1
|
Brooks A, Zhang Y, Chen J, Zhao CX. Cancer Metastasis-on-a-Chip for Modeling Metastatic Cascade and Drug Screening. Adv Healthc Mater 2024; 13:e2302436. [PMID: 38224141 DOI: 10.1002/adhm.202302436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Microfluidic chips are valuable tools for studying intricate cellular and cell-microenvironment interactions. Traditional in vitro cancer models lack accuracy in mimicking the complexities of in vivo tumor microenvironment. However, cancer-metastasis-on-a-chip (CMoC) models combine the advantages of 3D cultures and microfluidic technology, serving as powerful platforms for exploring cancer mechanisms and facilitating drug screening. These chips are able to compartmentalize the metastatic cascade, deepening the understanding of its underlying mechanisms. This article provides an overview of current CMoC models, focusing on distinctive models that simulate invasion, intravasation, circulation, extravasation, and colonization, and their applications in drug screening. Furthermore, challenges faced by CMoC and microfluidic technologies are discussed, while exploring promising future directions in cancer research. The ongoing development and integration of these models into cancer studies are expected to drive transformative advancements in the field.
Collapse
Affiliation(s)
- Anastasia Brooks
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Jiezhong Chen
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
2
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [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: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Zeissig MN, Hewett DR, Mrozik KM, Panagopoulos V, Wallington-Gates CT, Spencer A, Dold SM, Engelhardt M, Vandyke K, Zannettino ACW. Expression of the chemokine receptor CCR1 decreases sensitivity to bortezomib in multiple myeloma cell lines. Leuk Res 2024; 139:107469. [PMID: 38479337 DOI: 10.1016/j.leukres.2024.107469] [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: 08/03/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND The proteasome inhibitor bortezomib is one of the primary therapies used for the haematological malignancy multiple myeloma (MM). However, intrinsic or acquired resistance to bortezomib, via mechanisms that are not fully elucidated, is a barrier to successful treatment in many patients. Our previous studies have shown that elevated expression of the chemokine receptor CCR1 in MM plasma cells in newly diagnosed MM patients is associated with poor prognosis. Here, we hypothesised that the poor prognosis conferred by CCR1 expression is, in part, due to a CCR1-mediated decrease in MM plasma cell sensitivity to bortezomib. METHODS In order to investigate the role of CCR1 in MM cells, CCR1 was knocked out in human myeloma cell lines OPM2 and U266 using CRISPR-Cas9. Additionally, CCR1 was overexpressed in the mouse MM cell line 5TGM1. The effect of bortezomib on CCR1 knockout or CCR1-overexpressing cells was then assessed by WST-1 assay, with or without CCL3 siRNA knockdown or addition of recombinant human CCL3. NSG mice were inoculated intratibially with OPM2-CCR1KO cells and were treated with 0.7 mg/kg bortezomib or vehicle twice per week for 3 weeks and GFP+ tumour cells in the bone marrow were quantitated by flow cytometry. The effect of CCR1 overexpression or knockout on unfolded protein response pathways was assessed using qPCR for ATF4, HSPA5, XBP1, ERN1 and CHOP and Western blot for IRE1α and p-Jnk. RESULTS Using CCR1 overexpression or CRIPSR-Cas9-mediated CCR1 knockout in MM cell lines, we found that CCR1 expression significantly decreases sensitivity to bortezomib in vitro, independent of the CCR1 ligand CCL3. In addition, CCR1 knockout rendered the human MM cell line OPM2 more sensitive to bortezomib in an intratibial MM model in NSG mice in vivo. Moreover, CCR1 expression negatively regulated the expression of the unfolded protein response receptor IRE1 and downstream target gene XBP1, suggesting this pathway may be responsible for the decreased bortezomib sensitivity of CCR1-expressing cells. CONCLUSIONS Taken together, these studies suggest that CCR1 expression may be associated with decreased response to bortezomib in MM cell lines.
Collapse
Affiliation(s)
- Mara N Zeissig
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Duncan R Hewett
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Vasilios Panagopoulos
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Craig T Wallington-Gates
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| | - Andrew Spencer
- Department of Haematology, Alfred Health-Monash University, Melbourne, Australia
| | - Sandra M Dold
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Monika Engelhardt
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kate Vandyke
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia; Central Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
4
|
Williams CMD, Noll JE, Bradey AL, Duggan J, Wilczek VJ, Masavuli MG, Grubor‐Bauk B, Panagopoulos RA, Hewett DR, Mrozik KM, Zannettino ACW, Vandyke K, Panagopoulos V. Myeloperoxidase creates a permissive microenvironmental niche for the progression of multiple myeloma. Br J Haematol 2023; 203:614-624. [PMID: 37699574 PMCID: PMC10952523 DOI: 10.1111/bjh.19102] [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: 05/25/2023] [Revised: 08/07/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
Expression of myeloperoxidase (MPO), a key inflammatory enzyme restricted to myeloid cells, is negatively associated with the development of solid tumours. Activated myeloid cell populations are increased in multiple myeloma (MM); however, the functional consequences of myeloid-derived MPO within the myeloma microenvironment are unknown. Here, the role of MPO in MM pathogenesis was investigated, and the capacity for pharmacological inhibition of MPO to impede MM progression was evaluated. In the 5TGM1-KaLwRij mouse model of myeloma, the early stages of tumour development were associated with an increase in CD11b+ myeloid cell populations and an increase in Mpo expression within the bone marrow (BM). Interestingly, MM tumour cell homing was increased towards sites of elevated myeloid cell numbers and MPO activity within the BM. Mechanistically, MPO induced the expression of key MM growth factors, resulting in tumour cell proliferation and suppressed cytotoxic T-cell activity. Notably, tumour growth studies in mice treated with a small-molecule irreversible inhibitor of MPO (4-ABAH) demonstrated a significant reduction in overall MM tumour burden. Taken together, our data demonstrate that MPO contributes to MM tumour growth, and that MPO-specific inhibitors may provide a new therapeutic strategy to limit MM disease progression.
Collapse
Affiliation(s)
- Connor M. D. Williams
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Jacqueline E. Noll
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Alanah L. Bradey
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Jvaughn Duggan
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Vicki J. Wilczek
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Makutiro G. Masavuli
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health ResearchUniversity of AdelaideAdelaideAustralia
| | - Branka Grubor‐Bauk
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Health ResearchUniversity of AdelaideAdelaideAustralia
| | - Romana A. Panagopoulos
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
- Breast Cancer Research Unit, Discipline of Surgery, Basil Hetzel Institute for Translational Health ResearchUniversity of AdelaideAdelaideAustralia
| | - Duncan R. Hewett
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Krzysztof M. Mrozik
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Andrew C. W. Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Kate Vandyke
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Vasilios Panagopoulos
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, School of BiomedicineUniversity of AdelaideAdelaideAustralia
- Solid Tumour Program, Precision Cancer Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| |
Collapse
|
5
|
Development of 99mTc-Hynic-Adh-1 Molecular Probe Specifically Targeting N-Cadherin and Its Preliminary Experimental Study in Monitoring Drug Resistance of Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:cancers15030755. [PMID: 36765712 PMCID: PMC9913320 DOI: 10.3390/cancers15030755] [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: 12/20/2022] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND N-cadherin is considered a characteristic protein of EMT and has been found to be closely related to tumor resistance. In this study, a novel molecular imaging probe, 99mTc-HYNIC-ADH-1, was developed, and its diagnostic value in monitoring drug resistance in NSCLC was preliminarily investigated. METHODS ADH-1 was labeled indirectly with 99mTc. Radiochemical purity and stability, partition coefficients and pharmacokinetics were evaluated. Additionally, the fluorescent probe of ADH-1 was synthesized to study tumor uptake in cells level and in vivo. Biodistribution analysis and small animal SPECT/CT were performed in PC9GR and PC9 tumor-bearing mice. RESULTS 99mTc-HYNIC-ADH-1 was highly stable (radiochemical purity ≥ 98% in PBS and serum after 24 h). A cell binding study and fluorescence imaging showed that the uptake was significantly higher in PC9GR cells (gefitinib-resistant) than in PC9 cells (nonresistant) (p < 0.05). Biodistribution analysis showed rapid blood clearance and significant uptake in the kidney and resistant tumor. Small animal SPECT/CT studies showed that uptake in PC9GR tumors (T/NT = 7.73 ± 0.54) was significantly higher than that in PC9 tumors (T/NT = 3.66 ± 0.78) at 1 h (p = 0.002). CONCLUSIONS The 99mTc-HYNIC-ADH-1 molecular probe has a short synthesis time, high labeling rate, high radiochemical purity and good stability, does not require purification, is characterized by rapid blood clearance and is mainly excreted through the urinary system. 99mTc-HYNIC-ADH-1 is considered a promising probe for monitoring drug resistance in NSCLC.
Collapse
|
6
|
Zerdan MB, Nasr L, Kassab J, Saba L, Ghossein M, Yaghi M, Dominguez B, Chaulagain CP. Adhesion molecules in multiple myeloma oncogenesis and targeted therapy. Int J Hematol Oncol 2022; 11:IJH39. [PMID: 35663420 PMCID: PMC9136637 DOI: 10.2217/ijh-2021-0017] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Every day we march closer to finding the cure for multiple myeloma. The myeloma cells inflict their damage through specialized cellular meshwork and cytokines system. Implicit in these interactions are cellular adhesion molecules and their regulators which include but are not limited to integrins and syndecan-1/CD138, immunoglobulin superfamily cell adhesion molecules, such as CD44, cadherins such as N-cadherin, and selectins, such as E-selectin. Several adhesion molecules are respectively involved in myelomagenesis such as in the transition from the precursor disorder monoclonal gammopathy of undetermined significance to indolent asymptomatic multiple myeloma (smoldering myeloma) then to active multiple myeloma or primary plasma cell leukemia, and in the pathological manifestations of multiple myeloma.
Collapse
Affiliation(s)
- Maroun Bou Zerdan
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| | - Lewis Nasr
- Saint-Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Joseph Kassab
- Saint-Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Ludovic Saba
- Saint-Joseph University, Faculty of Medicine, Beirut, Lebanon
| | - Myriam Ghossein
- Department of Medicine & Medical Sciences, University of Balamand, Balamand, Lebanon
| | - Marita Yaghi
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| | - Barbara Dominguez
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| | - Chakra P Chaulagain
- Department of Hematology-Oncology, Myeloma & Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL 33331, USA
| |
Collapse
|
7
|
Ebert LM, Vandyke K, Johan MZ, DeNichilo M, Tan LY, Myo Min KK, Weimann BM, Ebert BW, Pitson SM, Zannettino ACW, Wallington-Beddoe CT, Bonder CS. Desmoglein-2 expression is an independent predictor of poor prognosis patients with multiple myeloma. Mol Oncol 2021; 16:1221-1240. [PMID: 34245117 PMCID: PMC8936512 DOI: 10.1002/1878-0261.13055] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy and is an incurable disease of neoplastic plasma cells (PC). Newly diagnosed MM patients currently undergo lengthy genetic testing to match chromosomal mutations with the most potent drug/s to decelerate disease progression. With only 17% of MM patients surviving 10‐years postdiagnosis, faster detection and earlier intervention would unequivocally improve outcomes. Here, we show that the cell surface protein desmoglein‐2 (DSG2) is overexpressed in ~ 20% of bone marrow biopsies from newly diagnosed MM patients. Importantly, DSG2 expression was strongly predictive of poor clinical outcome, with patients expressing DSG2 above the 70th percentile exhibiting an almost 3‐fold increased risk of death. As a prognostic factor, DSG2 is independent of genetic subtype as well as the routinely measured biomarkers of MM activity (e.g. paraprotein). Functional studies revealed a nonredundant role for DSG2 in adhesion of MM PC to endothelial cells. Together, our studies suggest DSG2 to be a potential cell surface biomarker that can be readily detected by flow cytometry to rapidly predict disease trajectory at the time of diagnosis.
Collapse
Affiliation(s)
- Lisa M Ebert
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Kate Vandyke
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Myeloma Research Laboratory, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - M Zahied Johan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Mark DeNichilo
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Lih Y Tan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Kay K Myo Min
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Benjamin M Weimann
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Brenton W Ebert
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Andrew C W Zannettino
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Myeloma Research Laboratory, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Craig T Wallington-Beddoe
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Flinders Medical Centre, Bedford Park, SA, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
8
|
Zeissig MN, Zannettino ACW, Vandyke K. Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma. Cancers (Basel) 2020; 12:cancers12123643. [PMID: 33291672 PMCID: PMC7761917 DOI: 10.3390/cancers12123643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Like in solid cancers, the process of dissemination is a critical feature of disease progression in the blood cancer multiple myeloma. At diagnosis, myeloma patients have cancer that has spread throughout the bone marrow, with patients with more disseminatory myeloma having worse outcomes for their disease. In this review, we discuss the current understanding of the mechanisms that underpin the dissemination process in multiple myeloma. Furthermore, we discuss the potential for the use of therapies that target the dissemination process as a novel means of improving outcomes for multiple myeloma patients. Abstract Multiple myeloma (MM) is a plasma cell (PC) malignancy characterised by the presence of MM PCs at multiple sites throughout the bone marrow. Increased numbers of peripheral blood MM PCs are associated with rapid disease progression, shorter time to relapse and are a feature of advanced disease. In this review, the current understanding of the process of MM PC dissemination and the extrinsic and intrinsic factors potentially driving it are addressed through analysis of patient-derived MM PCs and MM cell lines as well as mouse models of homing and dissemination. In addition, we discuss how patient cytogenetic subgroups that present with highly disseminated disease, such as t(4;14), t(14;16) and t(14;20), suggest that intrinsic properties of MM PC influence their ability to disseminate. Finally, we discuss the possibility of using therapeutic targeting of tumour dissemination to slow disease progression and prevent overt relapse.
Collapse
Affiliation(s)
- Mara N. Zeissig
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| | - Andrew C. W. Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- Central Adelaide Local Health Network, Adelaide 5000, Australia
- Centre for Cancer Biology, University of South Australia, Adelaide 5000, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- Correspondence: ; Tel.: +61-8-8128-4694
| |
Collapse
|
9
|
Multiplatform genomic profiling and magnetic resonance imaging identify mechanisms underlying intratumor heterogeneity in meningioma. Nat Commun 2020; 11:4803. [PMID: 32968068 PMCID: PMC7511976 DOI: 10.1038/s41467-020-18582-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
Meningiomas are the most common primary intracranial tumors, but the molecular drivers of meningioma tumorigenesis are poorly understood. We hypothesized that investigating intratumor heterogeneity in meningiomas would elucidate biologic drivers and reveal new targets for molecular therapy. To test this hypothesis, here we perform multiplatform molecular profiling of 86 spatially-distinct samples from 13 human meningiomas. Our data reveal that regional alterations in chromosome structure underlie clonal transcriptomic, epigenomic, and histopathologic signatures in meningioma. Stereotactic co-registration of sample coordinates to preoperative magnetic resonance images further suggest that high apparent diffusion coefficient (ADC) distinguishes meningioma regions with proliferating cells enriched for developmental gene expression programs. To understand the function of these genes in meningioma, we develop a human cerebral organoid model of meningioma and validate the high ADC marker genes CDH2 and PTPRZ1 as potential targets for meningioma therapy using live imaging, single cell RNA sequencing, CRISPR interference, and pharmacology.
Collapse
|
10
|
The long non-coding RNA CRNDE regulates growth of multiple myeloma cells via an effect on IL6 signalling. Leukemia 2020; 35:1710-1721. [PMID: 32879426 DOI: 10.1038/s41375-020-01034-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Multiple myeloma (MM) is a currently incurable malignancy of antibody-secreting plasma cells. Long non-coding RNAs (lncRNAs) have been recognised as an important class of regulatory molecules which are increasingly implicated in tumorigenesis. While recent studies have demonstrated changes in expression of lncRNAs in MM, the functional significance and molecular pathways downstream of these changes remain poorly characterised. In this study, we have performed CRISPR-mediated deletion of the locus encoding the lncRNA Colorectal Neoplasia Differentially Expressed (CRNDE), a known oncogenic lncRNA that is overexpressed in plasma cells of MM patients and is a marker of poor prognosis. We found that CRISPR-mediated deletion of the CRNDE locus in MM cells decreases proliferation and adhesion properties, increases sensitivity to Dexamethasone and reduces tumour growth in an in vivo xenograft model. Transcriptomic profiling in CRNDE-deleted MM cells demonstrated that CRNDE activates expression of a number of genes previously implicated in the aetiology of MM, including IL6R. We further demonstrate that deletion of the CRNDE locus diminishes IL6 signalling and proliferative responses in MM cells. Altogether this study reveals the IL6 signalling pathway as a novel mechanism by which CRNDE impacts upon MM cell growth and disease progression.
Collapse
|
11
|
Friend NL, Hewett DR, Panagopoulos V, Noll JE, Vandyke K, Mrozik KM, Fitter S, Zannettino AC. Characterization of the role of Samsn1 loss in multiple myeloma development. FASEB Bioadv 2020; 2:554-572. [PMID: 32923989 PMCID: PMC7475304 DOI: 10.1096/fba.2020-00027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 04/26/2020] [Accepted: 06/29/2020] [Indexed: 12/23/2022] Open
Abstract
The protein SAMSN1 was recently identified as a putative tumor suppressor in multiple myeloma, with re-expression of Samsn1 in the 5TGM1/KaLwRij murine model of myeloma leading to a near complete abrogation of intramedullary tumor growth. Here, we sought to clarify the mechanism underlying this finding. Intratibial administration of 5TGM1 myeloma cells into KaLwRij mice revealed that Samsn1 had no effect on primary tumor growth, but that its expression significantly inhibited the metastasis of these primary tumors. Notably, neither in vitro nor in vivo migration was affected by Samsn1 expression. Both knocking-out SAMSN1 in the RPMI-8226 and JJN3 human myeloma cell lines, and retrovirally expressing SAMSN1 in the LP-1 and OPM2 human myeloma cell lines had no effect on either cell proliferation or migration in vitro. Altering SAMSN1 expression in these human myeloma cells did not affect the capacity of the cells to establish either primary or metastatic intramedullary tumors when administered intratibially into immune deficient NSG mice. Unexpectedly, the tumor suppressive and anti-metastatic activity of Samsn1 in 5TGM1 cells were not evidenced following cell administration either intratibially or intravenously to NSG mice. Crucially, the growth of Samsn1-expressing 5TGM1 cells was limited in C57BL/6/Samsn1-/- mice but not in C57BL/6 Samsn1+/+ mice. We conclude that the reported potent in vivo tumor suppressor activity of Samsn1 can be attributed, in large part, to graft-rejection from Samsn1-/- recipient mice. This has broad implications for the design and interpretation of experiments that utilize cancer cells and knockout mice that are mismatched for expression of specific proteins.
Collapse
Affiliation(s)
- Natasha L. Friend
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Duncan R. Hewett
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Vasilios Panagopoulos
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Jacqueline E. Noll
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Kate Vandyke
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Krzysztof M. Mrozik
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Stephen Fitter
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
| | - Andrew C.W. Zannettino
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research InstituteAdelaideAustralia
- Central Adelaide Local Health NetworkAdelaideAustralia
| |
Collapse
|
12
|
Mrozik KM, Cheong CM, Hewett DR, Noll JE, Opperman KS, Adwal A, Russell DL, Blaschuk OW, Vandyke K, Zannettino ACW. LCRF-0006, a small molecule mimetic of the N-cadherin antagonist peptide ADH-1, synergistically increases multiple myeloma response to bortezomib. FASEB Bioadv 2020; 2:339-353. [PMID: 32617520 PMCID: PMC7325588 DOI: 10.1096/fba.2019-00073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
N-cadherin is a homophilic cell-cell adhesion molecule that plays a critical role in maintaining vascular stability and modulating endothelial barrier permeability. Pre-clinical studies have shown that the N-cadherin antagonist peptide, ADH-1, increases the permeability of tumor-associated vasculature thereby increasing anti-cancer drug delivery to tumors and enhancing tumor response. Small molecule library screens have identified a novel compound, LCRF-0006, that is a mimetic of the classical cadherin His-Ala-Val sequence-containing region of ADH-1. Here, we evaluated the vascular permeability-enhancing and anti-cancer properties of LCRF-0006 using in vitro vascular disruption and cell apoptosis assays, and a well-established pre-clinical model (C57BL/KaLwRij/5TGM1) of the hematological cancer multiple myeloma (MM). We found that LCRF-0006 disrupted endothelial cell junctions in a rapid, transient and reversible manner, and increased vascular permeability in vitro and at sites of MM tumor in vivo. Notably, LCRF-0006 synergistically increased the in vivo anti-MM tumor response to low-dose bortezomib, a frontline anti-MM agent, leading to regression of disease in 100% of mice. Moreover, LCRF-0006 and bortezomib synergistically induced 5TGM1 MM tumor cell apoptosis in vitro. Our findings demonstrate the potential clinical utility of LCRF-0006 to significantly increase bortezomib effectiveness and enhance the depth of tumor response in patients with MM.
Collapse
Affiliation(s)
- Krzysztof M. Mrozik
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Chee M. Cheong
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Duncan R. Hewett
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Jacqueline E. Noll
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Khatora S. Opperman
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Alaknanda Adwal
- Ovarian and Reproductive Cancer Biology LaboratoryRobinson Research InstituteThe University of AdelaideAdelaideAustralia
| | - Darryl L. Russell
- Ovarian and Reproductive Cancer Biology LaboratoryRobinson Research InstituteThe University of AdelaideAdelaideAustralia
| | - Orest W. Blaschuk
- Division of UrologyDepartment of SurgeryMcGill UniversityMontrealCanada
| | - Kate Vandyke
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
| | - Andrew C. W. Zannettino
- Myeloma Research LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Central Adelaide Local Health NetworkAdelaideAustralia
| |
Collapse
|
13
|
Cheong CM, Mrozik KM, Hewett DR, Bell E, Panagopoulos V, Noll JE, Licht JD, Gronthos S, Zannettino ACW, Vandyke K. Twist-1 is upregulated by NSD2 and contributes to tumour dissemination and an epithelial-mesenchymal transition-like gene expression signature in t(4;14)-positive multiple myeloma. Cancer Lett 2020; 475:99-108. [PMID: 32014459 DOI: 10.1016/j.canlet.2020.01.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/11/2022]
Abstract
Approximately 15% of patients with multiple myeloma (MM) harbour the t(4;14) chromosomal translocation, leading to the overexpression of the histone methyltransferase NSD2. Patients with this translocation display increased tumour dissemination, accelerated disease progression and rapid relapse. Using publicly available gene expression profile data from NSD2high (n = 135) and NSD2low (n = 878) MM patients, we identified 39 epithelial-mesenchymal transition (EMT)-associated genes which are overexpressed in NSD2high MM plasma cells. In addition, our analyses identified Twist-1 as a key transcription factor upregulated in NSD2high MM patients and t(4;14)-positive cell lines. Overexpression and knockdown studies confirmed that Twist-1 is involved in driving the expression of EMT-associated genes in the human MM cell line KMS11 and promoted the migration of myeloma cell lines in vitro. Notably, Twist-1 overexpression in the mouse MM cell line 5TGM1 significantly increased tumour dissemination in an intratibial tumour model. These findings demonstrate that Twist-1, downstream of NSD2, contributes to the induction of an EMT-like signature in t(4;14)-positive MM and enhances the dissemination of MM plasma cells in vivo, which may, in part, explain the aggressive disease features associated with t(4;14)-positive MM.
Collapse
Affiliation(s)
- Chee Man Cheong
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Duncan R Hewett
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Elyse Bell
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Vasilios Panagopoulos
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Jacqueline E Noll
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Jonathan D Licht
- Departments of Medicine, Biochemistry and Molecular Biology and University of Florida Health Cancer Center, The University of Florida, Gainesville, FL, USA
| | - Stan Gronthos
- Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia; Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, Australia.
| |
Collapse
|
14
|
Yu W, Yang L, Li T, Zhang Y. Cadherin Signaling in Cancer: Its Functions and Role as a Therapeutic Target. Front Oncol 2019; 9:989. [PMID: 31637214 PMCID: PMC6788064 DOI: 10.3389/fonc.2019.00989] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Cadherin family includes lists of transmembrane glycoproteins which mediate calcium-dependent cell-cell adhesion. Cadherin-mediated adhesion regulates cell growth and differentiation throughout life. Through the establishment of the cadherin-catenin complex, cadherins provide normal cell-cell adhesion and maintain homeostatic tissue architecture. In the process of cell recognition and adhesion, cadherins act as vital participators. As results, the disruption of cadherin signaling has significant implications on tumor formation and progression. Altered cadherin expression plays a vital role in tumorigenesis, tumor progression, angiogenesis, and tumor immune response. Based on ongoing research into the role of cadherin signaling in malignant tumors, cadherins are now being considered as potential targets for cancer therapies. This review will demonstrate the mechanisms of cadherin involvement in tumor progression, and consider the clinical significance of cadherins as therapeutic targets.
Collapse
Affiliation(s)
- Weina Yu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Ting Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China.,School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Opperman KS, Vandyke K, Clark KC, Coulter EA, Hewett DR, Mrozik KM, Schwarz N, Evdokiou A, Croucher PI, Psaltis PJ, Noll JE, Zannettino AC. Clodronate-Liposome Mediated Macrophage Depletion Abrogates Multiple Myeloma Tumor Establishment In Vivo. Neoplasia 2019; 21:777-787. [PMID: 31247457 PMCID: PMC6593350 DOI: 10.1016/j.neo.2019.05.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Multiple myeloma is a fatal plasma cell malignancy that is reliant on the bone marrow microenvironment. The bone marrow is comprised of numerous cells of mesenchymal and hemopoietic origin. Of these, macrophages have been implicated to play a role in myeloma disease progression, angiogenesis, and drug resistance; however, the role of macrophages in myeloma disease establishment remains unknown. In this study, the antimyeloma efficacy of clodronate-liposome treatment, which globally and transiently depletes macrophages, was evaluated in the well-established C57BL/KaLwRijHsd murine model of myeloma. Our studies show, for the first time, that clodronate-liposome pretreatment abrogates myeloma tumor development in vivo. Clodronate-liposome administration resulted in depletion of CD169+ bone marrow-resident macrophages. Flow cytometric analysis revealed that clodronate-liposome pretreatment impaired myeloma plasma cell homing and retention within the bone marrow 24 hours postmyeloma plasma cell inoculation. This was attributed in part to decreased levels of macrophage-derived insulin-like growth factor 1. Moreover, a single dose of clodronate-liposome led to a significant reduction in myeloma tumor burden in KaLwRij mice with established disease. Collectively, these findings support a role for CD169-expressing bone marrow-resident macrophages in myeloma disease establishment and progression and demonstrate the potential of targeting macrophages as a therapy for myeloma patients.
Collapse
Affiliation(s)
- Khatora S Opperman
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Kate Vandyke
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Kimberley C Clark
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Elizabeth A Coulter
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Duncan R Hewett
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Nisha Schwarz
- Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Andreas Evdokiou
- Discipline of Surgery, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Basil Hetzel Institute, 37 Woodville Road, Woodville, 5011
| | - Peter I Croucher
- Bone Biology Laboratory, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010
| | - Peter J Psaltis
- Heart and Vascular Health Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Jacqueline E Noll
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001
| | - Andrew Cw Zannettino
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, 5005; Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, 5001; Centre for Cancer Biology, University of South Australia and SA Pathology, PO Box 2471, Adelaide, 5001.
| |
Collapse
|
16
|
Jeong MH, Park SY, Lee SH, Seo J, Yoo JY, Park SH, Kim MJ, Lee S, Jang S, Choi HK, Lee JE, Shin SJ, Choi KC, Cheong JH, Yoon HG. EPB41L5 Mediates TGFβ-Induced Metastasis of Gastric Cancer. Clin Cancer Res 2019; 25:3617-3629. [PMID: 30814110 DOI: 10.1158/1078-0432.ccr-18-2959] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/27/2018] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Because of disease heterogeneity, limited studies on effective chemotherapies and therapeutic agents for advanced gastric cancer are available. Erythrocyte membrane protein band 4.1-like 5 (EPB41L5) has critical roles in renal and breast cancer metastasis. However, its role in metastatic gastric cancer remains unknown. EXPERIMENTAL DESIGN The specimens of 78 gastric cancer patients were analyzed by oligonucleotide microarray and survival analysis. In vitro experiments and metastatic mice models were used to assess the effects of EPB41L5 on gastric cancer metastasis. RESULTS Gastric cancer patients with high EPB41L5 levels had poor prognosis and low survival rate. Further, TGFβ1-induced EPB41L5 expression promoted gastric cancer cell migration and invasion by Smad-dependent TGFβ signaling. Phospho-Smad3 recruitment to the EPB41L5 promoter was significantly inhibited by a TGFβ inhibitor. EPB41L5 overexpression increased lung metastasis of gastric cancer cells in nude mice, which was completely reversed by anti-EPB41L5 monoclonal antibody treatment. Importantly, p120-catenin knockdown abolished EPB41L5-enhanced gastric cancer cell metastasis. Anti-EPB41L5 monoclonal antibody treatment blocked the association of EPB41L5 with p120-catenin. CONCLUSIONS TGFβ/EPB41L5/p120-catenin axis regulates gastric cancer cell metastasis, and EPB41L5 is a promising therapeutic target for advanced gastric cancer.
Collapse
Affiliation(s)
- Mi-Hyeon Jeong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Yeon Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSung Seo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Yoon Yoo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Ho Park
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Jeong Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - SooYeon Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Subhin Jang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo-Kyoung Choi
- Division of Nutrition and Metabolism Research Group, Korea Food Research Institute, Gyeonggi-do, Korea
| | - Jae Eun Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Kyung-Chul Choi
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, Korea.
| | - Jae-Ho Cheong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea. .,Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
17
|
Cultrara CN, Kozuch SD, Ramasundaram P, Heller CJ, Shah S, Beck AE, Sabatino D, Zilberberg J. GRP78 modulates cell adhesion markers in prostate Cancer and multiple myeloma cell lines. BMC Cancer 2018; 18:1263. [PMID: 30563499 PMCID: PMC6299583 DOI: 10.1186/s12885-018-5178-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022] Open
Abstract
Background Glucose regulated protein 78 (GRP78) is a resident chaperone of the endoplasmic reticulum and a master regulator of the unfolded protein response under physiological and pathological cell stress conditions. GRP78 is overexpressed in many cancers, regulating a variety of signaling pathways associated with tumor initiation, proliferation, adhesion and invasion which contributes to metastatic spread. GRP78 can also regulate cell survival and apoptotic pathways to alter responsiveness to anticancer drugs. Tumors that reside in or metastasize to the bone and bone marrow (BM) space can develop pro-survival signals through their direct adhesive interactions with stromal elements of this niche thereby resisting the cytotoxic effects of drug treatment. In this study, we report a direct correlation between GRP78 and the adhesion molecule N-cadherin (N-cad), known to play a critical role in the adhesive interactions of multiple myeloma and metastatic prostate cancer with the bone microenvironment. Methods N-cad expression levels (transcription and protein) were evaluated upon siRNA mediated silencing of GRP78 in the MM.1S multiple myeloma and the PC3 metastatic prostate cancer cell lines. Furthermore, we evaluated the effects of GRP78 knockdown (KD) on epithelial-mesenchymal (EMT) transition markers, morphological changes and adhesion of PC3 cells. Results GRP78 KD led to concomitant downregulation of N-cad in both tumors types. In PC3 cells, GRP78 KD significantly decreased E-cadherin (E-cad) expression likely associated with the induction in TGF-β1 expression. Furthermore, GRP78 KD also triggered drastic changes in PC3 cells morphology and decreased their adhesion to osteoblasts (OSB) dependent, in part, to the reduced N-cad expression. Conclusion This work implicates GRP78 as a modulator of cell adhesion markers in MM and PCa. Our results may have clinical implications underscoring GRP78 as a potential therapeutic target to reduce the adhesive nature of metastatic tumors to the bone niche. Electronic supplementary material The online version of this article (10.1186/s12885-018-5178-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher N Cultrara
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Stephen D Kozuch
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Poornema Ramasundaram
- Center for Discovery and Innovation, Hackensack University Medical Center, 340 Kingsland Street, Building 102, Nutley, NJ, 07110, USA
| | - Claudia J Heller
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Sunil Shah
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Adah E Beck
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - David Sabatino
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Jenny Zilberberg
- Center for Discovery and Innovation, Hackensack University Medical Center, 340 Kingsland Street, Building 102, Nutley, NJ, 07110, USA.
| |
Collapse
|
18
|
Mrozik KM, Blaschuk OW, Cheong CM, Zannettino ACW, Vandyke K. N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer 2018; 18:939. [PMID: 30285678 PMCID: PMC6167798 DOI: 10.1186/s12885-018-4845-0] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
In many types of solid tumours, the aberrant expression of the cell adhesion molecule N-cadherin is a hallmark of epithelial-to-mesenchymal transition, resulting in the acquisition of an aggressive tumour phenotype. This transition endows tumour cells with the capacity to escape from the confines of the primary tumour and metastasise to secondary sites. In this review, we will discuss how N-cadherin actively promotes the metastatic behaviour of tumour cells, including its involvement in critical signalling pathways which mediate these events. In addition, we will explore the emerging role of N-cadherin in haematological malignancies, including bone marrow homing and microenvironmental protection to anti-cancer agents. Finally, we will discuss the evidence that N-cadherin may be a viable therapeutic target to inhibit cancer metastasis and increase tumour cell sensitivity to existing anti-cancer therapies.
Collapse
Affiliation(s)
- Krzysztof Marek Mrozik
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | | | - Chee Man Cheong
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andrew Christopher William Zannettino
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.,Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia. .,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.
| |
Collapse
|
19
|
Effects of N-cadherin on neuronal migration during chicken optic tectum development. Histochem Cell Biol 2018; 151:239-248. [PMID: 30250974 DOI: 10.1007/s00418-018-1733-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2018] [Indexed: 10/28/2022]
Abstract
N-cadherin, a member of the cadherin family, plays an important role in neural development. In addition, N-cadherin has been reported to be crucial in neuronal migration, axonal outgrowth, and axonal path-finding. However, the mechanism underlying the effects of N-cadherin in neuronal migration is not entirely clear. In this study, we investigated the overexpression or knockdown of N-cadherin in the optic tectum during chicken embryo development, and then analyzed the effect of N-cadherin on neuronal migration. The results showed that compared with the control group, in the N-cadherin knockdown group, the neuronal migration of the optic tectum was significantly affected and could not arrive at destination. The stratum griseum central layer of the optic tectum mainly includes multipolar neurons, which could not be formed after the knockdown of N-cadherin, and more neurons form the bipolar or monopolar neurons compared with the control group. Compared with the control group, more cells stayed in the neuroepithelium layer. The axonal length in the optic tectum was significantly (P < 0.001) shorter in the N-cadherin knockdown group than in the control group. These results reveal that the knockdown of N-cadherin mainly affects the length of axons and formation of multipolar neurons in the development of the chicken optic tectum, which eventually results in the inhibition of neuronal migration.
Collapse
|
20
|
Wallington-Beddoe CT, Bennett MK, Vandyke K, Davies L, Zebol JR, Moretti PAB, Pitman MR, Hewett DR, Zannettino ACW, Pitson SM. Sphingosine kinase 2 inhibition synergises with bortezomib to target myeloma by enhancing endoplasmic reticulum stress. Oncotarget 2018; 8:43602-43616. [PMID: 28467788 PMCID: PMC5546428 DOI: 10.18632/oncotarget.17115] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 12/22/2022] Open
Abstract
The proteasome inhibitor bortezomib has proven to be invaluable in the treatment of myeloma. By exploiting the inherent high immunoglobulin protein production of malignant plasma cells, bortezomib induces endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), resulting in myeloma cell death. In most cases, however, the disease remains incurable highlighting the need for new therapeutic targets. Sphingosine kinase 2 (SK2) has been proposed as one such therapeutic target for myeloma. Our observations that bortezomib and SK2 inhibitors independently elicited induction of ER stress and the UPR prompted us to examine potential synergy between these agents in myeloma. Targeting SK2 synergistically contributed to ER stress and UPR activation induced by bortezomib, as evidenced by activation of the IRE1 pathway and stress kinases JNK and p38MAPK, thereby resulting in potent synergistic myeloma apoptosis in vitro. The combination of bortezomib and SK2 inhibition also exhibited strong in vivo synergy and favourable effects on bone disease. Therefore, our studies suggest that perturbations of sphingolipid signalling can synergistically enhance the effects seen with proteasome inhibition, highlighting the potential for the combination of these two modes of increasing ER stress to be formally evaluated in clinical trials for the treatment of myeloma patients.
Collapse
Affiliation(s)
- Craig T Wallington-Beddoe
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Australia
| | - Melissa K Bennett
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Australia
| | - Kate Vandyke
- SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Australia.,South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Lorena Davies
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia
| | - Julia R Zebol
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia
| | - Paul A B Moretti
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia
| | - Melissa R Pitman
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia
| | - Duncan R Hewett
- School of Medicine, University of Adelaide, Australia.,South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andrew C W Zannettino
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Australia.,South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.,SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Australia
| |
Collapse
|
21
|
Active enhancer and chromatin accessibility landscapes chart the regulatory network of primary multiple myeloma. Blood 2018. [PMID: 29519805 DOI: 10.1182/blood-2017-09-808063] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma (MM) is an aggressive cancer that originates from antibody-secreting plasma cells. Although genetically and transcriptionally well characterized, the aberrant gene regulatory networks that underpin this disease remain poorly understood. Here, we mapped regulatory elements, open chromatin, and transcription factor (TF) footprints in primary MM cells. In comparison with normal antibody-secreting cells, MM cells displayed consistent changes in enhancer activity that are connected to superenhancer (SE)-mediated deregulation of TF genes. MM cells also displayed widespread decompaction of heterochromatin that was associated with activation of regulatory elements and in a major subset of patients' deregulation of the cyclic adenosine monophosphate pathway. Finally, building SE-associated TF-based regulatory networks allowed identification of several novel TFs that are central to MM biology. Taken together, these findings significantly add to our understanding of the aberrant gene regulatory network that underpins MM.
Collapse
|
22
|
Christopoulos PF, Sfikakis PP, Nikolaou E, Terpos E, Koutsilieris M, Kyrtsonis MC. Cadherin-11 (CDH11) expression in the peripheral blood of patients with active Multiple Myeloma. Br J Haematol 2016; 177:813-816. [PMID: 27118219 DOI: 10.1111/bjh.14105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Panagiotis F Christopoulos
- First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eftychia Nikolaou
- First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Marie-Christine Kyrtsonis
- First Department of Propaedeutic and Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
23
|
Jiang R, Zhao C, Wang X, Wang S, Sun X, Tian Y, Song W. Resistin-Like Molecule-β Promotes Invasion and Migration of Gastric Carcinoma Cells. Med Sci Monit 2016; 22:937-42. [PMID: 27001185 PMCID: PMC4806584 DOI: 10.12659/msm.895598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Resistin-like molecule-β (RELMβ) is a novel secretory protein from intestinal goblet cells and participates in epithelial differentiation, tumor occurrence, and immune response. RELMβ is absent in normal gastric mucosa but is abundantly expressed in gastric carcinoma tissues, and is correlated with tumor invasion and metastasis. Epithelial-mesenchymal transition (EMT) is an important mechanism governing tumor cell invasion. This study thus investigated the modulation of RELMβ in gastric cancer metastasis and its correlation with EMT. Material/Methods We used RELMβ-low expression AGS cell line of gastric cancer and normal mucosa cell line GES1 as in vitro models, on which RELMβ0-expressing vector was transfected. The invasion and migration of cells were quantified by Transwell assay. EMT-related protein including E-cadherin, N-cadherin, Snail, and Vimentin were detected by Western blotting in transfected AGS cells. Results RELMβ transfection significantly potentiated invasion and migration abilities of AGS cells, whose RELMβ protein level was significantly elevated compared to those in untransfected AGS or GES1 cells. After RELMβ transfection, EMT-related proteins, including N-cadherin, Snail, and Vimentin levels, were elevated, but E-cadherin expression was depressed. Conclusions RELMβ-overexpression can facilitate invasion and migration of gastric carcinoma cells and it increases the expression of EMT-related proteins, such as N-cadherin, Snail, Vimentin, but decreases E-cadherin level, thus promoting the progression of EMT.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Chunming Zhao
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Xinyu Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Shengxi Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaogang Sun
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Yang Tian
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Wei Song
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
24
|
Noll JE, Vandyke K, Hewett DR, Mrozik KM, Bala RJ, Williams SA, Kok CH, Zannettino AC. PTTG1 expression is associated with hyperproliferative disease and poor prognosis in multiple myeloma. J Hematol Oncol 2015; 8:106. [PMID: 26445238 PMCID: PMC4595141 DOI: 10.1186/s13045-015-0209-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/28/2015] [Indexed: 01/08/2023] Open
Abstract
Background Multiple myeloma (MM) is an incurable haematological malignancy characterised by the clonal proliferation of malignant plasma cells within the bone marrow. We have previously identified pituitary tumour transforming gene 1 (Pttg1) as a gene that is significantly upregulated in the haematopoietic compartment of the myeloma-susceptible C57BL/KaLwRij mouse strain, when compared with the myeloma-resistant C57BL/6 mouse. Over-expression of PTTG1 has previously been associated with malignant progression and an enhanced proliferative capacity in solid tumours. Methods In this study, we investigated PTTG1 gene and protein expression in MM plasma cells from newly diagnosed MM patients. Gene expression profiling was used to identify gene signatures associated with high PTTG1 expression in MM patients. Additionally, we investigated the effect of short hairpin ribonucleic acid (shRNA)-mediated PTTG1 knockdown on the proliferation of the murine myeloma plasma cell line 5TGM1 in vitro and in vivo. Results PTTG1 was found to be over-expressed in 36–70 % of MM patients, relative to normal controls, with high PTTG1 expression being associated with poor patient outcomes (hazard ratio 2.49; 95 % CI 1.28 to 4.86; p = 0.0075; log-rank test). In addition, patients with high PTTG1 expression exhibited increased expression of cell proliferation-associated genes including CCNB1, CCNB2, CDK1, AURKA, BIRC5 and DEPDC1. Knockdown of Pttg1 in 5TGM1 cells decreased cellular proliferation, without affecting cell cycle distribution or viability, and decreased expression of Ccnb1, Birc5 and Depdc1 in vitro. Notably, Pttg1 knockdown significantly reduced MM tumour development in vivo, with an 83.2 % reduction in tumour burden at 4 weeks (p < 0.0001, two-way ANOVA). Conclusions This study supports a role for increased PTTG1 expression in augmenting tumour development in a subset of MM patients. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0209-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacqueline E Noll
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Kate Vandyke
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia. .,SA Pathology, Adelaide, Australia.
| | - Duncan R Hewett
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Rachel J Bala
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Sharon A Williams
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Chung H Kok
- Leukaemia Research Group, Cancer Theme, SAHMRI, Adelaide, Australia.
| | - Andrew Cw Zannettino
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia. .,Discipline of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide, Cancer Theme, Level 5 South, SAHMRI, PO Box 11060, Adelaide, SA, 5001, Australia.
| |
Collapse
|