overview of the anticancer therapeutic strategies that targeting CD44 and preclinical and clinical trials evaluating the pharmacokinetics, efficacy, and drug-related toxicity about CD44-targeted therapies. This review provides up-to-date information about the roles of CD44 in neoplastic diseases, which may open new perspectives in the field of cancer treatment through targeting CD44.
Collapse
Affiliation(s)
- Yiming Xu
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziyi Bai
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tianxia Lan
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Chenying Fu
- Laboratory of Aging and Geriatric Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ping Cheng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
5
|
MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int J Mol Sci 2024; 25:4102. [PMID: 38612911 PMCID: PMC11012648 DOI: 10.3390/ijms25074102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation within tumors that promote cancer progression, metastasis, and recurrence due to their self-renewal capacity and resistance to conventional therapies. CSC-specific markers and signaling pathways highly active in CSCs have emerged as a promising strategy for improving patient outcomes. This review provides a comprehensive overview of the therapeutic targets associated with CSCs of solid tumors across various cancer types, including key molecular markers aldehyde dehydrogenases, CD44, epithelial cellular adhesion molecule, and CD133 and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. We discuss a wide array of therapeutic modalities ranging from targeted antibodies, small molecule inhibitors, and near-infrared photoimmunotherapy to advanced genetic approaches like RNA interference, CRISPR/Cas9 technology, aptamers, antisense oligonucleotides, chimeric antigen receptor (CAR) T cells, CAR natural killer cells, bispecific T cell engagers, immunotoxins, drug-antibody conjugates, therapeutic peptides, and dendritic cell vaccines. This review spans developments from preclinical investigations to ongoing clinical trials, highlighting the innovative targeting strategies that have been informed by CSC-associated pathways and molecules to overcome therapeutic resistance. We aim to provide insights into the potential of these therapies to revolutionize cancer treatment, underscoring the critical need for a multi-faceted approach in the battle against cancer. This comprehensive analysis demonstrates how advances made in the CSC field have informed significant developments in novel targeted therapeutic approaches, with the ultimate goal of achieving more effective and durable responses in cancer patients.
Collapse
Affiliation(s)
- Maya R. MacLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Olivia L. Walker
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Raj Pranap Arun
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Wasundara Fernando
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Nova Scotia Health Authority, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
6
|
Zeng L, Xie L, Hu J, He C, Liu A, Lu X, Zhou W. Osteopontin-driven partial epithelial-mesenchymal transition governs the development of middle ear cholesteatoma. Cell Cycle 2024; 23:537-554. [PMID: 38662954 PMCID: PMC11135870 DOI: 10.1080/15384101.2024.2345481] [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: 09/10/2023] [Accepted: 04/17/2024] [Indexed: 05/28/2024] Open
Abstract
Cholesteatoma is a common disease of the middle ear. Currently, surgical removal is the only treatment option and patients face a high risk of relapse. The molecular basis of cholesteatoma remains largely unknown. Here, we show that Osteopontin (OPN), a predominantly secreted protein, plays a crucial role in the development of middle ear cholesteatoma. Global transcriptome analysis revealed the loss of epithelial features and an enhanced immune response in human cholesteatoma tissues. Quantitative RT-PCR and immunohistochemical staining of middle ear cholesteatoma validated the reduced expression of epithelial markers, as well as the elevated expression of mesenchymal markers including Vimentin and Fibronectin, but not N-Cadherin, α-smooth muscle actin (α-SMA) or ferroptosis suppressor protein 1 (FSP1), indicating a partial epithelial-mesenchymal transition (EMT) state. Besides, the expression of OPN was significantly elevated in human cholesteatoma tissues. Treatment with OPN promoted cell proliferation, survival and migration and led to a partial EMT in immortalized human keratinocyte cells. Importantly, blockade of OPN signaling could remarkably improve the cholesteatoma-like symptoms in SD rats. Our mechanistic study demonstrated that the AKT-zinc finger E-box binding homeobox 2 (ZEB2) axis mediated the effects of OPN. Overall, these findings suggest that targeting the OPN signaling represents a promising strategy for the treatment of middle ear cholesteatoma.
Collapse
Affiliation(s)
- Lingling Zeng
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Xie
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jin Hu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao He
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aiguo Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Lu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Zhou
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
7
|
Vogl DP, Mateos B, Migotti M, Felkl M, Conibear AC, Konrat R, Becker CFW. Semisynthesis of segmentally isotope-labeled and site-specifically palmitoylated CD44 cytoplasmic tail. Bioorg Med Chem 2024; 100:117617. [PMID: 38306881 DOI: 10.1016/j.bmc.2024.117617] [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: 11/30/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
CD44, a ubiquitously expressed transmembrane receptor, plays a crucial role in cell growth, migration, and tumor progression. Dimerization of CD44 is a key event in signal transduction and has emerged as a potential target for anti-tumor therapies. Palmitoylation, a posttranslational modification, disrupts CD44 dimerization and promotes CD44 accumulation in ordered membrane domains. However, the effects of palmitoylation on the structure and dynamics of CD44 at atomic resolution remain poorly understood. Here, we present a semisynthetic approach combining solid-phase peptide synthesis, recombinant expression, and native chemical ligation to investigate the impact of palmitoylation on the cytoplasmic domain (residues 669-742) of CD44 (CD44ct) by NMR spectroscopy. A segmentally isotope-labeled and site-specifically palmitoylated CD44 variant enabled NMR studies, which revealed chemical shift perturbations and indicated local and long-range conformational changes induced by palmitoylation. The long-range effects suggest altered intramolecular interactions and potential modulation of membrane association patterns. Semisynthetic, palmitoylated CD44ct serves as the basis for studying CD44 clustering, conformational changes, and localization within lipid rafts, and could be used to investigate its role as a tumor suppressor and to explore its therapeutic potential.
Collapse
Affiliation(s)
- Dominik P Vogl
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Str. 38, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Borja Mateos
- Max Perutz Laboratories, Vienna Biocenter Campus 5, 1030 Vienna, Austria
| | - Mario Migotti
- Max Perutz Laboratories, Vienna Biocenter Campus 5, 1030 Vienna, Austria; Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, A-1030 Vienna, Austria
| | - Manuel Felkl
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Str. 38, 1090 Vienna, Austria
| | - Anne C Conibear
- TU Wien, Institute of Applied Synthetic Chemistry, Getreidemarkt 9, 1060 Vienna, Austria
| | - Robert Konrat
- Max Perutz Laboratories, Vienna Biocenter Campus 5, 1030 Vienna, Austria
| | - Christian F W Becker
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria.
| |
Collapse
|
8
|
Shi T, Zhu J, Zhang X, Mao X. The Role of Hypoxia and Cancer Stem Cells in Development of Glioblastoma. Cancers (Basel) 2023; 15:cancers15092613. [PMID: 37174078 PMCID: PMC10177528 DOI: 10.3390/cancers15092613] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma multiform (GBM) is recognized as the most malignant brain tumor with a high level of hypoxia, containing a small population of glioblastoma stem like cells (GSCs). These GSCs have the capacity of self-renewal, proliferation, invasion and recapitulating the parent tumor, and are major causes of radio-and chemoresistance of GBM. Upregulated expression of hypoxia inducible factors (HIFs) in hypoxia fundamentally contributes to maintenance and progression of GSCs. Therefore, we thoroughly reviewed the currently acknowledged roles of hypoxia-associated GSCs in development of GBM. In detail, we recapitulated general features of GBM, especially GSC-related features, and delineated essential responses resulted from interactions between GSC and hypoxia, including hypoxia-induced signatures, genes and pathways, and hypoxia-regulated metabolic alterations. Five hypothesized GSC niches are discussed and integrated into one comprehensive concept: hypoxic peri-arteriolar niche of GSCs. Autophagy, another protective mechanism against chemotherapy, is also closely related to hypoxia and is a potential therapeutic target for GBM. In addition, potential causes of therapeutic resistance (chemo-, radio-, surgical-, immuno-), and chemotherapeutic agents which can improve the therapeutic effects of chemo-, radio-, or immunotherapy are introduced and discussed. At last, as a potential approach to reverse the hypoxic microenvironment in GBM, hyperbaric oxygen therapy (HBOT) might be an adjuvant therapy to chemo-and radiotherapy after surgery. In conclusion, we focus on demonstrating the important role of hypoxia on development of GBM, especially by affecting the function of GSCs. Important advantages have been made to understand the complicated responses induced by hypoxia in GBM. Further exploration of targeting hypoxia and GSCs can help to develop novel therapeutic strategies to improve the survival of GBM patients.
Collapse
Affiliation(s)
- Tingyu Shi
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- Tangdu Hospital, Fourth Military Medical University, Xi'an 710024, China
| | - Jun Zhu
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiang Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xinggang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
9
|
Patterson SD, Copland M. The Bone Marrow Immune Microenvironment in CML: Treatment Responses, Treatment-Free Remission, and Therapeutic Vulnerabilities. Curr Hematol Malig Rep 2023; 18:19-32. [PMID: 36780103 PMCID: PMC9995533 DOI: 10.1007/s11899-023-00688-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 02/14/2023]
Abstract
PURPOSE OF REVIEW Tyrosine kinase inhibitors (TKIs) are very successful for the treatment of chronic myeloid leukaemia (CML) but are not curative in most patients due to persistence of TKI-resistant leukaemia stem cells (LSCs). The bone marrow immune microenvironment (BME) provides protection to the LSC through multidimensional interactions, driving therapy resistance, and highlighting the need to circumvent these protective niches therapeutically. This review updates the evidence for interactions between CML cells and the immune microenvironment with a view to identifying targetable therapeutic vulnerabilities and describes what is known about the role of immune regulation in treatment-free remission (TFR). RECENT FINDINGS Intracellular signalling downstream of the chemotactic CXCL12-CXCR4 axis, responsible for disrupted homing in CML, has been elucidated in LSCs, highlighting novel therapeutic opportunities. In addition, LSCs expressing CXCL12-cleaving surface protein CD26 were highly correlated with CML burden, building on existing evidence. Newer findings implicate the adhesion molecule CD44 in TKI resistance, while JAK/STAT-mediated resistance to TKIs may occur downstream of extrinsic signalling in the BME. Exosomal BME-LSC cross-communication has also been explored. Finally, further detail on the phenotypes of natural killer (NK) cells putatively involved in maintaining successful TFR has been published, and NK-based immunotherapies are discussed. Recent studies highlight and build on our understanding of the BME in CML persistence and TKI resistance, pinpointing therapeutically vulnerable interactions. Repurposing existing drugs and/or the development of novel inhibitors targeting these relationships may help to overcome these issues in TKI-resistant CML and be used as adjuvant therapy for sustained TFR.
Collapse
Affiliation(s)
- Shaun David Patterson
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Paul O'Gorman Leukaemia Research Centre, University of Glasgow, 21 Shelley Road, Glasgow, G12 0ZD, UK.
| | - Mhairi Copland
- School of Cancer Sciences, College of Medical, Veterinary and Life Sciences, Paul O'Gorman Leukaemia Research Centre, University of Glasgow, 21 Shelley Road, Glasgow, G12 0ZD, UK.
| |
Collapse
|
10
|
Hong X, Zhang J, Zou J, Ouyang J, Xiao B, Wang P, Peng X. Role of COL6A2 in malignant progression and temozolomide resistance of glioma. Cell Signal 2023; 102:110560. [PMID: 36521657 DOI: 10.1016/j.cellsig.2022.110560] [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: 08/26/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Gliomas are one of the most common primary malignant tumors of the central nervous system, and have an unfavorable prognosis. Even combining precise surgery, chemotherapy and radiotherapy, the survival rate is still unsatisfactory. Chemotherapy resistance is one of main reasons for its adverse prognosis. As shown by several studies, glioma stem cells (GSCs) were correlated with radiotherapy/chemotherapy resistance and high relapse rate. This study aimed to find a new biomarker related to GSCs and chemotherapy resistance. METHODS TCGA, CGGA, GSE16011, GSE23806 and GDSC datasets were used to screen the genes related to GSCs, Temozolomide (TMZ) resistance, and survival. In the TCGA, GTEx, GSE16011 and CGGA datasets, mRNA level, prognostic value, and correlation with immune infiltration in the selected genes were analyzed through methods including Kaplan-Meier analysis, Cox analysis, the ESTIMATE algorithm, and the CIBERSORT algorithm. The expression of COL6A2 mRNA and protein in different groups was detected by RT-qPCR and western blot. A MTT assay and flow cytometry were used to measure the effect of COL6A2 on proliferation and apoptosis of glioma cells. RESULTS COL6A2 was positively correlated with glioma stemness and TMZ resistance. Its expression was up-regulated in GBM, and high expression was correlated with adverse prognosis. As shown by Cox analysis, COL6A2 was an independent prognostic factor for glioma. COL6A2 mRNA was increased with the glioma grade. It was over-expressed in MGMT non-methylation and IDH wild-type specimens. The results of in vitro experiments showed that COL6A2 promots proliferation of glioma cells and inhibits their apoptosis. Meanwhile, the expression of COL6A2 in TMZ-resistant glioma cells was significantly higher than that in ordinary glioma cells. As shown by GO and KEGG pathway analysis, COL6A2 was correlated with glioma proliferation, migration, invasion, and immunity. In particular, it was significantly positively correlated with PD-1, PD-L2, PD-L1, B7-H3, CTLA-4, IDO1 and TIM-3 expression, further verifying that it may play an important role in immune response. In addition, COL6A2 might influence immune cell infiltration in the glioma microenvironment. CONCLUSION COL6A2 high-expression is an indicator for adverse glioma prognosis, and is correlated with TMZ-resistant and immune response. Meanwhile, it may be a prospective biomarker for treatment.
Collapse
Affiliation(s)
- Xia Hong
- Medical School of Jingchu University of Technology, Jingmen 448000, China
| | - Jingjing Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Jianmin Zou
- The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, China
| | - Jiecai Ouyang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Boan Xiao
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Peng Wang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| | - Xiaobin Peng
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| |
Collapse
|
11
|
Matthiesen I, Jury M, Rasti Boroojeni F, Ludwig SL, Holzreuter M, Buchmann S, Åman Träger A, Selegård R, Winkler TE, Aili D, Herland A. Astrocyte 3D culture and bioprinting using peptide functionalized hyaluronan hydrogels. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2023; 24:2165871. [PMID: 36733710 PMCID: PMC9888471 DOI: 10.1080/14686996.2023.2165871] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 06/18/2023]
Abstract
Astrocytes play an important role in the central nervous system, contributing to the development of and maintenance of synapses, recycling of neurotransmitters, and the integrity and function of the blood-brain barrier. Astrocytes are also linked to the pathophysiology of various neurodegenerative diseases. Astrocyte function and organization are tightly regulated by interactions mediated by the extracellular matrix (ECM). Engineered hydrogels can mimic key aspects of the ECM and can allow for systematic studies of ECM-related factors that govern astrocyte behaviour. In this study, we explore the interactions between neuroblastoma (SH-SY5Y) and glioblastoma (U87) cell lines and human fetal primary astrocytes (FPA) with a modular hyaluronan-based hydrogel system. Morphological analysis reveals that FPA have a higher degree of interactions with the hyaluronan-based gels compared to the cell lines. This interaction is enhanced by conjugation of cell-adhesion peptides (cRGD and IKVAV) to the hyaluronan backbone. These effects are retained and pronounced in 3D bioprinted structures. Bioprinted FPA using cRGD functionalized hyaluronan show extensive and defined protrusions and multiple connections between neighboring cells. Possibilities to tailor and optimize astrocyte-compatible ECM-mimicking hydrogels that can be processed by means of additive biofabrication can facilitate the development of advanced tissue and disease models of the central nervous system.
Collapse
Affiliation(s)
- Isabelle Matthiesen
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Jury
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Fatemeh Rasti Boroojeni
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Saskia L. Ludwig
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Muriel Holzreuter
- AIMES, Center for Integrated Medical and Engineering Science, Department of Neuroscience, Karolinska Institute, Solna, Sweden
| | - Sebastian Buchmann
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- AIMES, Center for Integrated Medical and Engineering Science, Department of Neuroscience, Karolinska Institute, Solna, Sweden
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Andrea Åman Träger
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Thomas E. Winkler
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- Institute of Microtechnology & Center of Pharmaceutical Engineering, Technische Universität Braunschweig, Braunschweig, Germany
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Anna Herland
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- AIMES, Center for Integrated Medical and Engineering Science, Department of Neuroscience, Karolinska Institute, Solna, Sweden
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
12
|
Zhang J, Hu Z, Horta CA, Yang J. Regulation of epithelial-mesenchymal transition by tumor microenvironmental signals and its implication in cancer therapeutics. Semin Cancer Biol 2023; 88:46-66. [PMID: 36521737 DOI: 10.1016/j.semcancer.2022.12.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated in various aspects of tumor development, including tumor invasion and metastasis, cancer stemness, and therapy resistance. Diverse stroma cell types along with biochemical and biophysical factors in the tumor microenvironment impinge on the EMT program to impact tumor progression. Here we provide an in-depth review of various tumor microenvironmental signals that regulate EMT in cancer. We discuss the molecular mechanisms underlying the role of EMT in therapy resistance and highlight new therapeutic approaches targeting the tumor microenvironment to impact EMT and tumor progression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Calista A Horta
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Novel Antibody Exerts Antitumor Effect through Downregulation of CD147 and Activation of Multiple Stress Signals. JOURNAL OF ONCOLOGY 2022; 2022:3552793. [PMID: 36385956 PMCID: PMC9652086 DOI: 10.1155/2022/3552793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022]
Abstract
CD147 is an immunoglobulin-like receptor that is highly expressed in various cancers and involved in the growth, metastasis, and activation of inflammatory pathways via interactions with various functional molecules, such as integrins, CD44, and monocarboxylate transporters. Through screening of CD147-targeting antibodies with antitumor efficacy, we discovered a novel rat monoclonal antibody #147D. This humanized IgG4-formatted antibody, h4#147D, showed potent antitumor efficacy in xenograft mouse models harboring the human PDAC cell line MIA PaCa-2, HCC cell line Hep G2, and CML cell line KU812, which featured low sensitivity to the corresponding standard-of-care drugs (gemcitabine, sorafenib, and imatinib, respectively). An analysis of tumor cells derived from MIA PaCa-2 xenograft mice treated with h4#147D revealed that cell surface expression of CD147 and its binding partners, including CD44 and integrin α3β1/α6β1, was significantly reduced by h4#147D. Inhibition of focal adhesion kinase (FAK), activation of multiple stress responsible signal proteins such as c-JunN-terminal kinase (JNK) and mitogen-activated protein kinase p38 (p38MAPK), and expression of SMAD4, as well as activation of caspase-3 were obviously observed in the tumor cells, suggesting that h4#147D induced tumor shrinkage by inducing multiple stress responsible signals. These results suggest that the anti-CD147 antibody h4#147D offers promise as a new antibody drug candidate.
Collapse
|
14
|
Faisal SM, Comba A, Varela ML, Argento AE, Brumley E, Abel C, Castro MG, Lowenstein PR. The complex interactions between the cellular and non-cellular components of the brain tumor microenvironmental landscape and their therapeutic implications. Front Oncol 2022; 12:1005069. [PMID: 36276147 PMCID: PMC9583158 DOI: 10.3389/fonc.2022.1005069] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Glioblastoma (GBM), an aggressive high-grade glial tumor, is resistant to therapy and has a poor prognosis due to its universal recurrence rate. GBM cells interact with the non-cellular components in the tumor microenvironment (TME), facilitating their rapid growth, evolution, and invasion into the normal brain. Herein we discuss the complexity of the interactions between the cellular and non-cellular components of the TME and advances in the field as a whole. While the stroma of non-central nervous system (CNS) tissues is abundant in fibrillary collagens, laminins, and fibronectin, the normal brain extracellular matrix (ECM) predominantly includes proteoglycans, glycoproteins, and glycosaminoglycans, with fibrillary components typically found only in association with the vasculature. However, recent studies have found that in GBMs, the microenvironment evolves into a more complex array of components, with upregulated collagen gene expression and aligned fibrillary ECM networks. The interactions of glioma cells with the ECM and the degradation of matrix barriers are crucial for both single-cell and collective invasion into neighboring brain tissue. ECM-regulated mechanisms also contribute to immune exclusion, resulting in a major challenge to immunotherapy delivery and efficacy. Glioma cells chemically and physically control the function of their environment, co-opting complex signaling networks for their own benefit, resulting in radio- and chemo-resistance, tumor recurrence, and cancer progression. Targeting these interactions is an attractive strategy for overcoming therapy resistance, and we will discuss recent advances in preclinical studies, current clinical trials, and potential future clinical applications. In this review, we also provide a comprehensive discussion of the complexities of the interconnected cellular and non-cellular components of the microenvironmental landscape of brain tumors to guide the development of safe and effective therapeutic strategies against brain cancer.
Collapse
Affiliation(s)
- Syed M. Faisal
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria L. Varela
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anna E. Argento
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Emily Brumley
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Clifford Abel
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R. Lowenstein
- Dept. of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Pedro R. Lowenstein,
| |
Collapse
|
15
|
Ahrens S, Appl B, Trochimiuk M, Dücker C, Feixas Serra G, Oliver Grau A, Reinshagen K, Pagerols Raluy L. Kigelia africana inhibits proliferation and induces cell death in stage 4 Neuroblastoma cell lines. Biomed Pharmacother 2022; 154:113584. [PMID: 36029541 DOI: 10.1016/j.biopha.2022.113584] [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: 06/09/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022] Open
Abstract
Neuroblastoma (NB) is one of the most common solid pediatric tumors and especially high-risk NBs still account for about 12-15% of cancer related deaths in children. Kigelia africana (KA) is a plant used in traditional African medicine which has already shown its anti-cancer potential in several in vitro and in vivo studies. The aim of this study is to evaluate the effect of KA fruit extract on stage 4 high-risk NB cells. Therefore, NB cell lines with and without MYCN amplification and non-neoplastic cells were treated with KA fruit extract at different concentrations. The effect of KA on cell viability and apoptosis rate were assessed by bioluminescence-/fluorescence-based assays. Several proteins involved in survival, tumor growth, inflammation and metastasis were detected via western blot and immunofluorescence. Secreted cytokines were detected via ELISA. Phytochemical composition of the extract was analyzed by liquid chromatography with tandem mass spectrometry (LC/MS/MS). Our group demonstrates a dose- and time-dependent selective cytotoxic effect of KA fruit extract on NB, especially in MYCN non-amplified tumor cells, by inhibiting cell proliferation and inducing cell death. Western blot and immunofluorescence results demonstrate a regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), disialoganglioside GD2 and epidermal growth factor receptor (EGFR) in KA-treated tumor cells. Our results evidence striking anti-cancer properties of KA fruit and pave the way for further surveys on the therapeutic properties and mechanisms of action in NB.
Collapse
Affiliation(s)
- Sofia Ahrens
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Birgit Appl
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Magdalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Charlotte Dücker
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Laia Pagerols Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
16
|
Handa T, Sasaki H, Takao M, Tano M, Uchida Y. Proteomics-based investigation of cerebrovascular molecular mechanisms in cerebral amyloid angiopathy by the FFPE-LMD-PCT-SWATH method. Fluids Barriers CNS 2022; 19:56. [PMID: 35778717 PMCID: PMC9250250 DOI: 10.1186/s12987-022-00351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) occurs in 80% of patients with Alzheimer’s disease (AD) and is mainly caused by the abnormal deposition of Aβ in the walls of cerebral blood vessels. Cerebrovascular molecular mechanisms in CAA were investigated by using comprehensive and accurate quantitative proteomics. Methods Concerning the molecular mechanisms specific to CAA, formalin-fixed paraffin-embedded (FFPE) sections were prepared from patients having AD neuropathologic change (ADNC) with severe cortical Aβ vascular deposition (ADNC +/CAA +), and from patients having ADNC without vascular deposition of Aβ (ADNC +/CAA −; so called, AD). Cerebral cortical vessels were isolated from FFPE sections using laser microdissection (LMD), processed by pressure cycling technology (PCT), and applied to SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Results The protein expression levels of 17 proteins in ADNC +/CAA +/H donors (ADNC +/CAA + donors with highly abundant Aβ in capillaries) were significantly different from those in ADNC +/CAA − and ADNC −/CAA − donors. Furthermore, we identified 56 proteins showing more than a 1.5-fold difference in average expression levels between ADNC +/CAA + and ADNC −/CAA − donors, and were significantly correlated with the levels of Aβ or Collagen alpha-2(VI) chain (COL6A2) (CAA markers) in 11 donors (6 ADNC +/CAA + and 5 ADNC −/CAA −). Over 70% of the 56 proteins showed ADNC +/CAA + specific changes in protein expression. The comparative analysis with brain parenchyma showed that more than 90% of the 56 proteins were vascular-specific pathological changes. A literature-based pathway analysis showed that 42 proteins are associated with fibrosis, oxidative stress and apoptosis. This included the increased expression of Heat shock protein HSP 90-alpha, CD44 antigen and Carbonic anhydrase 1 which are inhibited by potential drugs against CAA. Conclusions The combination of LMD-based isolation of vessels from FFPE sections, PCT-assisted sample processing and SWATH analysis (FFPE-LMD-PCT-SWATH method) revealed for the first time the changes in the expression of many proteins that are involved in fibrosis, ROS production and cell death in ADNC +/CAA + (CAA patients) vessels. The findings reported herein would be useful for developing a better understanding of the pathology of CAA and for promoting the discovery and development of drugs and biomarkers for CAA. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00351-x.
Collapse
Affiliation(s)
- Takumi Handa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hayate Sasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan.,Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan. .,Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
17
|
Ma Z, Shi S, Ren M, Pang C, Zhan Y, An H, Sun F. Molecular Mechanism of CD44 Homodimerization Modulated by Palmitoylation and Membrane Environments. Biophys J 2022; 121:2671-2683. [PMID: 35733341 DOI: 10.1016/j.bpj.2022.06.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/21/2022] [Accepted: 06/16/2022] [Indexed: 11/15/2022] Open
Abstract
The homodimerization of CD44 plays a key role in an intercellular-to-extracellular signal transduction and tumor progression. Acylated modification and specific membrane environments have been reported to mediate translocation and oligomerization of CD44, however, the underlying molecular mechanism remains elusive. In this study, extensive molecular dynamics simulations are performed to characterize the dimerization of palmitoylated CD44 variants in different bilayer environments. CD44 forms homodimer depending on the cysteines on the juxta-membrane domains, and the dimerization efficiency and packing configurations are defected by their palmitoylated modifications. In the phase-segregated (raft included) membrane, homodimerization of the palmitoylated CD44 is hardly observed, whereas PIP2 addition compensates to realize dimerization. However, the structure of CD44 homodimer formed in the phase-segregated bilayer turns susceptive and PIP2 addition allows for an extensive conformation of the cytoplasmic domain, a proposal prerequisite to access the cytoskeleton linker proteins. The results unravel a delicate competitive relationship between PIP2, lipid raft and palmitoylation in mediating protein homodimerization, which helps to clarify the dynamic dimer conformations and involved cellular signaling of the CD44 likewise proteins.
Collapse
Affiliation(s)
- Ziyi Ma
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Sai Shi
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China; State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300401, China; Key Laboratory of Electromagnetic Field and Electrical Apparatus Reliability of Hebei Province, Hebei University of Technology, Tianjin 300401, China
| | - Meina Ren
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Chunli Pang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Yong Zhan
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China; State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300401, China; Key Laboratory of Electromagnetic Field and Electrical Apparatus Reliability of Hebei Province, Hebei University of Technology, Tianjin 300401, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China; State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300401, China; Key Laboratory of Electromagnetic Field and Electrical Apparatus Reliability of Hebei Province, Hebei University of Technology, Tianjin 300401, China.
| | - Fude Sun
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China.
| |
Collapse
|
18
|
Hsiao Y, Chi J, Li C, Chen L, Chen Y, Liang H, Lo Y, Hong J, Chuu C, Hung L, Du J, Chang W, Wang J. Disruption of the pentraxin 3/CD44 interaction as an efficient therapy for triple-negative breast cancers. Clin Transl Med 2022; 12:e724. [PMID: 35090088 PMCID: PMC8797470 DOI: 10.1002/ctm2.724] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/29/2022] Open
Abstract
Due to the heterogeneity and high frequency of genome mutations in cancer cells, targeting vital protumour factors found in stromal cells in the tumour microenvironment may represent an ideal strategy in cancer therapy. However, the regulation and mechanisms of potential targetable therapeutic candidates need to be investigated. An in vivo study demonstrated that loss of pentraxin 3 (PTX3) in stromal cells significantly decreased the metastasis and growth of cancer cells. Clinically, our results indicate that stromal PTX3 expression correlates with adverse prognostic features and is associated with worse survival outcomes in triple-negative breast cancer (TNBC). We also found that transforming growth factor beta 1 (TGF-β1) induces PTX3 expression by activating the transcription factor CCAAT/enhancer binding protein delta (CEBPD) in stromal fibroblasts. Following PTX3 stimulation, CD44, a PTX3 receptor, activates the downstream ERK1/2, AKT and NF-κB pathways to specifically contribute to the metastasis/invasion and stemness of TNBC MDA-MB-231 cells. Two types of PTX3 inhibitors were developed to disrupt the PTX3/CD44 interaction and they showed a significant effect on attenuating growth and restricting the metastasis/invasion of MDA-MB-231 cells, suggesting that targeting the PTX3/CD44 interaction could be a new strategy for future TNBC therapies.
Collapse
Affiliation(s)
- Yu‐Wei Hsiao
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jhih‐Ying Chi
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Chien‐Feng Li
- Department of PathologyChi‐Mei Medical CenterTainanTaiwan R. O. C.
| | - Lei‐Yi Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Yi‐Ting Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Hsin‐Yin Liang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Yu‐Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jhen‐Yi Hong
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Chin‐Pin Chuu
- Institute of Cellular and System MedicineNational Health Research InstitutesMiaoli CountyTaiwan R. O. C.
| | - Liang‐Yi Hung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Jyun‐Yi Du
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
| | - Wen‐Chang Chang
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan R. O. C.
| | - Ju‐Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and BiotechnologyNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan R. O. C.
- International Research Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Department of Physiology, College of MedicineNational Cheng Kung UniversityTainanTaiwan R. O. C.
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan R. O. C.
| |
Collapse
|
19
|
Wei J, Gilboa E, Calin GA, Heimberger AB. Immune Modulatory Short Noncoding RNAs Targeting the Glioblastoma Microenvironment. Front Oncol 2021; 11:682129. [PMID: 34532286 PMCID: PMC8438301 DOI: 10.3389/fonc.2021.682129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas are heterogeneous and have a poor prognosis. Glioblastoma cells interact with their neighbors to form a tumor-permissive and immunosuppressive microenvironment. Short noncoding RNAs are relevant mediators of the dynamic crosstalk among cancer, stromal, and immune cells in establishing the glioblastoma microenvironment. In addition to the ease of combinatorial strategies that are capable of multimodal modulation for both reversing immune suppression and enhancing antitumor immunity, their small size provides an opportunity to overcome the limitations of blood-brain-barrier (BBB) permeability. To enhance glioblastoma delivery, these RNAs have been conjugated with various molecules or packed within delivery vehicles for enhanced tissue-specific delivery and increased payload. Here, we focus on the role of RNA therapeutics by appraising which types of nucleotides are most effective in immune modulation, lead therapeutic candidates, and clarify how to optimize delivery of the therapeutic RNAs and their conjugates specifically to the glioblastoma microenvironment.
Collapse
Affiliation(s)
- Jun Wei
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eli Gilboa
- Department of Microbiology & Immunology, Dodson Interdisciplinary Immunotherapy Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| | - George A Calin
- Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amy B Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
20
|
Vasse GF, Nizamoglu M, Heijink IH, Schlepütz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol 2021; 254:344-357. [PMID: 33506963 PMCID: PMC8252758 DOI: 10.1002/path.5632] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Fibrosis results from aberrant wound healing and is characterized by an accumulation of extracellular matrix, impairing the function of an affected organ. Increased deposition of extracellular matrix proteins, disruption of matrix degradation, but also abnormal post-translational modifications alter the biochemical composition and biophysical properties of the tissue microenvironment - the stroma. Macrophages are known to play an important role in wound healing and tissue repair, but the direct influence of fibrotic stroma on macrophage behaviour is still an under-investigated element in the pathogenesis of fibrosis. In this review, the current knowledge on interactions between macrophages and (fibrotic) stroma will be discussed from biochemical, biophysical, and cellular perspectives. Furthermore, we provide future perspectives with regard to how macrophage-stroma interactions can be examined further to ultimately facilitate more specific targeting of these interactions in the treatment of fibrosis. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gwenda F Vasse
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of PulmonologyGroningenThe Netherlands
| | - Marco Schlepütz
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Patrick van Rijn
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Barbro N Melgert
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
21
|
Wuelling M, Neu C, Thiesen AM, Kitanovski S, Cao Y, Lange A, Westendorf AM, Hoffmann D, Vortkamp A. Epigenetic Mechanisms Mediating Cell State Transitions in Chondrocytes. J Bone Miner Res 2021; 36:968-985. [PMID: 33534175 DOI: 10.1002/jbmr.4263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 01/06/2023]
Abstract
Epigenetic modifications play critical roles in regulating cell lineage differentiation, but the epigenetic mechanisms guiding specific differentiation steps within a cell lineage have rarely been investigated. To decipher such mechanisms, we used the defined transition from proliferating (PC) into hypertrophic chondrocytes (HC) during endochondral ossification as a model. We established a map of activating and repressive histone modifications for each cell type. ChromHMM state transition analysis and Pareto-based integration of differential levels of mRNA and epigenetic marks revealed that differentiation-associated gene repression is initiated by the addition of H3K27me3 to promoters still carrying substantial levels of activating marks. Moreover, the integrative analysis identified genes specifically expressed in cells undergoing the transition into hypertrophy. Investigation of enhancer profiles detected surprising differences in enhancer number, location, and transcription factor binding sites between the two closely related cell types. Furthermore, cell type-specific upregulation of gene expression was associated with increased numbers of H3K27ac peaks. Pathway analysis identified PC-specific enhancers associated with chondrogenic genes, whereas HC-specific enhancers mainly control metabolic pathways linking epigenetic signature to biological functions. Since HC-specific enhancers show a higher conservation in postnatal tissues, the switch to metabolic pathways seems to be a hallmark of differentiated tissues. Surprisingly, the analysis of H3K27ac levels at super-enhancers revealed a rapid adaption of H3K27ac occupancy to changes in gene expression, supporting the importance of enhancer modulation for acute alterations in gene expression. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Manuela Wuelling
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Christoph Neu
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Andrea M Thiesen
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Simo Kitanovski
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Yingying Cao
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Anja Lange
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Andrea Vortkamp
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
22
|
Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, Qiao Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther 2021; 6:153. [PMID: 33888679 PMCID: PMC8062524 DOI: 10.1038/s41392-021-00544-0] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is one of the major components of tumors that plays multiple crucial roles, including mechanical support, modulation of the microenvironment, and a source of signaling molecules. The quantity and cross-linking status of ECM components are major factors determining tissue stiffness. During tumorigenesis, the interplay between cancer cells and the tumor microenvironment (TME) often results in the stiffness of the ECM, leading to aberrant mechanotransduction and further malignant transformation. Therefore, a comprehensive understanding of ECM dysregulation in the TME would contribute to the discovery of promising therapeutic targets for cancer treatment. Herein, we summarized the knowledge concerning the following: (1) major ECM constituents and their functions in both normal and malignant conditions; (2) the interplay between cancer cells and the ECM in the TME; (3) key receptors for mechanotransduction and their alteration during carcinogenesis; and (4) the current therapeutic strategies targeting aberrant ECM for cancer treatment.
Collapse
Affiliation(s)
- Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Lele Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Dalong Wan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shengzhang Lin
- School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China.
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
23
|
Wang H, Feng J, Ao F, Tang Y, Xu P, Wang M, Huang M. Tumor-derived exosomal microRNA-7-5p enhanced by verbascoside inhibits biological behaviors of glioblastoma in vitro and in vivo. MOLECULAR THERAPY-ONCOLYTICS 2020; 20:569-582. [PMID: 33768139 PMCID: PMC7972934 DOI: 10.1016/j.omto.2020.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022]
Abstract
Verbascoside (VB), a glycosylated phenylpropane compound, has been widely used in traditional medicine showing anti-inflammatory and anti-tumor effects in many diseases. The current study aimed to investigate the mechanism underlying the inhibitor effect of VB on glioblastoma (GBM). We isolated and identified the tumor-derived exosomes (TEXs) secreted by GBM cells before and after treatment with VB, after which, we detected expression of microRNA (miR)-7-5p in cells and TEXs by qRT-PCR. Loss- and gain-function assays were conducted to determine the role of miR-7-5p in GBM cells with the proliferation, apoptosis, invasion, migration, and microtubule formation of GBM cells detected. A subcutaneous tumor model and tumor metastasis model of nude mice were established to validate the in vitro findings. We found that VB promoted the expression of miR-7-5p in GBM and transferred miR-7-5p to recipient GBM cells by exosomal delivery. Consequently, miR-7-5p downregulated epidermal growth factor receptor (EGFR) expression to inactivate the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway, causing inhibition in the proliferation, migration, invasion, and microtubule formation of GBM cells in vitro, as well as decline in tumor formation and metastasis in vivo. Overall, VB can promote the expression of miR-7-5p in GBM cells and transfer miR-7-5p via exosomes, thereby inhibiting the occurrence of GBM.
Collapse
Affiliation(s)
- Huan Wang
- Department of Neurosurgery, Jiangxi Provincial Corps Hospital of Chinese People’s Armed Police Forces, Nanchang 330001, PR China
| | - Jiugeng Feng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Fan Ao
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Yiqiang Tang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Pengliang Xu
- Department of Neurosurgery, Jiangxi Provincial Corps Hospital of Chinese People’s Armed Police Forces, Nanchang 330001, PR China
| | - Min Wang
- Department of Neurosurgery, Jiangxi Provincial Corps Hospital of Chinese People’s Armed Police Forces, Nanchang 330001, PR China
| | - Min Huang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
- Corresponding author: Min Huang, Department of Radiation Oncology, Jiangxi Cancer Hospital, No. 519, Eastern Beijing Road, Nanchang 330029, Jiangxi Province, PR China.
| |
Collapse
|
24
|
Wang C, Wang Z, Chen C, Fu X, Wang J, Fei X, Yan X, Xu R. A low MW inhibitor of CD44 dimerization for the treatment of glioblastoma. Br J Pharmacol 2020; 177:3009-3023. [PMID: 32080830 PMCID: PMC7280016 DOI: 10.1111/bph.15030] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE As a hallmark of glioblastoma multiforme (GBM), CD44 plays a crucial role in promoting glioblastoma stem cell (GSC) stemness phenotypes and multiple drug resistance. The therapeutic potential of CD44 has been validated by the clinical successes of several CD44 inhibitors, including antibodies and hyaluronan-related drugs. EXPERIMENTAL APPROACH We used systemsDock software to predict verbascoside as a candidate CD44 inhibitor. Microscale thermophoresis was used to confirm the interaction between CD44 and verbascoside. Four glioblastoma cell lines and a patient-derived glioblastoma cell line were used to test the influences of verbascoside on glioblastoma. CD44-overexpressing and CD44-knockout cell lines were also used. Real-time quantitative PCR and western blot analyses were performed. A xenograft mouse model was used to test verbascoside. KEY RESULTS Verbascoside bound to CD44 and suppressed its dimerization. By inhibiting CD44 dimerization, verbascoside decreased the release of the CD44 intracellular domain (CD44ICD) and suppressed the expression of CD44 downstream genes. Verbascoside treatment suppressed the stemness phenotypes of cells with high CD44 expression. In a mouse model of glioma, verbascoside treatment highly reduced the growth of intracranial tumours and inhibited CD44ICD release. Both stem cell marker and mesenchymal GBM subtype marker genes were down-regulated in verbascoside-treated mice. CONCLUSION AND IMPLICATIONS Verbascoside suppressed growth of glioblastoma cells by inhibiting CD44 dimerization. Stem cell-like cell properties and tumour cell growth were also suppressed by verbascoside, both in vitro and in vivo. Verbascoside significantly prolonged survival of xenografted mice.
Collapse
Affiliation(s)
- Chongwu Wang
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
| | - Zhaotao Wang
- Department of NeurosurgeryThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Chen Chen
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
| | - Xiaojun Fu
- Chinese PLA General HospitalMedicine School of Chinese PLABeijingChina
| | - Ji Wang
- School of basic medical scienceSouthern Medicine UniversityGuangzhouChina
| | - Xiaowei Fei
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
| | - Xiaojing Yan
- State Key Laboratory of Tree Genetics and BreedingChinese Academy of ForestryBeijingChina
| | - Ruxiang Xu
- The 7th Medical center of Chinese PLA general hospitalChinese PLA General Hospital Afflicted the Seventh Medical CenterBeijingChina
- School of basic medical scienceSouthern Medicine UniversityGuangzhouChina
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|