1
|
Zhou CY, Yang YL, Han ZY, Chen YX, Liu HL, Fan K, Li MC, Tu SH, Wen Q, Zhou XY, Ma L. Peripheral blood MR1 tetramer-positive mucosal-associated invariant T-cell function is modulated by mammalian target of rapamycin complex 1 in patients with active tuberculosis. Immunology 2024; 173:497-510. [PMID: 39022997 DOI: 10.1111/imm.13834] [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: 03/22/2023] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Tuberculosis (TB) is still an urgent global public health problem. Notably, mucosal-associated invariant T (MAIT) cells play an important role in early anti-TB immune response. Targeted control of them may be an effective method to improve vaccine efficacy and TB treatment. However, the biology and signal regulation mechanisms of MAIT cells in TB patients are still poorly understood. Previous studies have been limited by the lack of reagents to specifically identify MAIT cells. In addition, the use of alternative markers may subsume non-MAIT cell into MAIT cell populations. In this study, the human MR1 tetramer which can specifically identify MAIT cells was used to further explore the effect and mechanism of MAIT cells in anti-TB immune response. Our results showed that the tetramer+ MAIT cells in peripheral blood of TB patients were mainly CD8+ or CD4-CD8- cells, and very few were CD4+ cells. After BCG infecting autologous antigen-presenting cells, MAIT cells in patients produced significantly higher levels of cytokines, lysis and proliferation compared with healthy controls. After suppression of mTORC1 by the mTORC1-specific inhibitor rapamycin, the immune response of MAIT cells in patients was significantly reduced. This study demonstrates that peripheral blood tetramer+ MAIT cells from TB patients have significant anti-TB immune effect, which is regulated by mTORC1. This could provide ideas and potential therapeutic targets for the development of novel anti-TB immunotherapy.
Collapse
Affiliation(s)
- Chao-Ying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ya-Long Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Zhen-Yu Han
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Yao-Xin Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Hong-Lin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ke Fan
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ming-Chong Li
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Si-Hang Tu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Xin-Ying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Martínez Sanmiguel JJ, Rincón-López JA, Rangel-López R, Hermann-Muñoz JA, Franco Molina M, Rodríguez Padilla C, Alvarado-Orozco JM, Zárate Triviño DG. In Vitro and In Vivo Biological Properties of Calcium Silicophosphate-Based Bone Grafts: Silicocarnotite and Nagelschmidtite. ACS APPLIED BIO MATERIALS 2024; 7:5318-5336. [PMID: 39029129 DOI: 10.1021/acsabm.4c00538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Accidents, trauma, bone defects, and oncological processes significantly impact patients' health and quality of life. While calcium phosphates and bioactive glasses are commonly used as bone fillers to facilitate bone regeneration in orthopedics and traumatology, they exhibit certain disadvantages compared to calcium silicophosphate phases. This study evaluates the in vitro cytocompatibility and in vivo osteogenic properties of two-third-generation ceramic phases: silicocarnotite (SC) and nagelschmidtite (Nagel). These phases were synthesized via a solid-state reaction and characterized using X-ray diffraction and scanning electron microscopy. In vitro behavior was assessed through bioactivity tests, cell viability, proliferation, and inflammatory profiles by detecting cytokines and reactive oxygen species. Osteogenic properties were evaluated by detecting bone-associated proteins in MG-G3, hFOB1.19, and MC3T3-E1 cell lines after 3, 7, and 14 days. 45S5 Bioactive glass (BG), hydroxyapatite (HAp), and osteogenic medium were employed as control standards for bone formation. SC and Nagel phases exhibited higher viability percentages as well as osteoconductive and osteoinductive behavior. Finally, SC and Nagel bone grafts were implanted in a Wistar rat model to assess their in vivo ability to induce bone formation, demonstrating complete osseointegration after 12 weeks. Histological evaluation revealed osteocytes forming osteons and the presence of blood vessels, particularly in rats implanted with Nagel. Given their favorable biological performance, SC and Nagel emerge as promising candidates for bone grafts in orthopedics, traumatology, and maxillofacial surgery.
Collapse
Affiliation(s)
- Juan José Martínez Sanmiguel
- Centro de Ingeniería y Desarrollo Industrial, Avenida Playa Pie de la Cuesta No. 702 Desarrollo San Pablo, Querétaro 76125, México
| | - July Andrea Rincón-López
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro Libramiento Norponiente #2000, Querétaro C.P. 76230, México
| | - Raúl Rangel-López
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| | - Jennifer Andrea Hermann-Muñoz
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro Libramiento Norponiente #2000, Querétaro C.P. 76230, México
| | - Moisés Franco Molina
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| | - Cristina Rodríguez Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| | - Juan Manuel Alvarado-Orozco
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro Libramiento Norponiente #2000, Querétaro C.P. 76230, México
| | - Diana G Zárate Triviño
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| |
Collapse
|
3
|
Lopez-Pardo A, Amarilla-Irusta A, Sandá V, Stan-Fontoba M, Borrego F, Amo L. Degranulation assay to evaluate NK cell natural and antibody-dependent cell-mediated cytotoxicity against A549 tumor spheroids. Methods Cell Biol 2024; 189:97-115. [PMID: 39393889 DOI: 10.1016/bs.mcb.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Adoptive natural killer (NK) cell-based immunotherapy is a promising treatment approach in cancer that is showing notable efficacy against hematological malignancies. However, the success of NK cell immunotherapy in patients with solid tumors is limited due to several barriers, which include the immunosuppressive tumor microenvironment (TME), heterogeneity of tumor cells and poor NK cell infiltration into the tumor. Advances in 3D in vitro culture technologies have opened new avenues for the development of more physiological human cancer models that mimic important tumor features which are absent in traditional 2D studies and may be essential for the improvement of immunotherapies against solid tumors. Here, we describe a comprehensive protocol to generate tumor spheroids from the A549 lung carcinoma cell line, then establish co-cultures with NK cells to, ultimately, determine NK cell functional response with a degranulation assay, a surrogate of NK cell cytotoxicity against tumor spheroids. Additionally, we studied degranulation by stimulating NK cell antibody-dependent cell-mediated cytotoxicity (ADCC) with cetuximab, an IgG1 monoclonal antibody used in cancer therapy. Likewise, other monoclonal antibodies or combination treatments could also be studied in this 3D co-culture system, providing very valuable information to define effective combinations of therapeutic agents able to generate NK cells with high cytotoxic potential that could lead to more successful adoptive NK cell-based therapies for the treatment of solid tumors.
Collapse
Affiliation(s)
- Ainara Lopez-Pardo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Víctor Sandá
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Mario Stan-Fontoba
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Francisco Borrego
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Laura Amo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
4
|
Aherne O, Mørch M, Ortiz R, Shannon O, Davies JR. A novel multiplex fluorescent-labeling method for the visualization of mixed-species biofilms in vitro. Microbiol Spectr 2024; 12:e0025324. [PMID: 38785429 PMCID: PMC11218471 DOI: 10.1128/spectrum.00253-24] [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: 01/30/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
In nature, bacteria usually exist as mixed-species biofilms, where they engage in a range of synergistic and antagonistic interactions that increase their resistance to environmental challenges. Biofilms are a major cause of persistent infections, and dispersal from initial foci can cause new infections at distal sites thus warranting further investigation. Studies of development and spatial interactions in mixed-species biofilms can be challenging due to difficulties in identifying the different bacterial species in situ. Here, we apply CellTrace dyes to studies of biofilm bacteria and present a novel application for multiplex labeling, allowing identification of different bacteria in mixed-species, in vitro biofilm models. Oral bacteria labeled with CellTrace dyes (far red, yellow, violet, and CFSE [green]) were used to create single- and mixed-species biofilms, which were analyzed with confocal spinning disk microscopy (CSDM). Biofilm supernatants were studied with flow cytometry (FC). Both Gram-positive and Gram-negative bacteria were well labeled and CSDM revealed biofilms with clear morphology and stable staining for up to 4 days. Analysis of CellTrace labeled cells in supernatants using FC showed differences in the biofilm dispersal between bacterial species. Multiplexing with different colored dyes allowed visualization of spatial relationships between bacteria in mixed-species biofilms and relative coverage by the different species was revealed through segmentation of the CSDM images. This novel application, thus, offers a powerful tool for studying structure and composition of mixed-species biofilms in vitro.IMPORTANCEAlthough most chronic infections are caused by mixed-species biofilms, much of our knowledge still comes from planktonic cultures of single bacterial species. Studies of formation and development of mixed-species biofilms are, therefore, required. This work describes a method applicable to labeling of bacteria for in vitro studies of biofilm structure and dispersal. Critically, labeled bacteria can be multiplexed for identification of different species in mixed-species biofilms using confocal spinning disk microscopy, facilitating investigation of biofilm development and spatial interactions under different environmental conditions. The study is an important step in increasing the tools available for such complex and challenging studies.
Collapse
Affiliation(s)
- Olivia Aherne
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- CR Competence, Lund, Sweden
| | - Martina Mørch
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | | | - Oonagh Shannon
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Julia R Davies
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| |
Collapse
|
5
|
Fourneaux C, Racine L, Koering C, Dussurgey S, Vallin E, Moussy A, Parmentier R, Brunard F, Stockholm D, Modolo L, Picard F, Gandrillon O, Paldi A, Gonin-Giraud S. Differentiation is accompanied by a progressive loss in transcriptional memory. BMC Biol 2024; 22:58. [PMID: 38468285 PMCID: PMC10929117 DOI: 10.1186/s12915-024-01846-9] [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: 04/05/2023] [Accepted: 02/13/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Cell differentiation requires the integration of two opposite processes, a stabilizing cellular memory, especially at the transcriptional scale, and a burst of gene expression variability which follows the differentiation induction. Therefore, the actual capacity of a cell to undergo phenotypic change during a differentiation process relies upon a modification in this balance which favors change-inducing gene expression variability. However, there are no experimental data providing insight on how fast the transcriptomes of identical cells would diverge on the scale of the very first two cell divisions during the differentiation process. RESULTS In order to quantitatively address this question, we developed different experimental methods to recover the transcriptomes of related cells, after one and two divisions, while preserving the information about their lineage at the scale of a single cell division. We analyzed the transcriptomes of related cells from two differentiation biological systems (human CD34+ cells and T2EC chicken primary erythrocytic progenitors) using two different single-cell transcriptomics technologies (scRT-qPCR and scRNA-seq). CONCLUSIONS We identified that the gene transcription profiles of differentiating sister cells are more similar to each other than to those of non-related cells of the same type, sharing the same environment and undergoing similar biological processes. More importantly, we observed greater discrepancies between differentiating sister cells than between self-renewing sister cells. Furthermore, a progressive increase in this divergence from first generation to second generation was observed when comparing differentiating cousin cells to self renewing cousin cells. Our results are in favor of a gradual erasure of transcriptional memory during the differentiation process.
Collapse
Affiliation(s)
- Camille Fourneaux
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Laëtitia Racine
- Ecole Pratique des Hautes Etudes, PSL Research University, Sorbonne Université, INSERM, CRSA, Paris, 75012, France
| | - Catherine Koering
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Sébastien Dussurgey
- Plateforme AniRA-Cytométrie, Université Claude Bernard Lyon 1, CNRS UAR3444, Inserm US8, ENS de Lyon, SFR Biosciences, Lyon, F-69007, France
| | - Elodie Vallin
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Alice Moussy
- Ecole Pratique des Hautes Etudes, PSL Research University, Sorbonne Université, INSERM, CRSA, Paris, 75012, France
| | - Romuald Parmentier
- Ecole Pratique des Hautes Etudes, PSL Research University, Sorbonne Université, INSERM, CRSA, Paris, 75012, France
| | - Fanny Brunard
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Daniel Stockholm
- Ecole Pratique des Hautes Etudes, PSL Research University, Sorbonne Université, INSERM, CRSA, Paris, 75012, France
| | - Laurent Modolo
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Franck Picard
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Olivier Gandrillon
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France
- Inria Center, Grenoble Rhone-Alpes, Equipe Dracula, Villeurbanne, F69100, France
| | - Andras Paldi
- Ecole Pratique des Hautes Etudes, PSL Research University, Sorbonne Université, INSERM, CRSA, Paris, 75012, France
| | - Sandrine Gonin-Giraud
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS, UMR5239, Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
6
|
Pierzchalski A, Zenclussen AC, Herberth G. A comprehensive battery of flow cytometric immunoassays for the in vitro testing of chemical effects in human blood cells. Front Immunol 2024; 14:1327960. [PMID: 38229911 PMCID: PMC10790304 DOI: 10.3389/fimmu.2023.1327960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024] Open
Abstract
Background There is a growing need for immunological assays to test toxic and modulatory effects of chemicals. The assays should be easy to use, reproducible and superior to cell line-based assays. We have therefore developed a comprehensive portfolio of assays based on primary human blood cells that are suitable for testing chemical effects. Methods The flow cytometry-based assays were designed to target a wide range of human peripheral blood mononuclear cells and whole blood, including T cells, NK cells, B cells, basophils and innate-like T cells such as γδT, MAIT and NKT cells. We have selected a set of activation markers for each immune cell, e.g: CD154 (T cells), CD137, CD107a (NK cells), CD63 (basophils), CD69, CD83 (B cells), CD69, IFN-γ (MAIT cells) and we selected cell specific stimuli: aCD3 antibodies (T cells); E. coli and cytokines IL-12/15/18 (MAIT cells); CpG ODN2006, R848 or aCD40 antibodies (B cells), fMLP or aFcϵR1 (basophils) or K562 cells (NK cells). Results By selecting immune cell-specific markers and cell-specific stimuli, we were able to induce particular immune responses from the targeted immune cells. For example, the response to stimulation with anti-CD3 antibodies was in 36.8% of CD107a+CD8+ cells. Cytokine stimulation induced the production of IFN-γ in 30% of MAIT cells. After stimulation with E. coli, around 50% of MAIT cells produced TNF. About 40% of basophils responded to aFcƐR1 stimulation. Similar activation ranges were achieved in K562-stimulated NK cells. Conclusion Our test portfolio covers the most relevant immune cells present in human blood, providing a solid basis for in vitro toxicity and immunomodulatory testing of chemicals. By using human blood, the natural composition of cells found in the blood can be determined and the effects of chemicals can be detected at the cellular level.
Collapse
Affiliation(s)
- Arkadiusz Pierzchalski
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany
| | - Ana C. Zenclussen
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany
- Perinatal Immunology Research Group, Medical Faculty, Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Gunda Herberth
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany
| |
Collapse
|
7
|
Bhatta R, Han J, Liu Y, Bo Y, Lee D, Zhou J, Wang Y, Nelson ER, Chen Q, Zhang XS, Hassaneen W, Wang H. Metabolic tagging of extracellular vesicles and development of enhanced extracellular vesicle based cancer vaccines. Nat Commun 2023; 14:8047. [PMID: 38052869 PMCID: PMC10697976 DOI: 10.1038/s41467-023-43914-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023] Open
Abstract
As key mediators of cellular communication, extracellular vesicles (EVs) have been actively explored for diagnostic and therapeutic applications. However, effective methods to functionalize EVs and modulate the interaction between EVs and recipient cells are still lacking. Here we report a facile and universal metabolic tagging technology that can install unique chemical tags (e.g., azido groups) onto EVs. The surface chemical tags enable conjugation of molecules via efficient click chemistry, for the tracking and targeted modulation of EVs. In the context of tumor EV vaccines, we show that the conjugation of toll-like receptor 9 agonists onto EVs enables timely activation of dendritic cells and generation of superior antitumor CD8+ T cell response. These lead to 80% tumor-free survival against E.G7 lymphoma and 33% tumor-free survival against B16F10 melanoma. Our study yields a universal technology to generate chemically tagged EVs from parent cells, modulate EV-cell interactions, and develop potent EV vaccines.
Collapse
Affiliation(s)
- Rimsha Bhatta
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - David Lee
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Erik Russell Nelson
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-, Champaign, IL, USA
| | - Qian Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xiaojia Shelly Zhang
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- National Center for Supercomputing Applications, Urbana, IL, 61801, USA
| | - Wael Hassaneen
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carle Foundation Hospital, Urbana, IL, 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
8
|
Xia M, Wang B, Sun W, Ji D, Zhou H, Huang X, Yu M, Su Z, Chen P, Qu K, Wang X. Lsd1 safeguards T-cell development via suppressing endogenous retroelements and interferon responses. Life Sci Alliance 2023; 6:e202302042. [PMID: 37429639 PMCID: PMC10345215 DOI: 10.26508/lsa.202302042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/12/2023] Open
Abstract
The histone demethylase Lsd1 has been shown to play multiple essential roles in mammalian biology. However, its physiological functions in thymocyte development remain elusive. We observed that the specific deletion of Lsd1 in thymocytes caused significant thymic atrophy and reduced peripheral T cell populations with impaired proliferation capacity. Single-cell RNA sequencing combined with strand-specific total RNA-seq and ChIP-seq analysis revealed that ablation of Lsd1 led to the aberrant derepression of endogenous retroelements, which resulted in a viral mimicry state and activated the interferon pathway. Furthermore, the deletion of Lsd1 blocked the programmed sequential down-regulation of CD8 expression at the DP→CD4+CD8lo stage and induced an innate memory phenotype in both thymic and peripheral T cells. Single-cell TCR sequencing revealed the kinetics of TCR recombination in the mouse thymus. However, the preactivation state after Lsd1 deletion neither disturbed the timeline of TCR rearrangement nor reshaped the TCR repertoire of SP cells. Overall, our study provides new insight into the function of Lsd1 as an important maintainer of endogenous retroelement homeostasis in early T-cell development.
Collapse
Affiliation(s)
- Miaoran Xia
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Bingbing Wang
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Capital Medical University, Beijing, China
| | - Wujianan Sun
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dengyu Ji
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Hang Zhou
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Xuefeng Huang
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Capital Medical University, Beijing, China
| | - Minghang Yu
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Capital Medical University, Beijing, China
| | - Ziyang Su
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Capital Medical University, Beijing, China
| | - Ping Chen
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
| | - Kun Qu
- Department of Oncology, The First Affiliated Hospital of USTC, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Xi Wang
- Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory for Tumor Invasion and Metastasis, Capital Medical University, Beijing, China
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Lee JY, Park S, Han AR, Hwang HS, Kim HJ. Therapeutic potential of FLT4-targeting peptide in acute myeloid leukemia. Cancer Immunol Immunother 2023; 72:2919-2925. [PMID: 36763100 PMCID: PMC10991567 DOI: 10.1007/s00262-023-03385-8] [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: 09/16/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
Previously, we found that dysfunctional natural killer (NK) cells with low interferon gamma (IFN-γ) were restored in acute myeloid leukemia (AML) by the FLT4 antagonist MAZ51. Here, we developed 12 peptides targeting FLT4 for clinical application and examined whether they restored the frequency of lymphocytes, especially T cells and NK cells, and high IFN-γ expression, as MAZ51 treatment did in our previous study. Although clinical data from using peptides are currently available, peptides targeting FLT4 to modulate immune cells have not been fully elucidated. In this study, we focus on novel peptide 4 (P4) from the intracellular domain of FLT4 because it had dominant negative activity. Similar to MAZ51, high IFN-γ levels were expressed in AML-mononuclear cells exposed to P4. Additionally, T and NK cell levels were restored, as were high IFN-γ levels, in a leukemic environment when P4 was treated. Interestingly, the regulatory T cells were significantly decreased by P4, implying the role of peptide in tumor niche. Overall, we demonstrated the therapeutic value of functionally modulating lymphocytes using a peptide targeting FLT4 and proposed the development of advanced therapeutic approaches against AML by using immune cells.
Collapse
Affiliation(s)
- Ji Yoon Lee
- CHA Advanced Research Institute, Bundang CHA Hospital, CHA University, Seongnam, Korea
| | - Soojin Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - A-Reum Han
- CHA Advanced Research Institute, Bundang CHA Hospital, CHA University, Seongnam, Korea
| | - Hee-Sun Hwang
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Catholic Hematology Hospital, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
- Department of Hematology, College of Medicine, Leukemia Research Institute, Catholic Hematology Hospital, Seoul St. Mary's Hospital, The Catholic University of Korea, 222, Banpo-Daero, Seocho-Gu, Seoul, 06591, Korea.
| |
Collapse
|
10
|
Poloni C, Schonhofer C, Ivison S, Levings MK, Steiner TS, Cook L. T-cell activation-induced marker assays in health and disease. Immunol Cell Biol 2023; 101:491-503. [PMID: 36825901 PMCID: PMC10952637 DOI: 10.1111/imcb.12636] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023]
Abstract
Activation-induced marker (AIM) assays have proven to be an accessible and rapid means of antigen-specific T-cell detection. The method typically involves short-term incubation of whole blood or peripheral blood mononuclear cells with antigens of interest, where autologous antigen-presenting cells process and present peptides in complex with major histocompatibility complex (MHC) molecules. Recognition of peptide-MHC complexes by T-cell receptors then induces upregulation of activation markers on the T cells that can be detected by flow cytometry. In this review, we highlight the most widely used activation markers for assays in the literature while identifying nuances and potential downfalls associated with the technique. We provide a summary of how AIM assays have been used in both discovery science and clinical studies, including studies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity. This review primarily focuses on AIM assays using human blood or peripheral blood mononuclear cell samples, with some considerations noted for tissue-derived T cells and nonhuman samples. AIM assays are a powerful tool that enables detailed analysis of antigen-specific T-cell frequency, phenotype and function without needing to know the precise antigenic peptides and their MHC restriction elements, enabling a wider analysis of immunity generated following infection and/or vaccination.
Collapse
Affiliation(s)
- Chad Poloni
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- BC Children's Hospital Research InstituteVancouverBCCanada
| | - Cole Schonhofer
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- BC Children's Hospital Research InstituteVancouverBCCanada
| | - Sabine Ivison
- BC Children's Hospital Research InstituteVancouverBCCanada
- Department of SurgeryUniversity of British ColumbiaVancouverBCCanada
| | - Megan K Levings
- BC Children's Hospital Research InstituteVancouverBCCanada
- Department of SurgeryUniversity of British ColumbiaVancouverBCCanada
| | - Theodore S Steiner
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- BC Children's Hospital Research InstituteVancouverBCCanada
| | - Laura Cook
- Division of Infectious Diseases, Department of MedicineUniversity of British ColumbiaVancouverBCCanada
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Department of Critical Care, Melbourne Medical SchoolUniversity of MelbourneMelbourneAustralia
| |
Collapse
|
11
|
Wang J, Jiang Y, Yang Y, Xu K, Wang X, Yang R, Xiao X, Sun H. Nanoparticulate impurities in the pharmaceutical excipient trehalose induce an early immune response. Eur J Pharm Biopharm 2023:S0939-6411(23)00164-9. [PMID: 37354998 DOI: 10.1016/j.ejpb.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/05/2023] [Accepted: 06/19/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Pharmaceutical excipients are an important part of biological products. However, few attempts have been made to distinguish between the risk of inflammation associated with the biological products themselves and that associated with excipients. The analysis of early immune response risk associated with excipients added to biological products is an important step in exploring the complex mechanism of side effects in susceptible patients. METHODS AND RESULTS In this study, nanoparticle impurities (NPIs) were extracted from trehalose and characterized. A mouse popliteal lymph node cell (PLNA) model, a mouse spleen lymphocyte model, a human peripheral blood mononuclear cell cytokine release model, and a macrophage complement activation model were established to comprehensively evaluate the early immune risk related to impurities in the trehalose excipient. Although popliteal lymph node cell counts in mice did not show significant differences, all other models indicated possible immune risk. In the PLNA model, NPIs caused significant toe thickening in mice, whereby the content of IgE and MCP-1 increased significantly. NPIs significantly increased the proliferation and differentiation of spleen lymphocytes according to the CCK-8 assay and flow cytometry. After treatment with NPIs, the release of IgE and a variety of cytokines (MIP-1α, IFN-γ, IL-2, IL-8, TNF-α, IL-6, IL-1α) in human peripheral blood cells was significantly increased according to ELISA, while a concomitant increase of C3a/C5a as well as C4a/Bb proved that NPIs activated the complement system. CONCLUSION NPIs from trehalose elicited an immune response in vitro, and the immune response to trehalose may be related to NPIs and not the excipient itself. Different batches of trehalose showed different immune response effects. The currents research suggests that when trehalose is applied in high-risk administration routes, NPIs should be assessed and reasonably controlled.
Collapse
Affiliation(s)
- Jue Wang
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China
| | - Ying Jiang
- Shanghai Medical Device and Cosmetics Evaluation and Verification Center, Shanghai, China
| | - Yang Yang
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China
| | - Kai Xu
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China
| | - Xiaofeng Wang
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China
| | - Rui Yang
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China
| | - Xinyue Xiao
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China
| | - Huimin Sun
- National Institutes for Food and Drug Control, National Key Laboratory for Quality Control of Pharmaceutical Excipients, Beijing, China.
| |
Collapse
|
12
|
Multi-component immune knockout: A strategy for studying the effective components of traditional Chinese medicine. J Chromatogr A 2023; 1692:463853. [PMID: 36780848 DOI: 10.1016/j.chroma.2023.463853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/09/2023]
Abstract
Periploca forrestii Schltr., a traditional Chinese medicine (TCM), is commonly used to treat autoimmune diseases such as rheumatoid arthritis (RA). However, its mechanism, involving a variety of cardiac glycosides, remains largely unknown. The immune knockout strategy can highly selectively deplete target components by immunoaffinity chromatography (IAC). We aimed to identify the common structural features of cardiac glycosides in P. forrestii and design IAC to specifically recognize these features to achieve the multi-component knockout of potential active substances from the extracts of P. forrestii. A content detection experiment confirmed that the content of a compound with periplogenin structure (CPS) in the extract of P. forrestii was reduced by 45% by IAC of periplogenin. The immunosuppressive ability of the extract on H9 human T lymphocytic cells was weakened after CPS knockout from P. forrestii extract. Molecular biology experiments showed that mRNA expression of interferon-γ (IFN-γ), interleukin-2 (IL-2), and interleukin-6 (IL-6) in H9 cells was up-regulated after CPS knockout, while no significant changes in the expression of interleukin-4 (IL-4) were found. CPS knockout from P. forrestii extract did not cause significant changes in the proliferation of lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage cells incubated with this extract. These results indicate that CPS exhibited immunosuppressive effects via inhibiting the T helper 1 (Th1) cell immune response and not via the anti-inflammatory components in P. forrestii. This is the first use of IAC to achieve multi-component knockout in TCM extracts for identifying effective compounds. This method is effective and reliable and warrants further exploration.
Collapse
|
13
|
Terrén I, Astarloa-Pando G, Amarilla-Irusta A, Borrego F. P815-based redirected degranulation assay to study human NK cell effector functions. Methods Cell Biol 2023; 173:33-48. [PMID: 36653084 DOI: 10.1016/bs.mcb.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Natural killer (NK) cells are part of the innate immune system, the classic cytotoxic population of innate lymphoid cells (ILCs). They can directly kill virus-infected or tumor cells through different mechanisms without prior sensitization using their lytic functions in response to different signals (target cell ligands and/or inflammatory cytokines) and secreting cytokines, such as interferon gamma (IFNγ) and tumor necrosis factor (TNF). NK cells use antibody-dependent cell-mediated cytotoxicity (ADCC) to recognize and kill cells expressing target antigens when they are antibody coated. Redirected cytotoxicity is a technique used to target cells that do not per se activate NK cells. Here, we use redirected degranulation, a surrogate technique that correlates with redirected lysis. The P815 cell line (mouse mastocytoma) express fragment crystallizable gamma receptor II (FcγRII) and therefore could bind the Fc portion of mouse IgG antibodies, which through their fragment antigen-binding (Fab) may recognize NK cells activating receptors leading to target cell lysis. This technique could be used to determine the inhibitory or activating capacity of different receptors or isoforms and in immunotherapy using T cell and NK cell activators.
Collapse
Affiliation(s)
- Iñigo Terrén
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, Barakaldo, Spain
| | | | | | - Francisco Borrego
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
14
|
Maddalon A, Iulini M, Melzi G, Corsini E, Galbiati V. New Approach Methodologies in Immunotoxicology: Challenges and Opportunities. Endocr Metab Immune Disord Drug Targets 2023; 23:1681-1698. [PMID: 37069707 DOI: 10.2174/1871530323666230413081128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 04/19/2023]
Abstract
To maintain the integrity of an organism, a well-functioning immune system is essential. Immunity is dynamic, with constant surveillance needed to determine whether to initiate an immune response or to not respond. Both inappropriate immunostimulation and decreased immune response can be harmful to the host. A reduced immune response can lead to high susceptibility to cancer or infections, whereas an increased immune response can be related to autoimmunity or hypersensitivity reactions. Animal testing has been the gold standard for hazard assessment in immunotoxicity but a lot of efforts are ongoing to develop non-animal-based test systems, and important successes have been achieved. The term "new approach methodologies" (NAMs) refer to the approaches which are not based on animal models. They are applied in hazard and risk assessment of chemicals and include approaches such as defined approaches for data interpretation and integrated approaches to testing and assessment. This review aims to summarize the available NAMs for immunotoxicity assessment, taking into consideration both inappropriate immunostimulation and immunosuppression, including implication for cancer development.
Collapse
Affiliation(s)
- Ambra Maddalon
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Toxicology, Università degli Studi di Milano, Milan, Italy
| | - Martina Iulini
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Toxicology, Università degli Studi di Milano, Milan, Italy
| | - Gloria Melzi
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Toxicology, Università degli Studi di Milano, Milan, Italy
| | - Emanuela Corsini
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Toxicology, Università degli Studi di Milano, Milan, Italy
| | - Valentina Galbiati
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Toxicology, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
15
|
Neddylation tunes peripheral blood mononuclear cells immune response in COVID-19 patients. Cell Death Dis 2022; 8:316. [PMID: 35831294 PMCID: PMC9277603 DOI: 10.1038/s41420-022-01115-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has reached 5.5 million deaths worldwide, generating a huge impact globally. This highly contagious viral infection produces a severe acute respiratory syndrome that includes cough, mucus, fever and pneumonia. Likewise, many hospitalized patients develop severe pneumonia associated with acute respiratory distress syndrome (ARDS), along an exacerbated and uncontrolled systemic inflammation that in some cases induces a fatal cytokine storm. Although vaccines clearly have had a beneficial effect, there is still a high percentage of unprotected patients that develop the pathology, due to an ineffective immune response. Therefore, a thorough understanding of the modulatory mechanisms that regulate the response to SARS-CoV-2 is crucial to find effective therapeutic alternatives. Previous studies describe the relevance of Neddylation in the activation of the immune system and its implications in viral infection. In this context, the present study postulates Neddylation, a reversible ubiquitin-like post-translational modification of proteins that control their stability, localization and activity, as a key regulator in the immune response against SARS-CoV-2. For the first time, we describe an increase in global neddylation levels in COVID-19 in the serum of patients, which is particularly associated with the early response to infection. In addition, the results showed that overactivation of neddylation controls activation, proliferation, and response of peripheral blood mononuclear cells (PBMCs) isolated from COVID-19 patients. Inhibition of neddylation, and the subsequent avoidance of activated PBMCs, reduces cytokine production, mainly IL-6 and MCP-1 and induce proteome modulation, being a critical mechanism and a potential approach to immunomodulate COVID-19 patients.
Collapse
|
16
|
Badiee P, Maritz MF, Dmochowska N, Cheah E, Thierry B. Intratumoral Anti-PD-1 Nanoformulation Improves Its Biodistribution. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15881-15893. [PMID: 35357803 DOI: 10.1021/acsami.1c22479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Intratumoral administration of immune checkpoint inhibitors, such as programmed cell death-1 antibodies (aPD-1), is a promising approach toward addressing both the low patients' responses and high off-target toxicity, but good preclinical results have not translated in phase I clinical studies as significant off-target toxicities were observed. We hypothesized that the nanoformulation of aPD-1 could alter both their loco-regional and systemic distribution following intratumoral administration. To test this hypothesis, we developed an aPD-1 nanoformulation (aPD-1 NPs) and investigated its biodistribution following intratumoral injection in an orthotopic mice model of head and neck cancer. Biodistribution analysis demonstrated a significantly lower distribution in off-target organs of the nanoformulated aPD-1 compared to free antibodies. On the other hand, both aPD-1 NPs and free aPD-1 yielded a significantly higher tumor and tumor draining lymph node accumulation than the systemically administrated free aPD-1 used as the current clinical benchmark. In a set of comprehensive in vitro biological studies, aPD-1 NPs effectively inhibited PD-1 expression on T-cells to a similar extent to free aPD-1 and efficiently potentiated the cytotoxicity of T-cells against head and neck cancer cells in vitro. Further studies are warranted to assess the potential of this intratumoral administration of aPD-1 nanoformulation in alleviating the toxicity and enhancing the tumor efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Parisa Badiee
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
- UniSA Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| | - Michelle F Maritz
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Nicole Dmochowska
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Edward Cheah
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
- UniSA Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
- UniSA Clinical and Health Sciences, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| |
Collapse
|
17
|
Ganesan N, Ronsmans S, Vanoirbeek J, Hoet PHM. Assessment of Experimental Techniques That Facilitate Human Granuloma Formation in an In Vitro System: A Systematic Review. Cells 2022; 11:cells11050864. [PMID: 35269486 PMCID: PMC8909410 DOI: 10.3390/cells11050864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
The process of granuloma formation is complex, and due to species differences, the validity of animal studies is somewhat questioned. Moreover, the large number of animals needed to observe the different stages of development also raises ethical questions. Therefore, researchers have explored the use of human peripheral blood mononuclear cells (PBMCs), a heterogeneous population of immune cells, in an in vitro model. This review included in vitro studies that focused on exposing PBMCs—from healthy, sensitized, or diseased individuals—to antigens derived from infectious agents—such as mycobacteria or Schistosoma spp.—or inorganic antigens—such as beryllium. The reviewed studies mainly explored how human in vitro granuloma models can contribute towards understanding the pathogenesis of granulomatous diseases, especially during the early stages of granuloma formation. The feasibility of granuloma modelling was thus largely assessed via experimental techniques including (1) granuloma scoring indices (GI), (2) cell surface markers and (3) cytokine secretion profiling. While granuloma scoring showed some similarities between studies, a large variability of culture conditions and endpoints measured have been identified. The lack of any standardization currently impedes the success of a human in vitro granuloma model.
Collapse
Affiliation(s)
- Nirosha Ganesan
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, 3001 Leuven, Belgium; (N.G.); (J.V.)
| | - Steven Ronsmans
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3001 Leuven, Belgium;
| | - Jeroen Vanoirbeek
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, 3001 Leuven, Belgium; (N.G.); (J.V.)
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3001 Leuven, Belgium;
| | - Peter H. M. Hoet
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, 3001 Leuven, Belgium; (N.G.); (J.V.)
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3001 Leuven, Belgium;
- Correspondence:
| |
Collapse
|
18
|
Wu HY, Li KX, Pan WY, Guo MQ, Qiu DZ, He YJ, Li YH, Huang YX. Venetoclax enhances NK cell killing sensitivity of AML cells through the NKG2D/NKG2DL activation pathway. Int Immunopharmacol 2022; 104:108497. [PMID: 34999394 DOI: 10.1016/j.intimp.2021.108497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Venetoclax, a selective B-cell lymphoma-2 (BCL2) inhibitor, has a potential therapeutic effect when combined with demethylating agents in the first-line setting of unfit elderly patients with acute myeloid leukaemia (AML); however, efficacy is still limited in refractory/recurrent AML. Therefore, exploration of a suitable novel treatment scheme is urgently needed.However, combining venetoclax with NK cell-based immunotherapy has not been studied. METHODS The cytotoxicity of NK cell combined with venetoclax was assessed in vitro using flow cytometry. Venetoclax-induced natural killer group 2 member D (NKG2D) ligand (NKG2DL) expression was detected by flow cytometry and western blotting. Mechanisms underlying venetoclax-induced NKG2DL expression were found by GSE127200 analysis and investigated using real-time PCR (Q-PCR) and western blotting. RESULTS Flow cytometric analysis showed that combining venetoclax with NK cells produced synergistic anti-leukaemia effects similar to those of venetoclax + azacitidine. Venetoclax could render AML cell lines and primary AML cells sensitive to NK cell killing by promoting NK cell degranulation, NK-AML cell recognition and NK cell secretion of interferon (IFN)-γ and granzyme B. The synergistic effect resulted from venetoclax-induced NKG2DL upregulation in AML cells and could be undermined by blocking NKG2D on NK cells. This finding suggests that venetoclax enhances NK cell killing activity by activating the NKG2D/NKG2DL ligand-receptor pathway. Furthermore, the nuclear factor-kappa-B (NFKB) signalling pathway was involved in venetoclax-induced NKG2DL upregulation. CONCLUSIONS Collectively, our data confirm that venetoclax combined with NK cells induces synergistic AML cell cytolysis and preliminarily revealed that venetoclax could selectively induce NKG2DLs on AML cells via NFKB signalling pathway.
Collapse
Affiliation(s)
- Hui-Yang Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Ke-Xin Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Wan-Ying Pan
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Meng-Qi Guo
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Dei-Zhi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yan-Jie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yu-Hua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yu-Xian Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China.
| |
Collapse
|
19
|
Dong F, Song X, Xing J, Tang X, Sheng X, Chi H, Zhan W. Immunological characteristics of dendritic cells marker CD83 in flounder (Paralichthys olivaceus). FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100030. [DOI: 10.1016/j.fsirep.2021.100030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
|
20
|
Isolation of primary human B lymphocytes from tonsils compared to blood as alternative source for ex vivo application. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1179:122853. [PMID: 34325309 DOI: 10.1016/j.jchromb.2021.122853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/18/2023]
Abstract
B lymphocytes ('B cells') are components of the human immune system with obvious potential for medical and biotechnological applications. Here, we discuss the isolation of primary human B cells from both juvenile and adult tonsillar material using a two-step procedure based on gradient centrifugation followed by separation on a nylon wool column as alternative to the current gold standard, i.e., negative immunosorting from buffy coats by antibody-coated magnetic beads. We show that the nylon wool separation is a low-cost method well suited to the isolation of large amounts of primary B cells reaching purities ≥ 80%. More importantly, this method allows the preservation of all B cell subsets, while MACS sorting seems to be biased against a certain B cell subtype, namely the CD27+ B cells. Importantly, compared to blood, the excellent recovery yield during purification of tonsillar B cells provides high number of cells, hence increases the number of subsequent experiments feasible with identical cell material, consequently improving comparability of results. The cultivability of the isolated B cells was demonstrated using pokeweed mitogen (PWM) as a stimulatory substance. Our results showed for the first time that the proliferative response of tonsillar B cells to mitogens declines with the age of the donor. Furthermore, we observed that PWM treatment stimulates the proliferation of a dedicated subpopulation and induces some terminal differentiation with ASCs signatures. Taken together this indicates that the proposed isolation procedure preserves the proliferative capability as well as the differentiation capacity of the B cells.
Collapse
|
21
|
Qi C, Wang D, Gong X, Zhou Q, Yue X, Li C, Li Z, Tian G, Zhang B, Wang Q, Wei X, Wu J. Co-Delivery of Curcumin and Capsaicin by Dual-Targeting Liposomes for Inhibition of aHSC-Induced Drug Resistance and Metastasis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16019-16035. [PMID: 33819006 DOI: 10.1021/acsami.0c23137] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Recent research studies have shown that the low survival rate of liver cancer is due to drug resistance and metastasis. In the tumor microenvironment (TME), activated hepatic stellate cells (aHSCs) have been proven to favor the development of liver cancer. Hence, the combination therapy dual-targeting aHSCs and tumor cells might be an effective strategy for treatment of liver cancer. In this study, the novel multifunctional liposomes (CAPS-CUR/GA&Gal-Lip) were prepared for co-delivery of curcumin (CUR) and capsaicin (CAPS), in which glycyrrhetinic acid (GA) and galactose (Gal) were chosen as targeting ligands to modify the liposomes (Lip) for dual-targeting liver cancer. To mimic TME, a novel HSCs+HepG2 (human hepatoma cell line) cocultured model was established for the antitumor effect in vitro. The results showed that, compared to HepG2 cells alone, the cocultured model promoted drug resistance and migration by upregulating the expression of P-glycoprotein (P-gp) and Vimentin, which were effectively inhibited by CAPS-CUR/GA&Gal-Lip. The efficacy of the in vivo antitumor was evaluated by three mice models: subcutaneous H22 (mouse hepatoma cell line) tumor-bearing mice, H22+m-HSC (mouse hepatic stellate cell) tumor-bearing mice, and orthotopic H22 cells-bearing mice. The results showed that CAPS-CUR/GA&Gal-Lip exhibited lesser extracellular matrix (ECM) deposition, lesser tumor angiogenesis, and superior antitumor effect compared with the no- and/or Gal-modified Lip, which was attributed to the simultaneous blocking of the activation of HSCs and inhibition of the metastasis of tumor cells. The dual-targeting method using Lip is thus a potential strategy for liver cancer treatment.
Collapse
Affiliation(s)
- Cuiping Qi
- School of Nursing, Weifang Medical University, Weifang 261053, P. R. China
| | - Di Wang
- School of Nursing, Weifang Medical University, Weifang 261053, P. R. China
| | - Xue Gong
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, P. R. China
| | - Qiyang Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, P. R. China
| | - Xinxin Yue
- School of Nursing, Weifang Medical University, Weifang 261053, P. R. China
| | - Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang 261053, P. R. China
| | - Zhipeng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, P. R. China
| | - Guixiang Tian
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, P. R. China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, P. R. China
| | - Qing Wang
- School of Basic Medicine, Weifang Medical University, Weifang 261053, P. R. China
| | - Xiuhong Wei
- School of Nursing, Weifang Medical University, Weifang 261053, P. R. China
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, P. R. China
| |
Collapse
|
22
|
Cencioni MT, Ali R, Nicholas R, Muraro PA. Defective CD19+CD24hiCD38hi transitional B-cell function in patients with relapsing-remitting MS. Mult Scler 2020; 27:1187-1197. [DOI: 10.1177/1352458520951536] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Multiple sclerosis (MS) is characterized by central nervous system (CNS) infiltration of T and B cells, excess inflammatory cytokine and chemokine production and failure of immune regulation. CD19+CD24hiCD38hi transitional B cells producing interleukin (IL)-10 have been shown to suppress interferon-γ (IFNγ) and tumour necrosis factor-α (TNFα) production by CD4+ T cells and to be dysfunctional in autoimmune arthritis and systemic lupus erythematosus. Objective: We hypothesized that transitional B-cell-dependent immune regulation could be defective in MS and examined their function in healthy subjects and patients with relapsing-remitting multiple sclerosis (RRMS). Methods: A total of 62 healthy donors and 21 RRMS subjects donated peripheral blood for the study. IL-10-producing B cells, IFNγ and TNFα-producing T cells and proliferating T cells were quantified by flow cytometry. Results: In healthy individuals, CD19+CD24hiCD38hi transitional B cells produce more IL-10 than CD19+CD24+CD38+ naive and CD19+CD24hiCD38− memory B cells and are able to suppress CD4+ T-cell proliferation and IFNγ and TNFα-production. In subjects with RRMS, CD19+CD24hiCD38hi transitional B cells produce significantly less IL-10 and to fail to suppress effector T-cell function. Conclusion: CD19+CD24hiCD38hi transitional B cells physiologically represent the most potent regulatory B-cell subset and are functionally defective in patients with RRMS, an abnormality that may contribute to the immune pathological process.
Collapse
Affiliation(s)
- Maria T Cencioni
- Department of Brain Sciences, Imperial College London, London, UK
| | - Rehiana Ali
- Department of Brain Sciences, Imperial College London, London, UK
| | - Richard Nicholas
- Department of Brain Sciences, Imperial College London, London, UK/Imperial College Healthcare NHS Trust, London, UK
| | - Paolo A Muraro
- Wolfson Neuroscience Laboratory, Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
23
|
Tang G, Yuan X, Luo Y, Lin Q, Chen Z, Xing X, Song H, Wu S, Hou H, Yu J, Mao L, Liu W, Wang F, Sun Z. Establishing immune scoring model based on combination of the number, function, and phenotype of lymphocytes. Aging (Albany NY) 2020; 12:9328-9343. [PMID: 32396527 PMCID: PMC7288950 DOI: 10.18632/aging.103208] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/17/2020] [Indexed: 12/31/2022]
Abstract
Background: Quantitatively assessing host immunity remains a challenge in clinical practice. Results: Most parameters in lymphocyte number, function and phenotype were correlated with age. The reference ranges of these parameters were established in four age groups (children, adolescents, adults, and elders). The numbers of CD4+ T cells, CD8+ T cells, B cells, but not NK cells, were negatively correlated with age. However, the function of CD4+ T cells, CD8+ T cells and NK cells was positively correlated with age. The expression of CD28 on T cells gradually decreased with increasing age and was negatively correlated with their function. An opposite phenomenon was observed in the expressions of HLA-DR and CD45RO on T cells. An immune scoring model was established by using 8 parameters (CD4+ T cell number × function, CD28+CD4+ T cell number, HLA-DR+CD4+ T cell number, CD45RO+CD4+ T cell number, CD8+ T cell number × function, CD28+CD8+ T cell number, HLA-DR+CD8+ T cell number, NK cell number × function) from the results of lymphocyte number, function, and phenotype. This immune scoring model showed sensitivities of 70% and 71.4% in determining hyper-immune and hypo-immune status, respectively. Conclusions: An immune scoring model based on combination of lymphocyte number, function, and phenotype shows potential value in quantitatively assessing host immunity. Methods: 261 healthy individuals aged 1 to 82 years were recruited from Tongji Hospital. The number, function, and phenotype of CD4+ T cells, CD8+ T cells and NK cells were simultaneously determined.
Collapse
Affiliation(s)
- Guoxing Tang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Yuan
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Luo
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Lin
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Xing
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijuan Song
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiji Wu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liyan Mao
- Center for Cellular and Molecular Diagnosis, Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Weiyong Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyong Sun
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|