1
|
Bhansali D, Tu NH, Inoue K, Teng S, Li T, Tran HD, Kim DH, Dong J, Peach CJ, Sokrat B, Jensen DD, Dolan JC, Yamano S, Robinson VM, Bunnett NW, Albertson DG, Leong KW, Schmidt BL. PAR 2 on oral cancer cells and nociceptors contributes to oral cancer pain that can be relieved by nanoparticle-encapsulated AZ3451. Biomaterials 2025; 314:122874. [PMID: 39418848 DOI: 10.1016/j.biomaterials.2024.122874] [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: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
Oral cancer is notoriously painful. Activation of protease-activated receptor 2 (PAR2, encoded by F2RL1) by proteases in the cancer microenvironment is implicated in oral cancer pain. PAR2 is a G protein-coupled receptor (GPCR) expressed on neurons and cells in the cancer microenvironment. Sustained signaling of PAR2 from endosomes of neurons mediates sensitization and nociception. We focused on the differential contribution of PAR2 on oral cancer cells and neurons to oral cancer pain and whether encapsulation of a PAR2 inhibitor, AZ3451 in nanoparticles (NP) more effectively reverses PAR2 activation. We report that F2RL1 was overexpressed in human oral cancers and cancer cell lines. Deletion of F2RL1 on cancer cells reduced cancer-associated mechanical allodynia. A third-generation polyamidoamine dendrimer, functionalized with cholesterol was self-assembled into NPs encapsulating AZ3451. NP encapsulated AZ3451 (PAMAM-Chol-AZ NPs) more effectively reversed activation of PAR2 at the plasma membrane and early endosomes than free drug. The PAMAM-Chol-AZ NPs showed greater efficacy in reversing nociception than free drug, with respect to both level and duration, in three preclinical mouse models of oral cancer pain. The antinociceptive efficacy was confirmed with an operant orofacial assay. Genetic deletion of F2RL1 on cancer cells or F2rl1 on neurons each partially reversed mechanical cancer allodynia. The remaining nociception could be effectively reversed by PAMAM-Chol-AZ NPs. These findings suggest that PAR2 on oral cancer cells and neurons contribute to oral cancer nociception and NPs loaded with a PAR2 antagonist provide increased antinociception and improved oral function compared to free drug.
Collapse
Affiliation(s)
- Divya Bhansali
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| | - Nguyen H Tu
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA; Pain Research Center, New York University, New York, NY, 10010, USA.
| | - Kenji Inoue
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA; Pain Research Center, New York University, New York, NY, 10010, USA.
| | - Shavonne Teng
- Pain Research Center, New York University, New York, NY, 10010, USA; Department of Molecular Pathobiology, New York University Dentistry, New York, NY, 10010, USA; Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| | - Hung D Tran
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA.
| | - Dong H Kim
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA.
| | - Jessy Dong
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA.
| | - Chloe J Peach
- Pain Research Center, New York University, New York, NY, 10010, USA; Department of Molecular Pathobiology, New York University Dentistry, New York, NY, 10010, USA; Centre of Membrane Proteins and Receptors, School of Life Sciences, Queen's Medical Centre, University of Nottingham, UK.
| | - Badr Sokrat
- Pain Research Center, New York University, New York, NY, 10010, USA; Department of Molecular Pathobiology, New York University Dentistry, New York, NY, 10010, USA.
| | - Dane D Jensen
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA; Pain Research Center, New York University, New York, NY, 10010, USA.
| | - John C Dolan
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA.
| | - Seiichi Yamano
- Department of Prosthodontics, New York University Dentistry, New York, NY, 10010, USA.
| | - Valeria Mezzano Robinson
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| | - Nigel W Bunnett
- Pain Research Center, New York University, New York, NY, 10010, USA; Department of Molecular Pathobiology, New York University Dentistry, New York, NY, 10010, USA; Neuroscience Institute, New York University Langone Health, New York, NY, 10016, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| | - Donna G Albertson
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA; Pain Research Center, New York University, New York, NY, 10010, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| | - Brian L Schmidt
- New York University Dentistry Translational Research Center, New York University Dentistry, New York, NY, 10010, USA; Pain Research Center, New York University, New York, NY, 10010, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
2
|
Petrescu DI, Yustein JT, Dasgupta A. Preclinical models for the study of pediatric solid tumors: focus on bone sarcomas. Front Oncol 2024; 14:1388484. [PMID: 39091911 PMCID: PMC11291195 DOI: 10.3389/fonc.2024.1388484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Sarcomas comprise between 10-15% of all pediatric malignancies. Osteosarcoma and Ewing sarcoma are the two most common pediatric bone tumors diagnosed in children and young adults. These tumors are commonly treated with surgery and/or radiation therapy and combination chemotherapy. However, there is a strong need for the development and utilization of targeted therapeutic methods to improve patient outcomes. Towards accomplishing this goal, pre-clinical models for these unique malignancies are of particular importance to design and test experimental therapeutic strategies prior to being introduced to patients due to their origination site and propensity to metastasize. Pre-clinical models offer several advantages for the study of pediatric sarcomas with unique benefits and shortcomings dependent on the type of model. This review addresses the types of pre-clinical models available for the study of pediatric solid tumors, with special attention to the bone sarcomas osteosarcoma and Ewing sarcoma.
Collapse
Affiliation(s)
- D. Isabel Petrescu
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - Atreyi Dasgupta
- The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Texas Children’s Cancer and Hematology Centers, Houston, TX, United States
| |
Collapse
|
3
|
Singh V, Chernatynskaya A, Qi L, Chuang HY, Cole T, Jeyalatha VM, Bhargava L, Yeudall WA, Farkas L, Yang H. Liposomes-Encapsulating Double-Stranded Nucleic Acid (Poly I:C) for Head and Neck Cancer Treatment. ACS Pharmacol Transl Sci 2024; 7:1612-1623. [PMID: 38751634 PMCID: PMC11092114 DOI: 10.1021/acsptsci.4c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 05/18/2024]
Abstract
Polyriboinosinic acid-polyribocytidylic acid (Poly I:C) serves as a synthetic mimic of viral double-stranded dsRNA, capable of inducing apoptosis in numerous cancer cells. Despite its potential, therapeutic benefits, the application of Poly I:C has been hindered by concerns regarding toxicity, stability, enzymatic degradation, and undue immune stimulation, leading to autoimmune disorders. To address these challenges, encapsulation of antitumor drugs within delivery systems such as cationic liposomes is often employed to enhance their efficacy while minimizing dosages. In this study, we investigated the potential of cationic liposomes to deliver Poly I:C into the Head and Neck 12 (HN12) cell line to induce apoptosis in the carcinoma cells and tumor model. Cationic liposomes made by the hydrodynamic focusing method surpass traditional methods by offering a continuous flow-based approach for encapsulating genes, which is ideal for efficient tumor delivery. DOTAP liposomes efficiently bind Poly I:C, confirmed by transmission electron microscopy images displaying their spherical morphology. Liposomes are easily endocytosed in HN12 cells, suggesting their potential for therapeutic gene and drug delivery in head and neck squamous carcinoma cells. Activation of apoptotic pathways involving MDA5, RIG-I, and TLR3 is evidenced by upregulated caspase-3, caspase-8, and IRF3 genes upon endocytosis of Poly(I:C)-encapsulated liposomes. Therapeutic evaluations revealed significant inhibition of tumor growth with Poly I:C liposomes, indicating the possibility of MDA5, RIG-I, and TLR3-induced apoptosis pathways via Poly I:C liposomes in HN12 xenografts in J:NU mouse models. Comparative histological analysis underscores enhanced cell death with Poly I:C liposomes, warranting further investigation into the precise mechanisms of apoptosis and inflammatory cytokine response in murine models for future research.
Collapse
Affiliation(s)
- Vidit Singh
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - Anna Chernatynskaya
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - Lin Qi
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - Hsin-Yin Chuang
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - Tristan Cole
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - Vimalin Mani Jeyalatha
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - Lavanya Bhargava
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| | - W. Andrew Yeudall
- Dental
College of Georgia, Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta 30912, Georgia, United States
| | - Laszlo Farkas
- Division
of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, Ohio State University, Columbus 43210-1132, Ohio, United States
| | - Hu Yang
- Linda
and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla 65409, Missouri, United States
| |
Collapse
|
4
|
Luo YL, Li YF, Zhu YP, Liang JQ, Chen DF, Chen HJ, Huang CQ, Li J, Li SY. Effects of human umbilical cord blood mononuclear cells on ovalbumin-induced asthma in mice. J Thorac Dis 2023; 15:5454-5465. [PMID: 37969283 PMCID: PMC10636480 DOI: 10.21037/jtd-22-1669] [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: 11/22/2022] [Accepted: 09/01/2023] [Indexed: 11/17/2023]
Abstract
Background Umbilical cord blood mononuclear cells (UCMNCs) show broad immune-modulation effects, which may be helpful for treating asthma. Effects of UCMNCs on asthma were investigated with mouse model in present study. Methods Asthma was induced in BALB/c mice by ovalbumin (OVA) immunization and challenge. Asthmatic mice were then treated on days 7 and 20 with intravenous injections of UCMNCs in doses of 4×105, 2×106, and 107 cells per mouse for the low-dose UCMNC (UCMNCL), medium-dose UCMNC (UCMNCM), and high-dose UCMNC (UCMNCH) groups, respectively. Fetal mouse blood mononuclear cells (FMMNCs) were administered to FMMNC group at a dose of 2×106 cells per mouse as approximate allograft control. Airway hyperresponsiveness (AHR), airway inflammation indexes, and CD4/CD8 T cell subsets were measured at day 25. Results Compared with the model group, AHR in the UCMNCL group, inflammation score of lung tissue in the UCMNCM group, interleukin (IL)-5 in bronchoalveolar lavage fluid (BALF) in UCMNCL group, IL-5 and IL-13 in BALF in UCMNCM group, and IL-17 in serum in UCMNCH group were significantly inhibited. Compared with the model group, CD4+CD8+ T cells were reduced in the UCMNCL group, while decrease of CD4-CD8- T cells and increase of CD4+CD8- T cells were further strengthened in UCMNCM group. FMMNC treatment significantly reduced the IL-13 and IL-17 in serum, decreased CD4-CD8- and CD4+CD8- T cells, and increased the CD4+CD8+ and CD4-CD8+ T cells in BALF. Conclusions UCMNCs can modulate AHR, T-helper (Th)2 inflammation, and airway injury in experimental asthma at appropriate dose.
Collapse
Affiliation(s)
- Yu-Long Luo
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun-Feng Li
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Yi-Ping Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin-Qiang Liang
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, China
| | - Di-Fei Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huan-Jie Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chu-Qing Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shi-Yue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Iwasaki M, Zhao H, Hu C, Saito J, Wu L, Sherwin A, Ishikawa M, Sakamoto A, Buggy D, Ma D. The differential cancer growth associated with anaesthetics in a cancer xenograft model of mice: mechanisms and implications of postoperative cancer recurrence. Cell Biol Toxicol 2023; 39:1561-1575. [PMID: 35953652 PMCID: PMC10425502 DOI: 10.1007/s10565-022-09747-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022]
Abstract
Anaesthetics may modify colorectal cancer cell biology which potentially affects long-term survival. This study aims to compare propofol and sevoflurane regarding with the direct anaesthetic effects on cancer malignancy and the indirect effects on host immunity in a cancer xenograft mode of mice. Cultured colon cancer cell (Caco-2) was injected subcutaneously to nude mice (day 1). Mice were exposed to either 1.5% sevoflurane for 1.5 h or propofol (20 μg g-1; ip injection) with or without 4 μg g-1 lipopolysaccharide (LPS; ip) from days 15 to 17, compared with those without anaesthetic exposure as controls. The clinical endpoints including tumour volumes over 70 mm3 were closely monitored up to day 28. Tumour samples from the other cohorts were collected on day 18 for PCR array, qRT-PCR, western blotting and immunofluorescent assessment. Propofol treatment reduced tumour size (mean ± SD; 23.0 ± 6.2mm3) when compared to sevoflurane (36.0 ± 0.3mm3) (p = 0.008) or control (23.6 ± 4.7mm3). Propofol decreased hypoxia inducible factor 1α (HIF1α), interleukin 1β (IL1β), and hepatocyte growth factor (HGF) gene expressions and increased tissue inhibitor of metalloproteinases 2 (TIMP-2) gene and protein expression in comparison to sevoflurane in the tumour tissue. LPS suppressed tumour growth in any conditions whilst increased TIMP-2 and anti-cancer neutrophil marker expressions and decreased macrophage marker expressions compared to those in the LPS-untreated groups. Our data indicated that sevoflurane increased cancer development when compared with propofol in vivo under non-surgical condition. Anaesthetics tested in this study did not alter the effects of LPS as an immune modulator in changing immunocyte phenotype and suppressing cancer development.
Collapse
Affiliation(s)
- Masae Iwasaki
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
- Department of Anaesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
| | - Cong Hu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
| | - Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Lingzhi Wu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
| | - Aislinn Sherwin
- Anaesthesiology and Perioperative Medicine, Mater University Hospital, University College Dublin, Dublin, Ireland
| | - Masashi Ishikawa
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
- Department of Anaesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Atsuhiro Sakamoto
- Department of Anaesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Donal Buggy
- Anaesthesiology and Perioperative Medicine, Mater University Hospital, University College Dublin, Dublin, Ireland
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Rd, Chelsea, London, SW10 9NH UK
| |
Collapse
|
6
|
Souto-Lopes M, Grenho L, Manrique YA, Dias MM, Fernandes MH, Monteiro FJ, Salgado CL. Full physicochemical and biocompatibility characterization of a supercritical CO 2 sterilized nano-hydroxyapatite/chitosan biodegradable scaffold for periodontal bone regeneration. BIOMATERIALS ADVANCES 2023; 146:213280. [PMID: 36682201 DOI: 10.1016/j.bioadv.2023.213280] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/07/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023]
Abstract
Despite bone's innate self-renewal capability, some periodontal pathologic and traumatic defects' size inhibits full spontaneous regeneration. This current research characterized a 3D porous biodegradable nano-hydroxyapatite/chitosan (nHAp/CS, 70/30) scaffold for periodontal bone regeneration, which preparation method includes the final solvent extraction and sterilization through supercritical CO2 (scCO2). Micro-CT analysis revealed the fully interconnected porous microstructure of the nHAp/CS scaffold (total porosity 78 %, medium pore size 200 μm) which is critical for bone regeneration. Scanning electron microscopy (SEM) showed HAp crystals forming on the surface of the nHAp/CS scaffold after 21 days in simulated body fluid, demonstrating its bioactivity in vitro. The presence of nHAp in the scaffolds promoted a significantly lower biodegradation rate compared to a plain CS scaffold in PBS. Dynamic mechanical analysis confirmed their viscoelasticity, but the presence of nHAp significantly enhanced the storage modulus (42.34 ± 6.09 kPa at 10 Hz after 28 days in PBS), showing that it may support bone ingrowth at low-load bearing bone defects. Both scaffold types significantly inhibited the growth, attachment and colony formation abilities of S. aureus and E. coli, enhancing the relevance of chitosan in the grafts' composition for the naturally contaminated oral environment. At SEM and laser scanning confocal microscopy, MG63 cells showed normal morphology and could adhere and proliferate inside the biomaterials' porous structure, especially for the nHAp/CS scaffold, reaching higher proliferative rate at day 14. MG63 cells seeded within nHAp/CS scaffolds presented a higher expression of RUNX2, collagen A1 and Sp7 osteogenic genes compared to the CS samples. The in vivo subcutaneous implantation in mice of both scaffold types showed lower biodegradability with the preservation of the scaffolds porous structure that allowed the ingrowth of connective tissue until 5 weeks. Histology shows an intensive and progressive ingrowth of new vessels and collagen between the 3rd and the 5th week, especially for the nHAp/CS scaffold. So far, the scCO2 method enabled the production of a cost-effective and environment-friendly ready-to-use nHAp/CS scaffold with microstructural, chemical, mechanical and biocompatibility features that make it a suitable bone graft alternative for defect sites in an adverse environment as in periodontitis and peri-implantitis.
Collapse
Affiliation(s)
- Mariana Souto-Lopes
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Liliana Grenho
- Faculty of Dental Medicine of the University of Porto, R. Dr. Manuel Pereira da Silva, 4200-393 Porto, Portugal; LAQV/REQUIMTE - Laboratório Associado para a Química Verde/Rede de Química e Tecnologia, Portugal
| | - Yaidelin Alves Manrique
- LSRE-LCM - Laboratory of Separation and Reaction Engineering - Laboratory of Catalysis and Materials, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Madalena Maria Dias
- LSRE-LCM - Laboratory of Separation and Reaction Engineering - Laboratory of Catalysis and Materials, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria Helena Fernandes
- Faculty of Dental Medicine of the University of Porto, R. Dr. Manuel Pereira da Silva, 4200-393 Porto, Portugal; LAQV/REQUIMTE - Laboratório Associado para a Química Verde/Rede de Química e Tecnologia, Portugal
| | - Fernando Jorge Monteiro
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Christiane Laranjo Salgado
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
7
|
Shigehiro T, Ueno M, Kijihira M, Takahashi R, Umemura C, Taha EA, Kurosaka C, Asayama M, Murakami H, Satoh A, Nakamura Y, Futami J, Masuda J. Immune State Conversion of the Mesenteric Lymph Node in a Mouse Breast Cancer Model. Int J Mol Sci 2022; 23:ijms231911035. [PMID: 36232335 PMCID: PMC9570492 DOI: 10.3390/ijms231911035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Secondary lymphoid tissues, such as the spleen and lymph nodes (LNs), contribute to breast cancer development and metastasis in both anti- and pro-tumoral directions. Although secondary lymphoid tissues have been extensively studied, very little is known about the immune conversion in mesenteric LNs (mLNs) during breast cancer development. Here, we demonstrate inflammatory immune conversion of mLNs in a metastatic 4T1 breast cancer model. Splenic T cells were significantly decreased and continuously suppressed IFN-γ production during tumor development, while myeloid-derived suppressor cells (MDSCs) were dramatically enriched. However, T cell numbers in the mLN did not decrease, and the MDSCs only moderately increased. T cells in the mLN exhibited conversion from a pro-inflammatory state with high IFN-γ expression to an anti-inflammatory state with high expression of IL-4 and IL-10 in early- to late-stages of breast cancer development. Interestingly, increased migration of CD103+CD11b+ dendritic cells (DCs) into the mLN, along with increased (1→3)-β-D-glucan levels in serum, was observed even in late-stage breast cancer. This suggests that CD103+CD11b+ DCs could prime cancer-reactive T cells. Together, the data indicate that the mLN is an important lymphoid tissue contributing to breast cancer development.
Collapse
Affiliation(s)
- Tsukasa Shigehiro
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| | - Maho Ueno
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Mayumi Kijihira
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Ryotaro Takahashi
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
| | - Chiho Umemura
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Eman A. Taha
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Chisaki Kurosaka
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Megumi Asayama
- Department of Applied Chemistry and Biotechnology, Faculty of Engineering, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Murakami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Ayano Satoh
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Junichiro Futami
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
| | - Junko Masuda
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan
- Division of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Department of Pharmacology, Tokyo Women’s Medical University, Shinjuku, Tokyo 162-8666, Japan
- Correspondence: (T.S.); (J.M.); Tel.: +81-47-121-4060 (T.S.); +81-86-251-8003 (J.M.)
| |
Collapse
|
8
|
Stoyanova E, Mihaylova N, Ralchev N, Ganova P, Bradyanova S, Manoylov I, Raynova Y, Idakieva K, Tchorbanov A. Antitumor Properties of Epitope-Specific Engineered Vaccine in Murine Model of Melanoma. Mar Drugs 2022; 20:md20060392. [PMID: 35736195 PMCID: PMC9227764 DOI: 10.3390/md20060392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Finding new effective compounds of natural origin for composing anti-tumor vaccines is one of the main goals of antitumor research. Promising anti-cancer agents are the gastropodan hemocyanins-multimeric copper-containing glycoproteins used so far for therapy of different tumors. The properties of hemocyanins isolated from the marine snail Rapana thomasiana (RtH) and the terrestrial snail Helix aspersa (HaH) upon their use as carrier-proteins in conjugated vaccines, containing ganglioside mimotope GD3P4 peptide, were studied in the developed murine melanoma model. Murine melanoma cell line B16F10 was used for solid tumor establishment in C57BL/6 mice using various schemes of therapy. Protein engineering, flow cytometry, and cytotoxicity assays were also performed. The administration of the protein-engineered vaccines RtH-GD3P4 or HaH-GD3P4 under the three different regimens of therapy in the B16F10 murine melanoma model suppressed tumor growth, decreased tumor incidence, and prolonged the survival of treated animals. The immunization of experimental mice induced an infiltration of immunocompetent cells into the tumors and generated cytotoxic tumor-specific T cells in the spleen. The treatment also generates significantly higher levels of tumor-infiltrated M1 macrophages, compared to untreated tumor-bearing control mice. This study demonstrated a promising approach for cancer therapy having potential applications for cancer vaccine research.
Collapse
Affiliation(s)
- Emiliya Stoyanova
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
| | - Nikolina Mihaylova
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
| | - Nikola Ralchev
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
| | - Petya Ganova
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
| | - Silviya Bradyanova
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
| | - Iliyan Manoylov
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
| | - Yuliana Raynova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (Y.R.); (K.I.)
| | - Krassimira Idakieva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (Y.R.); (K.I.)
| | - Andrey Tchorbanov
- Laboratory of Experimental Immunology, Stefan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (E.S.); (N.M.); (N.R.); (P.G.); (S.B.); (I.M.)
- Correspondence: ; Tel.: + 359-2-979-6357; Fax: +359-2-870-0109
| |
Collapse
|
9
|
Preclinical Evaluation of CAR T Cell Function: In Vitro and In Vivo Models. Int J Mol Sci 2022; 23:ijms23063154. [PMID: 35328572 PMCID: PMC8955360 DOI: 10.3390/ijms23063154] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 01/12/2023] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR) T cells is a rapidly emerging modality that engineers T cells to redirect tumor-specific cytotoxicity. CAR T cells have been well characterized for their efficacy against B cell malignancies, and rigorously studied in other types of tumors. Preclinical evaluation of CAR T cell function, including direct tumor killing, cytokine production, and memory responses, is crucial to the development and optimization of CAR T cell therapies. Such comprehensive examinations are usually performed in different types of models. Model establishment should focus on key challenges in the clinical setting and the capability to generate reliable data to indicate CAR T cell therapeutic potency in the clinic. Further, modeling the interaction between CAR T cells and tumor microenvironment provides additional insight for the future endeavors to enhance efficacy, especially against solid tumors. This review will summarize both in vitro and in vivo models for CAR T cell functional evaluation, including how they have evolved with the needs of CAR T cell research, the information they can provide for preclinical assessment of CAR T cell products, and recent technology advances to test CAR T cells in more clinically relevant models.
Collapse
|
10
|
Candiota AP, Arús C. Establishing Imaging Biomarkers of Host Immune System Efficacy during Glioblastoma Therapy Response: Challenges, Obstacles and Future Perspectives. Metabolites 2022; 12:metabo12030243. [PMID: 35323686 PMCID: PMC8950145 DOI: 10.3390/metabo12030243] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
This hypothesis proposal addresses three major questions: (1) Why do we need imaging biomarkers for assessing the efficacy of immune system participation in glioblastoma therapy response? (2) Why are they not available yet? and (3) How can we produce them? We summarize the literature data supporting the claim that the immune system is behind the efficacy of most successful glioblastoma therapies but, unfortunately, there are no current short-term imaging biomarkers of its activity. We also discuss how using an immunocompetent murine model of glioblastoma, allowing the cure of mice and the generation of immune memory, provides a suitable framework for glioblastoma therapy response biomarker studies. Both magnetic resonance imaging and magnetic resonance-based metabolomic data (i.e., magnetic resonance spectroscopic imaging) can provide non-invasive assessments of such a system. A predictor based in nosological images, generated from magnetic resonance spectroscopic imaging analyses and their oscillatory patterns, should be translational to clinics. We also review hurdles that may explain why such an oscillatory biomarker was not reported in previous imaging glioblastoma work. Single shot explorations that neglect short-term oscillatory behavior derived from immune system attack on tumors may mislead actual response extent detection. Finally, we consider improvements required to properly predict immune system-mediated early response (1–2 weeks) to therapy. The sensible use of improved biomarkers may enable translatable evidence-based therapeutic protocols, with the possibility of extending preclinical results to human patients.
Collapse
Affiliation(s)
- Ana Paula Candiota
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, 08193 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Carles Arús
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Cerdanyola del Vallès, 08193 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Correspondence:
| |
Collapse
|
11
|
Huang Y, Nan L, Xiao C, Dong J, Li K, Cheng J, Ji Q, Wei Q, Bao G, Liu Y. Outer Membrane Vesicles Coating Nano-Glycyrrhizic Acid Confers Protection Against Borderella bronchiseptica Through Th1/Th2/Th17 Responses. Int J Nanomedicine 2022; 17:647-663. [PMID: 35177904 PMCID: PMC8846627 DOI: 10.2147/ijn.s350846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose Outer membrane vesicles (OMVs) are spherical nano-sized proteolipids secreted by numerous pathogenic Gram-negative bacteria. Due to the immunostimulatory properties and protective efficacy, OMVs have received increasing attention as a candidate for the vaccine to prevent and treat bacterial infections. However, the immune response remains elusive due to the low structural stability and poor size homogeneity of the vesicles. In this study, OMVs were used to coat self-assembled glycyrrhizic acid nanoparticles (GANs) and obtain a stable OMV vaccine. The immunoprotective effects and anti-infection efficacy were evaluated in vivo and in vitro. Methods The OMVs were prepared by ultrafiltration method and fused with GAN through mechanical extrusion. The characteristics, including morphology, hydrodynamic size, zeta potential, and stability were evaluated. The in vitro immunological function of GAN-OMV on the macrophages and in vivo immune efficacy and anti-infection effect were examined and compared. Results The results showed that the GAN-OMV were homogenous with a size of 130 nm and a stable core-shell structure. Micropinocytosis-dependent and clathrin-mediated endocytotic pathways effectively internalized the GAN-OMV into the macrophages and promoted cell proliferation, cytokine secretion, and M1 polarization. Furthermore, subcutaneous GAN-OMV vaccination contributed to significantly higher Borderella bronchiseptica (Bb)-specific antibody production and lymphocyte proliferation. The splenic lymphocytes of mice immunized with GAN-OMVs displayed a higher ratio of CD4+/CD8+ T cells and CD19+ B cells and produced significantly higher levels of Th1/Th2/Th17 cytokines. GAN-OMV also effectively prevented Bb reinfection. Conclusion In this study, GAN-OMV was developed successfully to stimulate Th1/Th2/Th17 immune responses against Bb and provide a promising strategy for novel vaccine development against the microbial pathogen.
Collapse
Affiliation(s)
- Yee Huang
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Li Nan
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Chenwen Xiao
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Jie Dong
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Ke Li
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Jvfen Cheng
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Quanan Ji
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Qiang Wei
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| | - Guolian Bao
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
- Correspondence: Guolian Bao; Yan Liu, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China, Email ;
| | - Yan Liu
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Science, Hangzhou, 310021, People’s Republic of China
| |
Collapse
|
12
|
Enhancing antibody-dependent cellular phagocytosis by Re-education of tumor-associated macrophages with resiquimod-encapsulated liposomes. Biomaterials 2020; 268:120601. [PMID: 33338932 DOI: 10.1016/j.biomaterials.2020.120601] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/13/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
Tumor-associated macrophages (TAMs) exist in nearly all tumors, and form a major part of the tumor microenvironment. TAMs are divided into two groups: tumor-suppressing M1 type and tumor-promoting M2 type. Most TAMs are educated by the tumor cells to become M2 type, which support tumor growth and make immunotherapy ineffective. Antibody-dependent cellular phagocytosis (ADCP) is an important mechanism for antibody cancer therapy, and this mechanism is dependent on TAMs. In this study, we found that the M1 type macrophages elicit a more efficient ADCP response than the M2 type, which was confirmed by three tumor cell lines, Raji, A431, and SKBR3, along with their corresponding therapeutic antibody Rituximab, anti-EGFR mouse monoclonal antibody (clone 528), and Trastuzumab, respectively. Resiquimod (R848), an immune system activating agent, has been shown to stimulate the M1 type macrophages, and re-educate the TAMs from M2 type to M1 type. By treating TAMs with R848, the ADCP response increased significantly in vitro and in in vivo mouse xenograft models. R848 encapsulated liposomes (R848-LPs) not only accumulated efficiently in the tumor tissues, but also distributed in the TAMs. Synergizing the R848-LPs with the anti-EGFR mouse monoclonal antibody (clone 528) significantly inhibited WiDr-tumor growth in vivo. Our study also revealed that the TAM-targeted delivery of R848 is able to re-educate the TAMs to M1 type, enhance the ADCP effect of the antibodies, and hence, enhance the anti-tumor effect of the therapeutic antibodies.
Collapse
|
13
|
Han JH, Yoon JS, Chang DY, Cho KG, Lim J, Kim SS, Suh-Kim H. CXCR4-STAT3 Axis Plays a Role in Tumor Cell Infiltration in an Orthotopic Mouse Glioblastoma Model. Mol Cells 2020; 43:539-550. [PMID: 32597394 PMCID: PMC7332361 DOI: 10.14348/molcells.2020.0098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 05/08/2020] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a fatal malignant tumor that is characterized by diffusive growth of tumor cells into the surrounding brain parenchyma. However, the diffusive nature of GBM and its relationship with the tumor microenvironment (TME) is still unknown. Here, we investigated the interactions of GBM with the surrounding microenvironment in orthotopic xenograft animal models using two human glioma cell lines, U87 and LN229. The GBM cells in our model showed different features on the aspects of cell growth rate during their development, dispersive nature of glioma tumor cells along blood vessels, and invasion into the brain parenchyma. Our results indicated that these differences in the two models are in part due to differences in the expression of CXCR4 and STAT3, both of which play an important role in tumor progression. In addition, the GBM shows considerable accumulation of resident microglia and peripheral macrophages, but polarizes differently into tumor-supporting cells. These results suggest that the intrinsic factors of GBM and their interaction with the TME determine the diffusive nature and probably the responsiveness to non-cancer cells in the TME.
Collapse
Affiliation(s)
- Ji-hun Han
- Department of Biomedical Sciences, Ajou Graduate School, Suwon 6499, Korea
- These authors contributed equally to this work.
| | - Jeong Seon Yoon
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea
- These authors contributed equally to this work.
| | - Da-Young Chang
- Department of Biomedical Sciences, Ajou Graduate School, Suwon 6499, Korea
| | - Kyung Gi Cho
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 1496, Korea
| | - Jaejoon Lim
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam 1496, Korea
| | - Sung-Soo Kim
- Department of Biomedical Sciences, Ajou Graduate School, Suwon 6499, Korea
| | - Haeyoung Suh-Kim
- Department of Biomedical Sciences, Ajou Graduate School, Suwon 6499, Korea
- Department of Anatomy, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
14
|
Eom YW, Akter R, Li W, Lee S, Hwang S, Kim J, Cho MY. M1 Macrophages Promote TRAIL Expression in Adipose Tissue-Derived Stem Cells, Which Suppresses Colitis-Associated Colon Cancer by Increasing Apoptosis of CD133 + Cancer Stem Cells and Decreasing M2 Macrophage Population. Int J Mol Sci 2020; 21:ijms21113887. [PMID: 32485960 PMCID: PMC7312348 DOI: 10.3390/ijms21113887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
We have previously reported that adipose tissue-derived stem cells (ASCs) cultured at high cell density can induce cancer cell death through the expression of type I interferons and tumor necrosis factor (TNF)-related apoptosis-inducing ligands (TRAIL). Here, we investigated whether TRAIL-expressing ASCs induced by M1 macrophages can alleviate colitis-associated cancer in an azoxymethane (AOM)/dextran sodium sulfate (DSS) animal model. M1 macrophages significantly increased the TRAIL expression in ASCs, which induced the apoptosis of LoVo cells in a TRAIL-dependent manner. However, CD133knockout LoVo cells, generated using the CRISPR-Cas9 gene-editing system, were resistant to TRAIL. In the AOM/DSS-induced colitis-associated cancer model, the intraperitoneal transplantation of TRAIL-expressing ASCs significantly suppressed colon cancer development. Moreover, immunohistochemical staining revealed a low CD133 expression in tumors from the AOM/DSS + ASCs group when compared with tumors from the untreated group. Additionally, the ASC treatment selectively reduced the number of M2 macrophages in tumoral (45.7 ± 4.2) and non-tumoral mucosa (30.3 ± 1.5) in AOM/DSS + ASCs-treated animals relative to those in the untreated group (tumor 71.7 ± 11.2, non-tumor 94.3 ± 12.5; p < 0.001). Thus, TRAIL-expressing ASCs are promising agents for anti-tumor therapy, particularly to alleviate colon cancer by inducing the apoptosis of CD133+ cancer stem cells and decreasing the M2 macrophage population.
Collapse
Affiliation(s)
- Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea; (Y.W.E.); (S.H.)
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea
| | - Rokeya Akter
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea; (R.A.); (W.L.); (S.L.)
| | - Wanlu Li
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea; (R.A.); (W.L.); (S.L.)
| | - Suji Lee
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea; (R.A.); (W.L.); (S.L.)
| | - Soonjae Hwang
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea; (Y.W.E.); (S.H.)
| | - Jiye Kim
- Department of Plastic and Reconstructive Surgery, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea;
| | - Mee-Yon Cho
- Regeneration Medicine Research Center, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-do 26426, Korea; (R.A.); (W.L.); (S.L.)
- Correspondence: ; Tel.: +82-33-731-1553
| |
Collapse
|
15
|
Intensive therapy with gastropodan hemocyanins increases their antitumor properties in murine model of colon carcinoma. Int Immunopharmacol 2020; 84:106566. [PMID: 32416451 DOI: 10.1016/j.intimp.2020.106566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/22/2022]
Abstract
Various natural compounds have been tested as anticancer therapeutics in clinical trials. Most promising direction for antitumor therapy is the use of substances which enhance the immune system response stimulating tumor-specific lymphocytes. Hemocyanins are large extracellular oxygen transport glycoproteins isolated from different arthropod and mollusk species which exhibit strong anticancer properties. Immunized in mammals they trigger Th1 immune response that promotes unspecific stimulation and adjuvant activity in experimental therapeutic vaccines for cancer and antibody development. In the present study we used two hemocyanins - one isolated from marine snail Rapana thomasiana (RtH) and another one, from the terrestrial snail Helix pomatia (HpH) which have been investigated by using different administration schedules (intensive and mild) in murine model of colon carcinoma. The treatment with RtH and HpH generated high levels of antitumor IgG antibodies, antibody-producing plasma cells and tumor-specific CTLs, stimulated secretion of proinflammatory cytokines, suppressed the manifestation of carcinoma symptoms as tumor growth and size, and prolonged the life span of treated mice. Our results showed a significant anti-cancer effect of RtH and HpH hemocyanins on a murine model of colon carcinoma with promising potential for immunotherapy in various schemes of administration based on cross-reactive tumor-associated epitopes.
Collapse
|
16
|
Smink AM, Najdahmadi A, Alexander M, Li S, Rodriquez S, van Goor H, Hillebrands JL, Botvinick E, Lakey JRT, de Vos P. The Effect of a Fast-Releasing Hydrogen Sulfide Donor on Vascularization of Subcutaneous Scaffolds in Immunocompetent and Immunocompromised Mice. Biomolecules 2020; 10:biom10050722. [PMID: 32384680 PMCID: PMC7277536 DOI: 10.3390/biom10050722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Islet transplantation into subcutaneous polymer scaffolds has shown to successfully induce normoglycemia in type 1 diabetes models. Vascularization of these scaffolds is imperative for optimal control of glucose levels. We studied the effect of the vascular stimulator hydrogen sulfide (H2S) on the degree of vascularization of a scaffold and the role of the immune system in this process. Scaffolds were subcutaneously implanted in immunocompetent C57BL/6 and immunocompromised nude mice. Mice received twice-daily intraperitoneal injections of the fast-releasing H2S donor sodium hydrosulfide (NaHS, 25 or 50 μmol/kg) or saline for 28 days. After 63 days the vascular network was analyzed by histology and gene expression. Here we showed that the vascularization of a subcutaneous scaffold in nude mice was significantly impaired by H2S treatment. Both the CD31 gene and protein expression were reduced in these scaffolds compared to the saline-treated controls. In C57BL/6 mice, the opposite was found, the vascularization of the scaffold was significantly increased by H2S. The mRNA expression of the angiogenesis marker CD105 was significantly increased compared to the controls as well as the number of CD31 positive blood vessels. In conclusion, the immune system plays an important role in the H2S mediated effect on vascularization of subcutaneous scaffolds.
Collapse
Affiliation(s)
- Alexandra M. Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (H.v.G.); (J.-L.H.); (P.d.V.)
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA; (M.A.); (S.L.); (S.R.); (E.B.); (J.R.T.L.)
- Correspondence: ; Tel.: +31-50-3610109
| | - Avid Najdahmadi
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, CA 92617, USA;
| | - Michael Alexander
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA; (M.A.); (S.L.); (S.R.); (E.B.); (J.R.T.L.)
| | - Shiri Li
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA; (M.A.); (S.L.); (S.R.); (E.B.); (J.R.T.L.)
| | - Samuel Rodriquez
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA; (M.A.); (S.L.); (S.R.); (E.B.); (J.R.T.L.)
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (H.v.G.); (J.-L.H.); (P.d.V.)
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (H.v.G.); (J.-L.H.); (P.d.V.)
| | - Elliot Botvinick
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA; (M.A.); (S.L.); (S.R.); (E.B.); (J.R.T.L.)
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, CA 92617, USA;
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - Jonathan R. T. Lakey
- Department of Surgery, University of California Irvine, Orange, CA 92868, USA; (M.A.); (S.L.); (S.R.); (E.B.); (J.R.T.L.)
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (H.v.G.); (J.-L.H.); (P.d.V.)
| |
Collapse
|
17
|
Alhasan R, Nasim MJ, Jacob C, Gaucher C. Selenoneine: a Unique Reactive Selenium Species From the Blood of Tuna With Implications for Human Diseases. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s40495-019-00175-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
18
|
Dietary Supplementation of Selenoneine-Containing Tuna Dark Muscle Extract Effectively Reduces Pathology of Experimental Colorectal Cancers in Mice. Nutrients 2018; 10:nu10101380. [PMID: 30262787 PMCID: PMC6212930 DOI: 10.3390/nu10101380] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
Selenoneine is an ergothioneine analog with greater antioxidant activity and is the major form of organic selenium in the blood, muscles, and other tissues of tuna. The aim of this study was to determine whether a selenoneine-rich diet exerts antioxidant activities that can prevent carcinogenesis in two types of colorectal cancer model in mice. We administrated selenoneine-containing tuna dark muscle extract (STDME) to mice for one week and used azoxymethane (AOM) and dextran sodium sulfate (DSS) for inducing colorectal carcinogenesis. Next, we examined the incidence of macroscopic polyps and performed functional analysis of immune cells from the spleen. In the AOM/DSS-induced colitis-associated cancer (CAC) model, the oral administration of STDME significantly decreased tumor incidence and inhibited the accumulation of myeloid-derived suppressor cells (MDSCs) while also inhibiting the downregulation of interferon-γ (IFN-γ) production during carcinogenesis. These results suggest that dietary STDME may be an effective agent for reducing colorectal tumor progression.
Collapse
|