1
|
Shen Q, Cao M, Yu C, Tang J, Song L, Ding Y, Ju L, Wei JF, Li L, Huang W. Biodegradable Mesoporous Organosilica-Based Nanostabilizer Targeting Mast Cells for Long-Term Treatment of Allergic Diseases. ACS NANO 2024; 18:16934-16946. [PMID: 38907988 DOI: 10.1021/acsnano.4c03069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Allergic diseases are immune system dysfunctions mediated by mast cell (MC) activation stimulated by specific allergens. However, current small molecular MC stabilizers for allergic disease prevention often require multiple doses over a long period of time and are associated with serious side effects. Herein, we develop a diselenide-bridged mesoporous silica nanostabilizer, proving that it could specifically target sensitized MCs via the recognition of IgE aptamer and IgE. Meantime, the IgE aptamer can also mitigate allergic reactions by preventing re-exposure of allergens from the surface of sensitized MCs. Furthermore, the diselenide-bridged scaffold can be reduced by the intracellular excessive ROS, subsequently achieving redox homeostasis via ROS depletion. Finally, the precise release of small molecular MC stabilizers along with the biodegradation of nanocarrier can stabilize the membranes of MCs. In vivo assays in passive cutaneous anaphylactic (PCA) and allergic rhinitis (AR) mice indicated that our current strategy further endowed it with a high efficacy, long-term therapeutic time window, as well as negligible inflammatory side effects for allergic diseases, offering a promising therapeutic strategy for the clinical generalization of allergic diseases.
Collapse
Affiliation(s)
- Qian Shen
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Mengda Cao
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210044, China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Jian Tang
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Lebin Song
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Yanan Ding
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Linjie Ju
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Ji-Fu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
- The Institute of Flexible Electronics, Xiamen University, Xiamen 361005, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE), School of Flexible Electronics (Future Technologies) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
- The Institute of Flexible Electronics, Xiamen University, Xiamen 361005, China
| |
Collapse
|
2
|
Praetzel R, Kepley C. Human Lung Mast Cells as a Possible Reservoir for Coronavirus: A Novel Unrecognized Mechanism for SARS-CoV-2 Immune-Mediated Pathology. Int J Mol Sci 2024; 25:6511. [PMID: 38928216 PMCID: PMC11204339 DOI: 10.3390/ijms25126511] [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: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
The pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global health concern. Cell entry of SARS-CoV-2 depends on viral spike (S) proteins binding to cellular receptors (ACE2) and their subsequent priming by host cell proteases (TMPRSS2). Assessing effects of viral-induced host response factors and determining which cells are used by SARS-CoV-2 for entry might provide insights into viral transmission, add clarity to the virus' pathogenesis, and possibly reveal therapeutic targets. Mast cells (MCs) are ubiquitously expressed tissue cells that act as immune sentinels given their ability to react specifically to pathogens at environmental interfaces, such as in the lung. Several lines of evidence suggest a critical role for MCs in SARS-CoV-2 infections based on patients' mediator profiles, especially the "cytokine storm" responsible for most morbidity and mortality. In this pilot study, we demonstrated that human lung MCs (n = 3 donors) are a source of renin and that they upregulate the membrane receptor for SARS-CoV-2 (ACE2) as well as the protease required for cellular entry (TMPRSS2) under certain conditions. We hypothesized that infection of human MCs with SARS-CoV-2 may be a heretofore-unrecognized mechanism of viral pathogenesis, and further studies are required to assess this question.
Collapse
Affiliation(s)
| | - Chris Kepley
- Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA
| |
Collapse
|
3
|
Praetzel R, Motaghed M, Fereydouni M, Ahani E, Kepley C. Description and Characterization of Three-Dimensional Human Mast Cell Progenitor Spheroids In Vitro. Cureus 2024; 16:e53708. [PMID: 38455803 PMCID: PMC10919245 DOI: 10.7759/cureus.53708] [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: 12/12/2023] [Accepted: 01/28/2024] [Indexed: 03/09/2024] Open
Abstract
Human mast cells (MC) are an essential component of the immune system as they uniquely store and release a wide range of soluble mediators through IgE and non-IgE mechanisms. Several tissue sources can be used to differentiate functional MC for in vitro and in vivo studies. Here we describe an improved method for obtaining large numbers of human MC from adipose tissue with advantages over current methods. We analyzed donor parameters (e.g. age, race) on MC-isolation following adipose and skin tissue digestion from healthy donors. Adipose and skin-derived MC were morphologically and immunophenotypically similar in all donors regardless of age. However, donor-dependent variations in MC numbers were observed following tissue digestion. In addition, we identified and characterized three-dimensional structures from which mature MC emerged in vitro using peripheral blood and human tissue sources. MC progenitor spheroids (MCPS) appeared approximately one week following progenitor isolation and were consistently observed to have mature MC attached, emerging, or nearby when cultured in a stem cell factor-containing medium. The overall characteristics of the MCPS were similar from each tissue source. We propose that these MCPS serve as the common source of human MC in vitro.
Collapse
Affiliation(s)
- Rebecca Praetzel
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Mona Motaghed
- Department of Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, USA
| | - Mohammad Fereydouni
- Department of Nanoscience, University of North Carolina at Greensboro, Greensboro, USA
| | - Elnaz Ahani
- Department of Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, USA
| | - Chris Kepley
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| |
Collapse
|
4
|
Fereydouni M, Ahani E, Desai P, Motaghed M, Dellinger A, Metcalfe DD, Yin Y, Lee SH, Kafri T, Bhatt AP, Dellinger K, Kepley CL. Human Tumor Targeted Cytotoxic Mast Cells for Cancer Immunotherapy. Front Oncol 2022; 12:871390. [PMID: 35574362 PMCID: PMC9097604 DOI: 10.3389/fonc.2022.871390] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/23/2022] [Indexed: 12/14/2022] Open
Abstract
The diversity of autologous cells being used and investigated for cancer therapy continues to increase. Mast cells (MCs) are tissue cells that contain a unique set of anti-cancer mediators and are found in and around tumors. We sought to exploit the anti-tumor mediators in MC granules to selectively target them to tumor cells using tumor specific immunoglobin E (IgE) and controllably trigger release of anti-tumor mediators upon tumor cell engagement. We used a human HER2/neu-specific IgE to arm human MCs through the high affinity IgE receptor (FcεRI). The ability of MCs to bind to and induce apoptosis of HER2/neu-positive cancer cells in vitro and in vivo was assessed. The interactions between MCs and cancer cells were investigated in real time using confocal microscopy. The mechanism of action using cytotoxic MCs was examined using gene array profiling. Genetically manipulating autologous MC to assess the effects of MC-specific mediators have on apoptosis of tumor cells was developed using siRNA. We found that HER2/neu tumor-specific IgE-sensitized MCs bound, penetrated, and killed HER2/neu-positive tumor masses in vitro. Tunneling nanotubes formed between MCs and tumor cells are described that parallel tumor cell apoptosis. In solid tumor, human breast cancer (BC) xenograft mouse models, infusion of HER2/neu IgE-sensitized human MCs co-localized to BC cells, decreased tumor burden, and prolonged overall survival without indications of toxicity. Gene microarray of tumor cells suggests a dependence on TNF and TGFβ signaling pathways leading to apoptosis. Knocking down MC-released tryptase did not affect apoptosis of cancer cells. These studies suggest MCs can be polarized from Type I hypersensitivity-mediating cells to cytotoxic cells that selectively target tumor cells and specifically triggered to release anti-tumor mediators. A strategy to investigate which MC mediators are responsible for the observed tumor killing is described so that rational decisions can be made in the future when selecting which mediators to target for deletion or those that could further polarize them to cytotoxic MC by adding other known anti-tumor agents. Using autologous human MC may provide further options for cancer therapeutics that offers a unique anti-cancer mechanism of action using tumor targeted IgE’s.
Collapse
Affiliation(s)
- Mohammad Fereydouni
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Elnaz Ahani
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural and Technical (AT) State University, Greensboro, NC, United States
| | - Parth Desai
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Mona Motaghed
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural and Technical (AT) State University, Greensboro, NC, United States
| | - Anthony Dellinger
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Dean D. Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yuzhi Yin
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Sung Hyun Lee
- Gene Therapy Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tal Kafri
- Gene Therapy Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Aadra P. Bhatt
- Lineberger Comprehensive Cancer Center, and the Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural and Technical (AT) State University, Greensboro, NC, United States
| | - Christopher L. Kepley
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, VA, United States
- *Correspondence: Christopher L. Kepley,
| |
Collapse
|
5
|
Fereydouni M, Motaghed M, Ahani E, Kafri T, Dellinger K, Metcalfe DD, Kepley CL. Harnessing the Anti-Tumor Mediators in Mast Cells as a New Strategy for Adoptive Cell Transfer for Cancer. Front Oncol 2022; 12:830199. [PMID: 35433433 PMCID: PMC9009255 DOI: 10.3389/fonc.2022.830199] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
The emergence of cancer immunotherapies utilizing adoptive cell transfer (ACT) continues to be one of the most promising strategies for cancer treatment. Mast cells (MCs) which occur throughout vascularized tissues, are most commonly associated with Type I hypersensitivity, bind immunoglobin E (IgE) with high affinity, produce anti-cancer mediators such as tumor necrosis factor alpha (TNF-α) and granulocyte macrophage colony-stimulating factor (GM-CSF), and generally populate the tumor microenvironments. Yet, the role of MCs in cancer pathologies remains controversial with evidence for both anti-tumor and pro-tumor effects. Here, we review the studies examining the role of MCs in multiple forms of cancer, provide an alternative, MC-based hypothesis underlying the mechanism of therapeutic tumor IgE efficacy in clinical trials, and propose a novel strategy for using tumor-targeted, IgE-sensitized MCs as a platform for developing new cellular cancer immunotherapies. This autologous MC cancer immunotherapy could have several advantages over current cell-based cancer immunotherapies and provide new mechanistic strategies for cancer therapeutics alone or in combination with current approaches.
Collapse
Affiliation(s)
- Mohammad Fereydouni
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina Greensboro (UNCG), Greensboro, NC, United States
| | - Mona Motaghed
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| | - Elnaz Ahani
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| | - Tal Kafri
- Gene Therapy Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, United States
| | - Dean D. Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Christopher L. Kepley
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, VA, United States
- *Correspondence: Christopher L. Kepley,
| |
Collapse
|
6
|
Hafezi B, Chan L, Knapp JP, Karimi N, Alizadeh K, Mehrani Y, Bridle BW, Karimi K. Cytokine Storm Syndrome in SARS-CoV-2 Infections: A Functional Role of Mast Cells. Cells 2021; 10:1761. [PMID: 34359931 PMCID: PMC8308097 DOI: 10.3390/cells10071761] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/27/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Cytokine storm syndrome is a cascade of escalated immune responses disposing the immune system to exhaustion, which might ultimately result in organ failure and fatal respiratory distress. Infection with severe acute respiratory syndrome-coronavirus-2 can result in uncontrolled production of cytokines and eventually the development of cytokine storm syndrome. Mast cells may react to viruses in collaboration with other cells and lung autopsy findings from patients that died from the coronavirus disease that emerged in 2019 (COVID-19) showed accumulation of mast cells in the lungs that was thought to be the cause of pulmonary edema, inflammation, and thrombosis. In this review, we present evidence that a cytokine response by mast cells may initiate inappropriate antiviral immune responses and cause the development of cytokine storm syndrome. We also explore the potential of mast cell activators as adjuvants for COVID-19 vaccines and discuss the medications that target the functions of mast cells and could be of value in the treatment of COVID-19. Recognition of the cytokine storm is crucial for proper treatment of patients and preventing the release of mast cell mediators, as impeding the impacts imposed by these mediators could reduce the severity of COVID-19.
Collapse
Affiliation(s)
- Bahareh Hafezi
- Department of Clinical Science, School of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad 9177948974, Iran; (B.H.); (N.K.)
| | - Lily Chan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Jason P. Knapp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Negar Karimi
- Department of Clinical Science, School of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad 9177948974, Iran; (B.H.); (N.K.)
| | - Kimia Alizadeh
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA;
| | - Yeganeh Mehrani
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.P.K.); (Y.M.)
| |
Collapse
|
7
|
Lin P, Cao M, Xia F, Liao H, Sun H, Wang Q, Lee J, Zhou Y, Guan Y, Zhang C, Xu Z, Li F, Wei J, Ling D. A Phosphatase-Mimetic Nano-Stabilizer of Mast Cells for Long-Term Prevention of Allergic Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004115. [PMID: 33898190 PMCID: PMC8061383 DOI: 10.1002/advs.202004115] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/30/2020] [Indexed: 06/12/2023]
Abstract
Allergic diseases are pathological immune responses with significant morbidity, which are closely associated with allergic mediators as released by allergen-stimulated mast cells (MCs). Prophylactic stabilization of MCs is regarded as a practical approach to prevent allergic diseases. However, most of the existing small molecular MC stabilizers exhibit a narrow therapeutic time window, failing to provide long-term prevention of allergic diseases. Herein, ceria nanoparticle (CeNP-) based phosphatase-mimetic nano-stabilizers (PMNSs) with a long-term therapeutic time window are developed for allergic disease prevention. By virtue of the regenerable catalytic hotspots of oxygen vacancies on the surface of CeNPs, PMNSs exhibit sustainable phosphatase-mimetic activity to dephosphorylate phosphoproteins in allergen-stimulated MCs. Consequently, PMNSs constantly modulate intracellular phospho-signaling cascades of MCs to inhibit the degranulation of allergic mediators, which prevents the initiation of allergic mediator-associated pathological responses, eventually providing protection against allergic diseases with a long-term therapeutic time window.
Collapse
Affiliation(s)
- Peihua Lin
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Mengda Cao
- Research Division of Clinical PharmacologyThe First Affiliated HospitalNanjing Medical UniversityNanjingJiangsu210029P. R. China
| | - Fan Xia
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
- Hangzhou Institute of Innovative MedicineZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Hongwei Liao
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Heng Sun
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Qiyue Wang
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jiyoung Lee
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Yan Zhou
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Yunan Guan
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Cheng Zhang
- Women & Children Central LaboratoryThe First Affiliated HospitalNanjing Medical UniversityNanjingJiangsu210036P. R. China
| | - Zhiqiang Xu
- Research Division of Clinical PharmacologyThe First Affiliated HospitalNanjing Medical UniversityNanjingJiangsu210029P. R. China
| | - Fangyuan Li
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
- Hangzhou Institute of Innovative MedicineZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Ji‐Fu Wei
- Research Division of Clinical PharmacologyThe First Affiliated HospitalNanjing Medical UniversityNanjingJiangsu210029P. R. China
| | - Daishun Ling
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
- Hangzhou Institute of Innovative MedicineZhejiang UniversityHangzhouZhejiang310058P. R. China
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240P. R. China
| |
Collapse
|
8
|
Duguay BA, Lu L, Arizmendi N, Unsworth LD, Kulka M. The Possible Uses and Challenges of Nanomaterials in Mast Cell Research. THE JOURNAL OF IMMUNOLOGY 2020; 204:2021-2032. [PMID: 32253270 DOI: 10.4049/jimmunol.1800658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 12/19/2019] [Indexed: 11/19/2022]
Abstract
Mast cells are tissue-resident immune cells that are involved in inflammation and fibrosis but also serve beneficial roles, including tissue maintenance, angiogenesis, pathogen clearance, and immunoregulation. Their multifaceted response and the ability of their mediators to target multiple organs and tissues means that mast cells play important roles in numerous conditions, including asthma, atopic dermatitis, drug sensitivities, ischemic heart disease, Alzheimer disease, arthritis, irritable bowel syndrome, infections (parasites, bacteria and viruses), and cancer. As a result, mast cells have become an important target for drug discovery and diagnostic research. Recent work has focused on applying novel nanotechnologies to explore cell biology. In this brief review, we will highlight the use of nanomaterials to modify mast cell functions and will discuss the potential of these technologies as research tools for understanding mast cell biology.
Collapse
Affiliation(s)
- Brett A Duguay
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Lei Lu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Narcy Arizmendi
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Larry D Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada; and
| | - Marianna Kulka
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta T6G 2M9, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
9
|
Alsaleh NB, Brown JM. Engineered Nanomaterials and Type I Allergic Hypersensitivity Reactions. Front Immunol 2020; 11:222. [PMID: 32117324 PMCID: PMC7033602 DOI: 10.3389/fimmu.2020.00222] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Type I allergic hypersensitivity disorders (atopy) including asthma, atopic dermatitis, allergic rhinitis, and food allergy are on the rise in developed and developing countries. Engineered nanomaterials (ENMs) span a large spectrum of material compositions including carbonic, metals, polymers, lipid-based, proteins, and peptides and are being utilized in a wide range of industries including healthcare and pharmaceuticals, electronics, construction, and food industry, and yet, regulations for the use of ENMs in consumer products are largely lacking. Prior evidence has demonstrated the potential of ENMs to induce and/or aggravate type I allergic hypersensitivity responses. Furthermore, previous studies have shown that ENMs could directly interact with and activate key T-helper 2 (Th2) effector cell types (such as mast cells) and the complement system, which could result in pseudoallergic (non-IgE-mediated) hypersensitivity reactions. Nevertheless, the underlying molecular mechanisms of ENM-mediated induction and/or exacerbation of type I immune responses are poorly understood. In this review, we first highlight key examples of studies that have demonstrated inherent immunomodulatory properties of ENMs in the context of type I allergic hypersensitivity reactions, and most importantly, we attempt to put together the potential molecular mechanisms that could drive ENM-mediated stimulation and/or aggravation of type I allergic hypersensitivity responses.
Collapse
Affiliation(s)
- Nasser B Alsaleh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Jared M Brown
- Department of Pharmaceutical Sciences, Colorado Center for Nanomedicine and Nanosafety, Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
10
|
Plotkin JD, Elias MG, Fereydouni M, Daniels-Wells TR, Dellinger AL, Penichet ML, Kepley CL. Human Mast Cells From Adipose Tissue Target and Induce Apoptosis of Breast Cancer Cells. Front Immunol 2019; 10:138. [PMID: 30833944 PMCID: PMC6387946 DOI: 10.3389/fimmu.2019.00138] [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: 10/26/2018] [Accepted: 01/16/2019] [Indexed: 01/09/2023] Open
Abstract
Mast cells (MC) are important immune sentinels found in most tissue and widely recognized for their role as mediators of Type I hypersensitivity. However, they also secrete anti-cancer mediators such as tumor necrosis factor alpha (TNF-α) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The purpose of this study was to investigate adipose tissue as a new source of MC in quantities that could be used to study MC biology focusing on their ability to bind to and kill breast cancer cells. We tested several cell culture media previously demonstrated to induce MC differentiation. We report here the generation of functional human MC from adipose tissue. The adipose-derived mast cells (ADMC) are phenotypically and functionally similar to connective tissue expressing tryptase, chymase, c-kit, and FcεRI and capable of degranulating after cross-linking of FcεRI. The ADMC, sensitized with anti-HER2/neu IgE antibodies with human constant regions (trastuzumab IgE and/or C6MH3-B1 IgE), bound to and released MC mediators when incubated with HER2/neu-positive human breast cancer cells (SK-BR-3 and BT-474). Importantly, the HER2/neu IgE-sensitized ADMC induced breast cancer cell (SK-BR-3) death through apoptosis. Breast cancer cell apoptosis was observed after the addition of cell-free supernatants containing mediators released from FcεRI-challenged ADMC. Apoptosis was significantly reduced when TNF-α blocking antibodies were added to the media. Adipose tissue represents a source MC that could be used for multiple research purposes and potentially as a cell-mediated cancer immunotherapy through the expansion of autologous (or allogeneic) MC that can be targeted to tumors through IgE antibodies recognizing tumor specific antigens.
Collapse
Affiliation(s)
- Jesse D Plotkin
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Michael G Elias
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Mohammad Fereydouni
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Tracy R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anthony L Dellinger
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.,The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States.,AIDS Institute, University of California, Los Angeles, Los Angeles, CA, United States.,The California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher L Kepley
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| |
Collapse
|
11
|
Plotkin JD, Elias MG, Dellinger AL, Kepley CL. NF-κB inhibitors that prevent foam cell formation and atherosclerotic plaque accumulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2037-2048. [PMID: 28457935 DOI: 10.1016/j.nano.2017.04.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/30/2017] [Accepted: 04/16/2017] [Indexed: 11/30/2022]
Abstract
The transformation of monocyte-derived macrophages into lipid-laden foam cells is one inflammatory process underlying atherosclerotic disease. Previous studies have demonstrated that fullerene derivatives (FDs) have inflammation-blunting properties. Thus, it was hypothesized that FD could inhibit the transformation process underlying foam cell formation. Fullerene derivatives inhibited the phorbol myristic acid/oxidized low-density lipoprotein-induced differentiation of macrophages into foam cells as determined by lipid staining and morphology.Lipoprotein-induced generation of TNF-α, C5a-induced MC activation, ICAM-1 driven adhesion, and CD36 expression were significantly inhibited in FD treated cells compared to non-treated cells. Inhibition appeared to be mediated through the NF-κB pathway as FD reduced expression of NF-κB and atherosclerosis-associated genes. Compared to controls, FD dramatically inhibited plaque formation in arteries of apolipoprotein E null mice. Thus, FD may be an unrecognized therapy to prevent atherosclerotic lesions via inhibition of foam cell formation and MC stabilization.
Collapse
Affiliation(s)
- Jesse D Plotkin
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States
| | - Michael G Elias
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States
| | - Anthony L Dellinger
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States
| | - Christopher L Kepley
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, United States.
| |
Collapse
|
12
|
Lebre F, Hearnden CH, Lavelle EC. Modulation of Immune Responses by Particulate Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5525-5541. [PMID: 27167228 DOI: 10.1002/adma.201505395] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/12/2016] [Indexed: 06/05/2023]
Abstract
Many biomaterials that are in both preclinical and clinical use are particulate in nature and there is a growing appreciation that the physicochemical properties of materials have a significant impact on their efficacy. The ability of particulates to modulate adaptive immune responses has been recognized for the past century but it is only in recent decades that a mechanistic understanding of how particulates can regulate these responses has emerged. It is now clear that particulate characteristics including size, charge, shape and porosity can influence the scale and nature of both the innate and adaptive immune responses. The potential to tailor biomaterials in order to regulate the type of innate immune response induced, offers significant opportunities in terms of designing systems with increased immune-mediated efficacy.
Collapse
Affiliation(s)
- Filipa Lebre
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, D02 PN40, Ireland
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Claire H Hearnden
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, D02 PN40, Ireland
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| |
Collapse
|
13
|
Shershakova N, Baraboshkina E, Andreev S, Purgina D, Struchkova I, Kamyshnikov O, Nikonova A, Khaitov M. Anti-inflammatory effect of fullerene C60 in a mice model of atopic dermatitis. J Nanobiotechnology 2016; 14:8. [PMID: 26810232 PMCID: PMC4727272 DOI: 10.1186/s12951-016-0159-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/13/2016] [Indexed: 12/15/2022] Open
Abstract
Background Water-soluble form of fullerene C60 is a promising tool for the control of ROS-dependent inflammation including allergic diseases. Anti-inflammatory effects of C60 (nC60) aqueous dispersion were evaluated in the mouse models of atopic dermatitis using subcutaneous (SC) and epicutaneous (EC) applications during 50 days period. A highly stable nC60 was prepared by exhaustive dialysis of water-organic C60 solution against water, where the size and ζ-potential of fullerene nanoparticles are about 100 nm and −30 mV, respectively. Results To induce skin inflammation, female BALB/c mice were EC sensitized with ovalbumin three times during one-weekly exposures. The nC60 solution was administrated in mice subcutaneously (SC) (0.1 mg/kg) and epicutaneously (EC) (1 mg/kg). Significant suppression of IgE and Th2 cytokines production and a concomitant rise in concentrations of Th1 cytokines were observed in nC60-treated groups. In addition, a significant increase in the levels of Foxp3+ and filaggrin mRNA expression was observed at EC application. Histological examination of skin samples indicated that therapeutic effect was achieved by both EC and SC treatment, but it was more effective with EC. Pronounced reduction of the eosinophil and leukocyte infiltration in treated skin samples was observed. Conclusions We suppose that nC60 treatment shifts immune response from Th2 to Th1 and restores to some extent the function of the skin barrier. This approach can be a good alternative to the treatment of allergic and other inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Sergey Andreev
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia.
| | - Daria Purgina
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia.
| | | | | | | | - Musa Khaitov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia.
| |
Collapse
|
14
|
Dellinger AL, Cunin P, Lee D, Kung AL, Brooks DB, Zhou Z, Nigrovic PA, Kepley CL. Inhibition of inflammatory arthritis using fullerene nanomaterials. PLoS One 2015; 10:e0126290. [PMID: 25879437 PMCID: PMC4400016 DOI: 10.1371/journal.pone.0126290] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 03/31/2015] [Indexed: 12/27/2022] Open
Abstract
Inflammatory arthritis (e.g. rheumatoid arthritis; RA) is a complex disease driven by the interplay of multiple cellular lineages. Fullerene derivatives have previously been shown to have anti-inflammatory capabilities mediated, in part, by their ability to prevent inflammatory mediator release by mast cells (MC). Recognizing that MC can serve as a cellular link between autoantibodies, soluble mediators, and other effector populations in inflammatory arthritis, it was hypothesized that fullerene derivatives might be used to target this inflammatory disease. A panel of fullerene derivatives was tested for their ability to affect the function of human skin-derived MC as well as other lineages implicated in arthritis, synovial fibroblasts and osteoclasts. It is shown that certain fullerene derivatives blocked FcγR- and TNF-α-induced mediator release from MC; TNF-α-induced mediator release from RA synovial fibroblasts; and maturation of human osteoclasts. MC inhibition by fullerene derivatives was mediated through the reduction of mitochondrial membrane potential and FcγR-mediated increases in cellular reactive oxygen species and NF-κB activation. Based on these in vitro data, two fullerene derivatives (ALM and TGA) were selected for in vivo studies using K/BxN serum transfer arthritis in C57BL/6 mice and collagen-induced arthritis (CIA) in DBA/1 mice. Dye-conjugated fullerenes confirmed localization to affected joints in arthritic animals but not in healthy controls. In the K/BxN moldel, fullerenes attenuated arthritis, an effect accompanied by reduced histologic inflammation, cartilage/bone erosion, and serum levels of TNF-α. Fullerenes remained capable of attenuating K/BxN arthritis in mast cell-deficient mice Cre-Master mice, suggesting that lineages beyond the MC represent relevant targets in this system. These studies suggest that fullerene derivatives may hold promise both as an assessment tool and as anti-inflammatory therapy of arthritis.
Collapse
Affiliation(s)
- Anthony L. Dellinger
- University of North Carolina Greensboro, Joint School of Nanosceince and Nanoengineering, Greensboro, North Carolina, United States of America
| | - Pierre Cunin
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, and Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David Lee
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Andrew L. Kung
- Dana Farber Institute, Boston, Massachusetts, United States of America
| | - D. Bradford Brooks
- Luna Innovations Incorporated, Danville, Virginia, United States of America
| | - Zhiguo Zhou
- Luna Innovations Incorporated, Danville, Virginia, United States of America
| | - Peter A. Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, and Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christopher L. Kepley
- University of North Carolina Greensboro, Joint School of Nanosceince and Nanoengineering, Greensboro, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Although the production and use of engineered nanomaterials (ENMs) is rapidly increasing, we lack sufficient knowledge regarding their capacity to induce and/or promote allergic disease. As novel ENMs are being developed and used for biomedical applications, such as drug delivery, it will be critical to understand the relationship between physicochemical properties of ENMs and possible mechanisms of immunomodulation. RECENT FINDINGS Cellular studies and a few animal studies have begun to examine the immunomodulatory effects of ENM exposure that may be predictive of developing allergic reactions. Specifically, the effects of direct ENM exposure on key immune cells recognized to facilitate allergic disease has been evaluated and will be discussed. However, few studies have reported specific physicochemical properties of ENMs that initiate allergic immune responses. Although limited, these descriptive studies point to the induction of cellular mechanisms that are well known to promote allergic disease. SUMMARY The limited data currently available suggest that there is a potential risk for the development of allergic responses following exposure to ENMs. As more ENMs are developed for consumer products and nanomedicines, further study on their potential for adverse immune interactions will be necessary for safe implementation of these novel materials.
Collapse
|
16
|
Finn DF, Walsh JJ. Twenty-first century mast cell stabilizers. Br J Pharmacol 2014; 170:23-37. [PMID: 23441583 DOI: 10.1111/bph.12138] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/30/2013] [Accepted: 02/13/2013] [Indexed: 12/14/2022] Open
Abstract
Mast cell stabilizing drugs inhibit the release of allergic mediators from mast cells and are used clinically to prevent allergic reactions to common allergens. Despite the relative success of the most commonly prescribed mast cell stabilizer, disodium cromoglycate, in use for the preventative treatment of bronchial asthma, allergic conjunctivitis and vernal keratoconjunctivitis, there still remains an urgent need to design new substances that are less expensive and require less frequent dosing schedules. In this regard, recent developments towards the discovery of the next generation of mast cell stabilizing drugs has included studies on substances isolated from natural sources, biological, newly synthesized compounds and drugs licensed for other indications. The diversity of natural products evaluated range from simple phenols, alkaloids, terpenes to simple amino acids. While in some cases their precise mode of action remains unknown it has nevertheless sparked interest in the development of synthetic derivatives with improved pharmacological properties. Within the purely synthetic class of inhibitors, particular attention has been devoted to the inhibition of important signalling molecules including spleen TK and JAK3. The statin class of cholesterol-lowering drugs as well as nilotinib, a TK inhibitor, are just some examples of clinically used drugs that have been evaluated for their anti-allergic properties. Here, we examine each approach under investigation, summarize the test data generated and offer suggestions for further preclinical evaluation before their therapeutic potential can be realized.
Collapse
Affiliation(s)
- D F Finn
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
| | | |
Collapse
|
17
|
Abstract
Mast cells (MC) are potent innate immune cells that accumulate in chronically inflamed tissues. MC express the IL-33 receptor IL-1 receptor-related protein ST2 at high level, and this IL-1 family cytokine both activates MC directly and primes them to respond to other proinflammatory signals. Whether IL-33 and ST2 play a role in MC survival remains to be defined. In skin-derived human MC, we found that IL-33 attenuated MC apoptosis without altering proliferation, an effect mediated principally through the antiapoptotic molecule B-cell lymphoma-X large (BCLXL). Murine MC demonstrated a similar mechanism, dependent entirely on ST2. In line with these observations, St2(-/-) mice exhibited reduced numbers of tissue MC in inflamed arthritic joints, in helminth-infected intestine, and in normal peritoneum. To confirm an MC-intrinsic role for ST2 in vivo, we performed peritoneal transfer of WT and St2(-/-) MC. In St2(-/-) hosts treated with IL-33 and in WT hosts subjected to thioglycollate peritonitis, WT MC displayed a clear survival advantage over coengrafted St2(-/-) MC. IL-33 blockade specifically attenuated this survival advantage, confirming IL-33 as the relevant ST2 ligand mediating MC survival in vivo. Together, these data reveal a cell-intrinsic role for the IL-33/ST2 axis in the regulation of apoptosis in MC, identifying thereby a previously unappreciated pathway supporting expansion of the MC population with inflammation.
Collapse
|
18
|
A novel gadolinium-based trimetasphere metallofullerene for application as a magnetic resonance imaging contrast agent. Invest Radiol 2014; 48:745-54. [PMID: 23748228 DOI: 10.1097/rli.0b013e318294de5d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Macromolecular contrast agents for magnetic resonance imaging (MRI) are useful blood-pool agents because of their long systemic half-life and have found applications in monitoring tumor vasculature and angiogenesis. Macromolecular contrast agents have been able to overcome some of the disadvantages of the conventional small-molecule contrast agent Magnevist (gadolinium-diethylenetriaminepentaacetic acid), such as rapid extravasation and quick renal clearance, which limits the viable MRI time. There is an urgent need for new MRI contrast agents that increase the sensitivity of detection with a higher relaxivity, longer blood half-life, and reduced toxicity from free Gd3+ ions. Here, we report on the characterization of a novel water-soluble, derivatized, gadolinium-enclosed metallofullerene nanoparticle (Hydrochalarone-1) in development as an MRI contrast agent. MATERIALS AND METHODS The physicochemical properties of Hydrochalarone-1 were characterized by dynamic light scattering (hydrodynamic diameter), atomic force microscopy (particle height), ζ potential analysis (surface charge), and inductively coupled plasma-mass spectrometry (gadolinium concentration). The blood compatibility of Hydrochalarone-1 was also assessed in vitro through analysis of hemolysis, platelet aggregation, and complement activation of human blood. In vitro relaxivities, in vivo pharmacokinetics, and a pilot in vivo acute toxicity study were also performed. RESULTS An extensive in vitro and in vivo characterization of Hydrochalarone-1 is described here. The hydrodynamic size of Hydrochalarone-1 was 5 to 7 nm depending on the dispersing media, and it was negatively charged at physiological pH. Hydrochalarone-1 showed compatibility with blood cells in vitro, and no significant hemolysis, platelet aggregation, or complement activation was observed in vitro. In addition, Hydrochalarone-1 had significantly higher r1 and r2 in vitro relaxivities in human plasma in comparison with Magnevist and was not toxic at the doses administered in an in vivo pilot acute-dose toxicity study in mice.In vivo MRI pharmacokinetic analysis after a single intravenous injection of Hydrochalarone-1 (0.2 mmol Gd/kg) showed that the volume of distribution at steady state was approximately 100 mL/kg, suggesting prolonged systemic circulation. Hydrochalarone-1 also had a long blood half-life (88 minutes) and increased relaxivity, suggesting application as a promising blood-pool MRI contrast agent. CONCLUSIONS The evidence suggests that Hydrochalarone-1, with its long systemic half-life, may have significant utility as a blood-pool MRI contrast agent.
Collapse
|
19
|
Dellinger AL, Zhou Z, Kepley CL. A steroid-mimicking nanomaterial that mediates inhibition of human lung mast cell responses. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1185-93. [PMID: 24566277 DOI: 10.1016/j.nano.2014.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 01/18/2014] [Accepted: 02/12/2014] [Indexed: 12/20/2022]
Abstract
UNLABELLED Water-soluble fullerenes can be engineered to regulate activation of mast cells (MC) and control MC-driven diseases in vivo. To further understand their anti-inflammatory mechanisms a C70-based fullerene conjugated to four myo-inositol molecules (C70-I) was examined in vitro for its effects on the signaling pathways leading to mediator release from human lung MC. The C70-I fullerene stabilizes MC and acts synergistically with long-acting β2-adrenergic receptor agonists (LABA) to enhance inhibition of MC mediator release through FcεRI-simulation. The inhibition was paralleled by the upregulation of dual-specificity phosphatase one (DUSP1) gene and protein levels. Concomitantly, increases in MAPK were blunted in C70-I treated cells. The increase in DUSP1 expression was due to the ability of C70-I to prevent the ubiquitination and degradation of DUSP1. These findings identify a mechanism of how fullerenes inhibit inflammatory mediator release from MC and suggest they could potentially be an alternative therapy for steroid resistant asthmatics. FROM THE CLINICAL EDITOR This study investigates the role and mechanism of action of fullerenes in deactivating mast cell-based inflammation, paving the way to the development of a novel, non-steroid therapy in reactive airway disease.
Collapse
Affiliation(s)
- Anthony L Dellinger
- Luna nanoWorks Division, Luna Innovations Inc., Danville, VA, USA; University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, USA
| | - Zhiguo Zhou
- Luna nanoWorks Division, Luna Innovations Inc., Danville, VA, USA
| | - Christopher L Kepley
- University of North Carolina Greensboro, Joint School of Nanoscience and Nanoengineering, Greensboro, NC, USA.
| |
Collapse
|
20
|
Dellinger A, Zhou Z, Connor J, Madhankumar AB, Pamujula S, Sayes CM, Kepley CL. Application of fullerenes in nanomedicine: an update. Nanomedicine (Lond) 2014; 8:1191-208. [PMID: 23837857 DOI: 10.2217/nnm.13.99] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fullerenes are carbon spheres presently being pursued globally for a wide range of applications in nanomedicine. These molecules have unique electronic properties that make them attractive candidates for diagnostic, therapeutic and theranostic applications. Herein, the latest research is discussed on developing fullerene-based therapeutics as antioxidants for inflammatory diseases, their potential as antiviral/bacterial agents, utility as a drug delivery device and the promise of endohedral fullerenes as new MRI contrast agents. The recent discovery that certain fullerene derivatives can stabilize immune effector cells to prevent or inhibit the release of proinflammatory mediators makes them potential candidates for several diseases such as asthma, arthritis and multiple sclerosis. Gadolinium-containing endohedral fullerenes are being pursued as diagnostic MRI contrast agents for several diseases. Finally, a new class of fullerene-based theranostics has been developed, which combine therapeutic and diagnostic capabilities to specifically detect and kill cancer cells.
Collapse
Affiliation(s)
- Anthony Dellinger
- Joint School of Nanoscience & Nanoengineering, 2907 East Lee Street, Greensboro, NC 27401, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Smith MJ, Brown JM, Zamboni WC, Walker NJ. From immunotoxicity to nanotherapy: the effects of nanomaterials on the immune system. Toxicol Sci 2014; 138:249-55. [PMID: 24431216 DOI: 10.1093/toxsci/kfu005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The potential for human exposure to the diverse and ever-changing world of nanoscale materials has raised concerns about their influence on health and disease. The novel physical and chemical properties of these materials, which are associated with their small size, complicate toxicological evaluations. Further, these properties may make engineered nanomaterials (ENMs) a prime target for interaction with the immune system following uptake by phagocytes. Undesired effects on antigen-presenting cells and other phagocytic cells are of concern due to the high likelihood of ENM uptake by these cells. In addition, ENM interactions with lymphocytes and other cell types can contribute to a varied spectrum of possible effects, including inflammation, hypersensitivity, and immunomodulation. Furthermore, the mast cell (a type of immune cell traditionally associated with allergy) appears to contribute to certain inflammatory and toxic effects associated with some ENMs. Although incidental exposure may be undesirable, nanomedicines engineered for various clinical applications provide opportunities to develop therapies that may or may not intentionally target the immune system. The interaction between ENMs and the immune system and the resulting pharmacokinetic and phenotypic responses are critical factors that dictate the balance between toxicity and clinical efficacy of nanotherapeutics.
Collapse
|
22
|
Abstract
A range of mediators are generated during anaphylaxis, with redundancy of effects, multiple overlapping pathways, and involvement of several cell types. Key steps in the reaction occur at the site of initial contact, and mediators may not be detectable systemically. Furthermore, the potencies of various mediators vary enormously, and clinical effects may occur below our level of detection. We also do not know what converts (amplifies) a local reaction into systemic anaphylaxis. Murine models have identified several novel mediators that may propagate and/or regulate this process and also indicate that circulating neutrophils may play an important role in reaction amplification. Differential expression of various genes within specific intracellular signalling pathways of mediator release may further explain the varying severities of anaphylactic reactions. As our knowledge of the mechanisms of activation, key mediators, and the regulation of mediator release improves, new treatments for prevention and acute management may emerge.
Collapse
Affiliation(s)
- Shelley F Stone
- Centre for Clinical Research in Emergency Medicine, Western Australian Institute for Medical Research, University of Western Australia, Perth, Western Australia, Australia.
| | | |
Collapse
|
23
|
Wang X, Reece SP, Brown JM. Immunotoxicological impact of engineered nanomaterial exposure: mechanisms of immune cell modulation. Toxicol Mech Methods 2013; 23:168-77. [PMID: 23256453 DOI: 10.3109/15376516.2012.757686] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abstract Engineered nanomaterials (ENMs) are increasingly being utilized in many consumer products and various medical applications, thereby leading to the potentiality of increased human exposures. Assessment of the adverse effects on the immune system is an important component for evaluating the overall health and safety of ENM. Tasked with eliminating pathogens and removing cancerous cells, the immune system is constantly functioning to maintain homeostasis. Small modifications to the immune system, which may occur following ENM exposure, could lead to impaired protection or an inappropriate immune response resulting in autoimmunity and damage to the host. This review seeks to survey and evaluate the current literature to better understand the impact of ENM exposure on cells critical to the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Xiaojia Wang
- Department of Pharmacology & Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| | | | | |
Collapse
|
24
|
Dellinger A, Olson J, Link K, Vance S, Sandros MG, Yang J, Zhou Z, Kepley CL. Functionalization of gadolinium metallofullerenes for detecting atherosclerotic plaque lesions by cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2013; 15:7. [PMID: 23324435 PMCID: PMC3562260 DOI: 10.1186/1532-429x-15-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/17/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The hallmark of atherosclerosis is the accumulation of plaque in vessel walls. This process is initiated when monocytic cells differentiate into macrophage foam cells under conditions with high levels of atherogenic lipoproteins. Vulnerable plaque can dislodge, enter the blood stream, and result in acute myocardial infarction and stroke. Imaging techniques such as cardiovascular magnetic resonance (CMR) provides one strategy to identify patients with plaque accumulation. METHODS We synthesized an atherosclerotic-targeting contrast agent (ATCA) in which gadolinium (Gd)-containing endohedrals were functionalized and formulated into liposomes with CD36 ligands intercalated into the lipid bilayer. In vitro assays were used to assess the specificity of the ATCA for foam cells. The ability of ATCA to detect atherosclerotic plaque lesions in vivo was assessed using CMR. RESULTS The ATCA was able to detect scavenger receptor (CD36)-expressing foam cells in vitro and were specifically internalized via the CD36 receptor as determined by focused ion beam/scanning electron microscopy (FIB-SEM) and Western blotting analysis of CD36 receptor-specific signaling pathways. The ATCA exhibited time-dependent accumulation in atherosclerotic plaque lesions of ApoE -/- mice as determined using CMR. No ATCA accumulation was observed in vessels of wild type (C57/b6) controls. Non-targeted control compounds, without the plaque-targeting moieties, were not taken up by foam cells in vitro and did not bind plaque in vivo. Importantly, the ATCA injection was well tolerated, did not demonstrate toxicity in vitro or in vivo, and no accumulation was observed in the major organs. CONCLUSIONS The ATCA is specifically internalized by CD36 receptors on atherosclerotic plaque providing enhanced visualization of lesions under physiological conditions. These ATCA may provide new tools for physicians to non-invasively detect atherosclerotic disease.
Collapse
Affiliation(s)
- Anthony Dellinger
- Luna Innovations Incorporated, Luna nanoWorks Division, 521 Bridge St, Danville, VA, 24541, USA
- Joint School of Nanoscience and Nanoengineering, 2907 E Lee St, Greensboro, NC, 27401, USA
| | - John Olson
- Center for Biomolecular Imaging, Wake Forest University, 1 Medical Center Blvd, Winston Salem, NC, 27157, USA
| | - Kerry Link
- Center for Biomolecular Imaging, Wake Forest University, 1 Medical Center Blvd, Winston Salem, NC, 27157, USA
| | - Stephen Vance
- Joint School of Nanoscience and Nanoengineering, 2907 E Lee St, Greensboro, NC, 27401, USA
| | - Marinella G Sandros
- Joint School of Nanoscience and Nanoengineering, 2907 E Lee St, Greensboro, NC, 27401, USA
| | - Jijin Yang
- Carl Zeiss Microscopy, LLC, One Zeiss Drive, Thornwood, NY, 10594, USA
| | - Zhiguo Zhou
- Luna Innovations Incorporated, Luna nanoWorks Division, 521 Bridge St, Danville, VA, 24541, USA
| | - Christopher L Kepley
- Joint School of Nanoscience and Nanoengineering, 2907 E Lee St, Greensboro, NC, 27401, USA
| |
Collapse
|
25
|
Norton SK, Wijesinghe DS, Dellinger A, Sturgill J, Zhou Z, Barbour S, Chalfant C, Conrad DH, Kepley CL. Epoxyeicosatrienoic acids are involved in the C(70) fullerene derivative-induced control of allergic asthma. J Allergy Clin Immunol 2012; 130:761-769.e2. [PMID: 22664166 PMCID: PMC3955256 DOI: 10.1016/j.jaci.2012.04.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 03/13/2012] [Accepted: 04/06/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Fullerenes are molecules being investigated for a wide range of therapeutic applications. We have shown previously that certain fullerene derivatives (FDs) inhibit mast cell (MC) function in vitro, and here we examine their in vivo therapeutic effect on asthma, a disease in which MCs play a predominant role. OBJECTIVE We sought to determine whether an efficient MC-stabilizing FD (C(70)-tetraglycolate [TGA]) can inhibit asthma pathogenesis in vivo and to examine its in vivo mechanism of action. METHODS Asthma was induced in mice, and animals were treated intranasally with TGA either simultaneously with treatment or after induction of pathogenesis. The efficacy of TGA was determined through the measurement of airway inflammation, bronchoconstriction, serum IgE levels, and bronchoalveolar lavage fluid cytokine and eicosanoid levels. RESULTS We found that TGA-treated mice have significantly reduced airway inflammation, eosinophilia, and bronchoconstriction. The TGA treatments are effective, even when given after disease is established. Moreover, we report a novel inhibitory mechanism because TGA stimulates the production of an anti-inflammatory P-450 eicosanoid metabolites (cis-epoxyeicosatrienoic acids [EETs]) in the lung. Inhibitors of these anti-inflammatory EETs reversed TGA inhibition. In human lung MCs incubated with TGA, there was a significant upregulation of CYP1B gene expression, and TGA also reduced IgE production from B cells. Lastly, MCs incubated with EET and challenged through FcεRI had a significant blunting of mediator release compared with nontreated cells. CONCLUSION The inhibitory capabilities of TGA reported here suggest that FDs might be used a platform for developing treatments for asthma.
Collapse
Affiliation(s)
- Sarah K Norton
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shannahan JH, Kodavanti UP, Brown JM. Manufactured and airborne nanoparticle cardiopulmonary interactions: a review of mechanisms and the possible contribution of mast cells. Inhal Toxicol 2012; 24:320-39. [PMID: 22486349 DOI: 10.3109/08958378.2012.668229] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human inhalation exposures to manufactured nanoparticles (NP) and airborne ultrafine particles (UFP) continues to increase in both occupational and environmental settings. UFP exposures have been associated with increased cardiovascular mortality and morbidity, while ongoing research supports adverse systemic and cardiovascular health effects after NP exposures. Adverse cardiovascular health effects include alterations in heart rate variability, hypertension, thrombosis, arrhythmias, increased myocardial infarction, and atherosclerosis. Exactly how UFP and NP cause these negative cardiovascular effects is poorly understood, however a variety of mediators and mechanisms have been proposed. UFP and NP, as well as their soluble components, are known to systemically translocate from the lung. Translocated particles could mediate cardiovascular toxicity through direct interactions with the vasculature, blood, and heart. Recent study suggests that sensory nerve stimulation within the lung may also contribute to UFP- and NP-induced acute cardiovascular alterations. Activation of sensory nerves, such as C-fibers, within the lung may result in altered cardiac rhythm and function. Lastly, release of pulmonary-derived mediators into systemic circulation has been proposed to facilitate cardiovascular effects. In general, these proposed pulmonary-derived mediators include proinflammatory cytokines, oxidatively modified macromolecules, vasoactive proteins, and prothrombotic factors. These pulmonary-derived mediators have been postulated to contribute to the subsequent prothrombotic, atherogenic, and inflammatory effects after exposure. This review will evaluate the potential contribution of individual mediators and mechanisms in facilitating cardiopulmonary toxicity following inhalation of UFP and NP. Lastly, we will appraise the literature and propose a hypothesis regarding the possible role of mast cells in contributing to these systemic effects.
Collapse
Affiliation(s)
- Jonathan H Shannahan
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | | |
Collapse
|