1
|
Gao Y, Wang N, Qi Y, Wang X, Zhang K, Zhang Y, Cao Y, Zang T, Wang T. The aluminum nanoparticle-encircled SQ-in-water emulsions (ANSWE) as a vaccine adjuvant-delivery system (VADS) for developing robust mucosal subunit vaccines. BIOMATERIALS ADVANCES 2025; 166:214076. [PMID: 39490192 DOI: 10.1016/j.bioadv.2024.214076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
The aluminum nanoparticle-encircled squalene (SQ)-in-water emulsions (ANSWE) were engineered as a VADS (vaccine adjuvant-delivery system) using a simple procedure for carrying antigens (Ag) to develop subunit vaccines. In vitro, due to possessing the synergistic adjuvanticity of both AN and SQ, ANSWE were efficiently taken up by APC (antigen-presenting cells) and triggered them to mature and make extra ROS (reactive oxygen species) and multiple cytokines, such as IL-12, IL-1β and IFN-β, which favor balanced Th1/Th2 immunoresponses. Within APC, ANSWE managed lysosome escape and consequently enhanced proteasome activities to facilitate Ag cross-presentation. Mice given twice ovalbumin-ANSWE via intrapulmonary vaccination (IPV) produced high levels of anti-Ag antibodies as well as cytotoxic T lymphocytes, which efficiently erased cells bearing cognate Ag. Thus, ANSWE as a potent VADS may be feasible for developing mucosal subunit vaccines that can elicit comprehensive immunity against infectious diseases, including especially the respiratory infections, and even aggressive cancers.
Collapse
Affiliation(s)
- Yuhao Gao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 420 Jade Road, Hefei, Anhui Province 230601, China
| | - Yuanyuan Qi
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Xiujuan Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Keyi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Yuxi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Yachen Cao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Tairan Zang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
2
|
Wang N, Wang C, Wei C, Chen M, Gao Y, Zhang Y, Wang T. Constructing the cGAMP-Aluminum Nanoparticles as a Vaccine Adjuvant-Delivery System (VADS) for Developing the Efficient Pulmonary COVID-19 Subunit Vaccines. Adv Healthc Mater 2024; 13:e2401650. [PMID: 39319481 DOI: 10.1002/adhm.202401650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/05/2024] [Indexed: 09/26/2024]
Abstract
The cGAMP-aluminum nanoparticles (CAN) are engineered as a vaccine adjuvant-delivery system to carry mixed RBD (receptor-binding domain) of the original severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its new variant for developing bivalent pulmonary coronavirus disease 2019 (COVID-19) vaccines (biRBD-CAN). High phosphophilicity/adsorptivity made intrapulmonary CAN instantly form the pulmonary ingredient-coated CAN (piCAN) to possess biomimetic features enhancing biocompatibility. In vitro biRBD-CAN sparked APCs (antigen-presenting cells) to mature and make extra reactive oxygen species, engendered lysosome escape effects and enhanced proteasome activities. Through activating the intracellular stimulator of interferon genes (STING) and nucleotide-binding domain and leucine-rich repeat and pyrin domain containing proteins 3 (NALP3) inflammasome pathways to exert synergy between cGAMP and AN, biRBD-CAN stimulated APCs to secret cytokines favoring mixed Th1/Th2 immunoresponses. Mice bearing twice intrapulmonary biRBD-CAN produced high levels of mucosal antibodies, the long-lasting systemic antibodies, and potent cytotoxic T lymphocytes which efficiently erased cells displaying cognate epitopes. Notably, biRBD-CAN existed in mouse lungs and different lymph nodes for at least 48 h, unveiling their sustained immunostimulatory activity as the main mechanism underlying the long-lasting immunity and memory. Hamsters bearing twice intrapulmonary biRBD-CAN developed high resistance to pseudoviral challenges performed using different recombinant strains including the ones with distinct SARS-CoV-2-spike mutations. Thus, biRBD-CAN as a broad-spectrum pulmonary COVID-19 vaccine candidate may provide a tool for controlling the emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 420 Jade Road, Hefei, Anhui Province, 230601, China
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Can Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
- Department of Pharmacy, The Second People's Hospital of Lianyungang, 41 Hailian East Road, Lianyungang, Jiangsu Province, 222006, China
| | - Chunliu Wei
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuhao Gao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuxi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| |
Collapse
|
3
|
Olagunju AS, Sardinha AVD, Amarante-Mendes GP. Long-Lasting, Fine-Tuned Anti-Tumor Activity of Recombinant Listeria monocytogenes Vaccine Is Controlled by Pyroptosis and Necroptosis Regulatory and Effector Molecules. Pathogens 2024; 13:828. [PMID: 39452700 PMCID: PMC11510422 DOI: 10.3390/pathogens13100828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
One of the main objectives of developing new anti-cancer vaccine strategies is to effectively induce CD8+ T cell-mediated anti-tumor immunity. Live recombinant vectors, notably Listeria monocytogenes, have been shown to elicit a robust in vivo CD8+ T-cell response in preclinical settings. Significantly, it has been demonstrated that Listeria induces inflammatory/immunogenic cell death mechanisms such as pyroptosis and necroptosis in immune cells that favorably control immunological responses. Therefore, we postulated that the host's response to Listeria-based vectors and the subsequent induction of CD8+ T cell-mediated immunity would be compromised by the lack of regulatory or effector molecules involved in pyroptosis or necroptosis. To test our hypothesis, we used recombinant L. monocytogenes carrying the ovalbumin gene (LM.OVA) to vaccinate wild-type (WT), caspase-1/11-/-, gsdmd-/-, ripk3-/-, and mlkl-/- C57Bl/6 mice. We performed an in vivo cytotoxicity assay to assess the efficacy of OVA-specific CD8+ T lymphocytes in eliminating target cells in wild-type and genetically deficient backgrounds. Furthermore, we evaluated the specific anti-tumor immune response in mice inoculated with the B16F0 and B16F0.OVA melanoma cell lines. Our findings demonstrated that while caspase-1/11 and GSDMD deficiencies interfere with the rapid control of LM.OVA infection, neither of the KOs seems to contribute to the early activation of OVA-specific CTL responses. In contrast, the individual deficiency of each one of these proteins positively impacts the generation of long-lasting effector CD8+ T cells.
Collapse
Affiliation(s)
- Abolaji S. Olagunju
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (A.S.O.); (A.V.D.S.)
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), São Paulo 05508-000, SP, Brazil
| | - Andrew V. D. Sardinha
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (A.S.O.); (A.V.D.S.)
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), São Paulo 05508-000, SP, Brazil
| | - Gustavo P. Amarante-Mendes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (A.S.O.); (A.V.D.S.)
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), São Paulo 05508-000, SP, Brazil
| |
Collapse
|
4
|
Coirada FC, Fernandes ER, Mello LRD, Schuch V, Soares Campos G, Braconi CT, Boscardin SB, Santoro Rosa D. Heterologous DNA Prime- Subunit Protein Boost with Chikungunya Virus E2 Induces Neutralizing Antibodies and Cellular-Mediated Immunity. Int J Mol Sci 2023; 24:10517. [PMID: 37445695 DOI: 10.3390/ijms241310517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Chikungunya virus (CHIKV) has become a significant public health concern due to the increasing number of outbreaks worldwide and the associated comorbidities. Despite substantial efforts, there is no specific treatment or licensed vaccine against CHIKV to date. The E2 glycoprotein of CHIKV is a promising vaccine candidate as it is a major target of neutralizing antibodies during infection. In this study, we evaluated the immunogenicity of two DNA vaccines (a non-targeted and a dendritic cell-targeted vaccine) encoding a consensus sequence of E2CHIKV and a recombinant protein (E2*CHIKV). Mice were immunized with different homologous and heterologous DNAprime-E2* protein boost strategies, and the specific humoral and cellular immune responses were accessed. We found that mice immunized with heterologous non-targeted DNA prime- E2*CHIKV protein boost developed high levels of neutralizing antibodies, as well as specific IFN-γ producing cells and polyfunctional CD4+ and CD8+ T cells. We also identified 14 potential epitopes along the E2CHIKV protein. Furthermore, immunization with recombinant E2*CHIKV combined with the adjuvant AS03 presented the highest humoral response with neutralizing capacity. Finally, we show that the heterologous prime-boost strategy with the non-targeted pVAX-E2 DNA vaccine as the prime followed by E2* protein + AS03 boost is a promising combination to elicit a broad humoral and cellular immune response. Together, our data highlights the importance of E2CHIKV for the development of a CHIKV vaccine.
Collapse
Affiliation(s)
- Fernanda Caroline Coirada
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Edgar Ruz Fernandes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Lucas Rodrigues de Mello
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04044-020, Brazil
| | - Viviane Schuch
- Departamento de Análises Clínicas e Toxicológicas, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Gúbio Soares Campos
- Laboratório de Virologia, Universidade Federal da Bahia (UFBA), Salvador 40110-909, Brazil
| | - Carla Torres Braconi
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| |
Collapse
|
5
|
Gao Y, Wang W, Yang Y, Zhao Q, Yang C, Jia X, Liu Y, Zhou M, Zeng W, Huang X, Chiu S, Jin T, Wu X. Developing Next-Generation Protein-Based Vaccines Using High-Affinity Glycan Ligand-Decorated Glyconanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204598. [PMID: 36398611 PMCID: PMC9839878 DOI: 10.1002/advs.202204598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Major diseases, such as cancer and COVID-19, are frightening global health problems, and sustained action is necessary to develop vaccines. Here, for the first time, ethoxy acetalated dextran nanoparticles (Ace-Dex-NPs) are functionalized with 9-N-(4H-thieno[3,2-c]chromene-2-carbamoyl)-Siaα2-3Galβ1-4GlcNAc (TCC Sia-LacNAc) targeting macrophages as a universal vaccine design platform. First, azide-containing oxidized Ace-Dex-NPs are synthesized. After the NPs are conjugated with ovalbumin (OVA) and resiquimod (Rd), they are coupled to TCC Sia-LacNAc-DBCO to produce TCC Sia-Ace-Dex-OVA-Rd, which induce a potent, long-lasting OVA-specific cytotoxic T-lymphocyte (CTL) response and high anti-OVA IgG, providing mice with superior protection against tumors. Next, this strategy is exploited to develop vaccines against infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein is the main target for neutralizing antibodies. The TCC Sia-Ace-Dex platform is preferentially used for designing an RBD-based vaccine. Strikingly, the synthetic TCC Sia-Ace-Dex-RBD-Rd elicited potent RBD-neutralizing antibodies against live SARS-CoV-2 infected Vero E6 cells. To develop a universal SARS-CoV-2 vaccine, the TCC Sia-Ace-Dex-N-Rd vaccine carrying SARS-CoV-2 nucleocapsid protein (N) is also prepared, which is highly conserved among SARS-CoV-2 and its variants of concern (VOCs), including Omicron (BA.1 to BA.5); this vaccine can trigger strong N-specific CTL responses against target cells infected with SARS-CoV-2 and its VOCs.
Collapse
Affiliation(s)
- Yanan Gao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Wei Wang
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Yunru Yang
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Qingyu Zhao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Chendong Yang
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
| | - Xiaoying Jia
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Yang Liu
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
| | - Minmin Zhou
- State Key Laboratory of VirologyWuhan Institute of VirologyCenter for Biosafety Mega‐ScienceChinese Academy of SciencesWuhan430071China
- University of the Chinese Academy of SciencesBeijing100049China
| | - Weihong Zeng
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical EngineeringInstitute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMichigan48824United States
| | - Sandra Chiu
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Tengchuan Jin
- Department of Basic Medical SciencesDivision of Molecular MedicineDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Xuanjun Wu
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐based MedicineShandong UniversityQingdaoShandong266237China
- Suzhou Research InstituteShandong UniversitySuzhouJiangsu215123China
| |
Collapse
|
6
|
Jiang S, Wu S, Zhao G, He Y, Guo X, Zhang Z, Hou J, Ding Y, Cheng A, Wang B. Identification of a promiscuous conserved CTL epitope within the SARS-CoV-2 spike protein. Emerg Microbes Infect 2022; 11:730-740. [PMID: 35171086 PMCID: PMC8890520 DOI: 10.1080/22221751.2022.2043727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The COVID-19 disease caused by infection with SARS-CoV-2 and its variants is devastating to the global public health and economy. To date, over a hundred COVID-19 vaccines are known to be under development, and the few that have been approved to fight the disease are using the spike protein as the primary target antigen. Although virus-neutralizing epitopes are mainly located within the RBD of the spike protein, the presence of T cell epitopes, particularly the CTL epitopes that are likely to be needed for killing infected cells, has received comparatively little attention. This study predicted several potential T cell epitopes with web-based analytic tools and narrowed them down from several potential MHC-I and MHC-II epitopes by ELIspot and cytolytic assays to a conserved MHC-I epitope. The epitope is highly conserved in current viral variants and compatible with a presentation by most HLA alleles worldwide. In conclusion, we identified a CTL epitope suitable for evaluating the CD8+ T cell-mediated cellular response and potentially for addition into future COVID-19 vaccine candidates to maximize CTL responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Sheng Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College(SHMC), Fudan University.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuting Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College(SHMC), Fudan University
| | - Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Yue He
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | | | - Zhiyu Zhang
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Jiawang Hou
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Yuan Ding
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Alex Cheng
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College(SHMC), Fudan University.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Advaccine Biopharmaceutics (Suzhou) Co. LTD, Jiangsu Province, China. dColby College, Waterville, Maine, USA
| |
Collapse
|
7
|
Gao Y, Zhao Q, Dong H, Xiao M, Huang X, Wu X. Developing Acid-Responsive Glyco-Nanoplatform Based Vaccines for Enhanced Cytotoxic T-lymphocyte Responses Against Cancer and SARS-CoV-2. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2105059. [PMID: 34512228 PMCID: PMC8420391 DOI: 10.1002/adfm.202105059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/04/2021] [Indexed: 05/05/2023]
Abstract
Cytotoxic T-lymphocytes (CTLs) are central for eliciting protective immunity against malignancies and infectious diseases. Here, for the first time, partially oxidized acetalated dextran nanoparticles (Ox-AcDEX NPs) with an average diameter of 100 nm are fabricated as a general platform for vaccine delivery. To develop effective anticancer vaccines, Ox-AcDEX NPs are conjugated with a representative CTL peptide epitope (CTLp) from human mucin-1 (MUC1) with the sequence of TSAPDTRPAP (referred to as Mp1) and an immune-enhancing adjuvant R837 (referred to as R) via imine bond formation affording AcDEX-(imine)-Mp1-R NPs. Administration of AcDEX-(imine)-Mp1-R NPs results in robust and long-lasting anti-MUC1 CTL immune responses, which provides mice with superior protection from the tumor. To verify its universality, this nanoplatform is also exploited to deliver epitopes from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to prevent coronavirus disease 2019 (COVID-19). By conjugating Ox-AcDEX NPs with the potential CTL epitope of SARS-CoV-2 (referred to as Sp) and R837, AcDEX-(imine)-Sp-R NPs are fabricated for anti-SARS-CoV-2 vaccine candidates. Several epitopes potentially contributing to the induction of potent and protective anti-SARS-CoV-2 CTL responses are examined and discussed. Collectively, these findings shed light on the universal use of Ox-AcDEX NPs to deliver both tumor-associated and virus-associated epitopes.
Collapse
Affiliation(s)
- Yanan Gao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based MedicineShandong UniversityQingdaoShandong266237China
| | - Qingyu Zhao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based MedicineShandong UniversityQingdaoShandong266237China
| | - Huiling Dong
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based MedicineShandong UniversityQingdaoShandong266237China
| | - Min Xiao
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based MedicineShandong UniversityQingdaoShandong266237China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical EngineeringInstitute for Quantitative Health Science and EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Xuanjun Wu
- National Glycoengineering Research CenterShandong Key Laboratory of Carbohydrate Chemistry and GlycobiologyNMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate‐Based MedicineShandong UniversityQingdaoShandong266237China
- Suzhou Research InstituteShandong UniversitySuzhouJiangsu215123China
| |
Collapse
|
8
|
Hernández-Torres M, Silva do Nascimento R, Rebouças MC, Cassado A, Matteucci KC, D'Império-Lima MR, Vasconcelos JRC, Bortoluci KR, Alvarez JM, Amarante-Mendes GP. Absence of Bim sensitizes mice to experimental Trypanosoma cruzi infection. Cell Death Dis 2021; 12:692. [PMID: 34247195 PMCID: PMC8272718 DOI: 10.1038/s41419-021-03964-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Chagas disease is a life-threatening disorder caused by the protozoan parasite Trypanosoma cruzi. Parasite-specific antibodies, CD8+ T cells, as well as IFN-γ and nitric oxide (NO) are key elements of the adaptive and innate immunity against the extracellular and intracellular forms of the parasite. Bim is a potent pro-apoptotic member of the Bcl-2 family implicated in different aspects of the immune regulation, such as negative selection of self-reactive thymocytes and elimination of antigen-specific T cells at the end of an immune response. Interestingly, the role of Bim during infections remains largely unidentified. To explore the role of Bim in Chagas disease, we infected WT, Bim+/-, Bim-/- mice with trypomastigotes forms of the Y strain of T. cruzi. Strikingly, our data revealed that Bim-/- mice exhibit a delay in the development of parasitemia followed by a deficiency in the control of parasite load in the bloodstream and a decreased survival compared to WT and Bim+/- mice. At the peak of parasitemia, peritoneal macrophages of Bim-/- mice exhibit decreased NO production, which correlated with a decrease in the pro-inflammatory Small Peritoneal Macrophage (SPM) subset. A similar reduction in NO secretion, as well as in the pro-inflammatory cytokines IFN-γ and IL-6, was also observed in Bim-/- splenocytes. Moreover, an impaired anti-T. cruzi CD8+ T-cell response was found in Bim-/- mice at this time point. Taken together, our results suggest that these alterations may contribute to the establishment of a delayed yet enlarged parasitic load observed at day 9 after infection of Bim-/- mice and place Bim as an important protein in the control of T. cruzi infections.
Collapse
Affiliation(s)
- Marcela Hernández-Torres
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), São Paulo, Brazil
| | | | - Monica Cardozo Rebouças
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), São Paulo, Brazil
| | - Alexandra Cassado
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Kely Catarine Matteucci
- Centro de Terapia Celular e Molecular - CTCMol - Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - José Ronnie C Vasconcelos
- Centro de Terapia Celular e Molecular - CTCMol - Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Karina R Bortoluci
- Centro de Terapia Celular e Molecular - CTCMol - Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Maria Alvarez
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Gustavo P Amarante-Mendes
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil.
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT-iii), São Paulo, Brazil.
| |
Collapse
|
9
|
Gao Y, Zhao Q, Xiao M, Huang X, Wu X. A versatile photothermal vaccine based on acid-responsive glyco-nanoplatform for synergistic therapy of cancer. Biomaterials 2021; 273:120792. [PMID: 33872856 DOI: 10.1016/j.biomaterials.2021.120792] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/24/2021] [Accepted: 03/29/2021] [Indexed: 01/10/2023]
Abstract
The race is on for therapeutic agents that stop cancer. An effective vaccine offers a safe and promising approach for cancer immunotherapy. However, substantial barriers to immunotherapy in cancer vaccines include the low immunogenicity of cancer antigens and the immunosuppression commonly present in solid tumors, resulting in significant challenges for developing a clinically effective cancer vaccine. Here, the state of the art of synergistic therapy, which includes the photothermal effect combined with immunotherapy, was investigated to target tumors. For the first time, indocyanine green (ICG, referred to as I), imiquimod (R837, referred to as R) and a foreign cytotoxic T lymphocyte antigen peptide (CTL-Ap, referred to as Ap) with the sequence of SIINFEKL from ovalbumin (OVA) were encapsulated by acetalated dextran (AcDEX) to form nanoparticles (NPs) averaging 92 nm in diameter as an immunogen. Administration of the resulting multifunctional vaccine I-R-Ap-AcDEX NPs enhanced antitumor cytotoxic T lymphocyte (CTL) immunotherapy. On the one hand, subcutaneous immunization of the NPs allows foreign Ap to enter the major histocompatibility complex class I (MHC-I) cross-presentation pathway of antigen-presenting cells, thereby presenting Ap and eliciting high levels of Ap-specific CTLs. On the other hand, intratumor/intravenous injections of the NPs allow foreign Ap to enter tumor cells and present Ap through the MHC-I cross-presentation pathway. Ap-specific CTLs can kill Ap-presented tumor cells. Furthermore, the NPs generated near-infrared laser triggered the photothermal killing of tumor cells. To our knowledge, this is the first report of AcDEX NPs in antitumor photothermal therapy. Strikingly, systemic administration of the I-R-Ap-AcDEX NPs combined with near-infrared laser irradiation allowed for complete protection to mice from the tumors when applied to two non-OVA tumor models. This quite impressive result displays the great promise of synergistic therapy by the vaccine I-R-Ap-AcDEX NPs, an approach that harnesses the photothermal effect to boost antitumor immunotherapy.
Collapse
Affiliation(s)
- Yanan Gao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China
| | - Qingyu Zhao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China
| | - Min Xiao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, United States
| | - Xuanjun Wu
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong, 266237, China; Suzhou Research Institute, Shandong University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
10
|
Loo Yau H, Bell E, Ettayebi I, de Almeida FC, Boukhaled GM, Shen SY, Allard D, Morancho B, Marhon SA, Ishak CA, Gonzaga IM, da Silva Medina T, Singhania R, Chakravarthy A, Chen R, Mehdipour P, Pommey S, Klein C, Amarante-Mendes GP, Roulois D, Arribas J, Stagg J, Brooks DG, De Carvalho DD. DNA hypomethylating agents increase activation and cytolytic activity of CD8 + T cells. Mol Cell 2021; 81:1469-1483.e8. [PMID: 33609448 DOI: 10.1016/j.molcel.2021.01.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/16/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Abstract
We demonstrate that DNA hypomethylating agent (HMA) treatment can directly modulate the anti-tumor response and effector function of CD8+ T cells. In vivo HMA treatment promotes CD8+ T cell tumor infiltration and suppresses tumor growth via CD8+ T cell-dependent activity. Ex vivo, HMAs enhance primary human CD8+ T cell activation markers, effector cytokine production, and anti-tumor cytolytic activity. Epigenomic and transcriptomic profiling shows that HMAs vastly regulate T cell activation-related transcriptional networks, culminating with over-activation of NFATc1 short isoforms. Mechanistically, demethylation of an intragenic CpG island immediately downstream to the 3' UTR of the short isoform was associated with antisense transcription and alternative polyadenylation of NFATc1 short isoforms. High-dimensional single-cell mass cytometry analyses reveal a selective effect of HMAs on a subset of human CD8+ T cell subpopulations, increasing both the number and abundance of a granzyme Bhigh, perforinhigh effector subpopulation. Overall, our findings support the use of HMAs as a therapeutic strategy to boost anti-tumor immune response.
Collapse
Affiliation(s)
- Helen Loo Yau
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Emma Bell
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Ilias Ettayebi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Felipe Campos de Almeida
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; Instituto de Investigação em Imunologia, Institutos Nacionais de Ciência e Tecnologia (INCT-iii), São Paulo 05403-900, Brazil
| | - Giselle M Boukhaled
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shu Yi Shen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - David Allard
- Centre de recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Beatriz Morancho
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO) and CIBERONC, 08035 Barcelona, Spain
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Charles A Ishak
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Isabela M Gonzaga
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Tiago da Silva Medina
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Translational Immuno-oncology Laboratory, A.C. Camargo Cancer Center, São Paulo 01509-001, Brazil
| | - Rajat Singhania
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Ankur Chakravarthy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Raymond Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Parinaz Mehdipour
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Sandra Pommey
- Centre de recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Gustavo P Amarante-Mendes
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; Instituto de Investigação em Imunologia, Institutos Nacionais de Ciência e Tecnologia (INCT-iii), São Paulo 05403-900, Brazil
| | - David Roulois
- UMR U1236, INSERM, Université de Rennes 1, EFS, 35000 Rennes, France
| | - Joaquín Arribas
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO) and CIBERONC, 08035 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - John Stagg
- Centre de recherche du Centre Hospitalier de l'Université de Montréal et Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada; Faculté de Pharmacie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - David G Brooks
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
11
|
Lang S, Tan Z, Wu X, Huang X. Synthesis of Carboxy-Dimethylmaleic Amide Linked Polymer Conjugate Based Ultra-pH-sensitive Nanoparticles for Enhanced Antitumor Immunotherapy. ACS Macro Lett 2020; 9:1693-1699. [PMID: 33224624 DOI: 10.1021/acsmacrolett.0c00755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are an important tool for anticancer immunotherapy. To elicit powerful CTL activities, ultra-pH-sensitive nanoparticles (NPs) based on methoxy poly(ethylene glycol)-b-[poly(diisopropylamino)ethyl methacrylate] have been synthesized as a vaccine delivery platform. A representative CTL epitope, ovalbumin (OVA) peptide antigen, was covalently conjugated to the polymer backbone through an acid responsive carboxy-dimethylmaleic amide linker (CDM) resulting in polymer P-CDM-OVA. Interestingly, while the P-CDM-OVA released OVA peptide slowly in a pH 6.4 buffer, the addition of bovine serum albumin (BSA) mimicking proteins encountered in a cellular and/or in vivo environment significantly accelerated the release process. Successful cell surface presentation of OVA was observed when P-CDM-OVA based ultra-pH-sensitive particles were incubated with antigen presenting cells. These P-CDM-OVA NPs greatly enhanced CTL responses in vivo compared to the free peptide or the previously reported acetalated dextran particles encapsulating OVA. The P-CDM was also investigated for adjuvant conjugation, and the coadministration of P-CDM-OVA and the P-CDM-adjuvant conjugate NPs further improved CTL responses in vivo and effectively reduced tumor growth in mice. Thus, the CDM linked polymer presents a promising platform for anticancer immunotherapy.
Collapse
|
12
|
Chauhan P, Hu S, Prasad S, Sheng WS, Lokensgard JR. Programmed death ligand-1 induction restrains the cytotoxic T lymphocyte response against microglia. Glia 2020; 69:858-871. [PMID: 33128485 DOI: 10.1002/glia.23932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/11/2022]
Abstract
Microglial cells are the main reservoir for HIV-1 within the brain and potential exists for negative immune checkpoint blockade therapies to purge this viral reservoir. Here, we investigated cytolytic responses of CD8+ T lymphocytes against microglia loaded with peptide epitopes. Initially, flow cytometric analysis demonstrated efficient killing of HIV-1 p24 AI9 or YI9 peptide-loaded splenocytes in MHC-matched recipients. Cytolytic killing of microglia was first demonstrated using ovalbumin (OVA) as a model antigen for in vitro cytotoxic T lymphocyte (CTL) assays. Peptide-loaded primary microglia obtained from programmed death ligand (PD-L) 1 knockout (KO) animals showed significantly more killing than cells from wild-type (WT) animals when co-cultured with activated CD8+ T-cells isolated from rAd5-OVA primed animals. Moreover, when peptide loaded-microglial cells from WT animals were treated with neutralizing α-PD-L1 Ab, significantly more killing was observed compared to either untreated or IgG isotype-treated cells. Most importantly, significantly increased in vivo killing of HIV-1 p24 YI9 peptide-loaded microglia from PD-L1 KO animals, as well as AI9 peptide-loaded BALB/c microglial cells treated with α-PD-L1, was observed within brains of rAd5-p24 primed-CNS boosted C57BL/6 or BALB/c mice, respectively. Finally, ex vivo responses of brain CD8+ T-cells in response to AI9 stimulation showed significantly increased IFN-γ and IL-2 production when treated with α-PD-1 Abs. Greater proliferation of CD8+ T-cells from the brain was also observed following blockade. Taken together, these studies demonstrate that PD-L1 induction on microglia restrains CTL responses and indicate that immune checkpoint blockade targeting this pathway may be beneficial in clearing viral brain reservoirs.
Collapse
Affiliation(s)
- Priyanka Chauhan
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shuxian Hu
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sujata Prasad
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wen S Sheng
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - James R Lokensgard
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
13
|
Rana A, de Almeida FC, Paico Montero HA, Gonzales Carazas MM, Bortoluci KR, Sad S, Amarante-Mendes GP. RIPK3 and Caspase-1/11 Are Necessary for Optimal Antigen-Specific CD8 T Cell Response Elicited by Genetically Modified Listeria monocytogenes. Front Immunol 2020; 11:536. [PMID: 32328060 PMCID: PMC7160319 DOI: 10.3389/fimmu.2020.00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 03/09/2020] [Indexed: 11/26/2022] Open
Abstract
Efficient induction of effector and long-term protective antigen-specific CD8+ T memory response by vaccination is essential to eliminate malignant and pathogen-infected cells. Intracellular infectious bacteria, including Listeria monocytogenes, have been considered potent vectors to carry multiple therapeutic proteins and generate antigen-specific CD8+ T cell responses. Although the role of molecules involved in inflammatory cell death pathways, such as necroptosis (RIPK3-mediated) and pyroptosis (Caspase-1/11-mediated), as effectors of immune response against intracellular bacteria are relatively well understood, their contribution to the adjuvant effect of recombinant bacterial vectors in the context of antigen-specific CD8+ T cell response remained obscure. Therefore, we evaluated the impact of RIPK3 and Caspase-1/11 (Casp-1/11) individual and combined deficiencies on the modulation of antigen-specific CD8+ T cell response during vaccination of mice with ovalbumin-expressing L. monocytogenes (LM-OVA). We observed that Casp-1/11 but not RIPK3 deficiency negatively impacts the capacity of mice to clear LM-OVA. Importantly, both RIPK3 and Casp-1/11 are necessary for optimal LM-OVA-mediated antigen-specific CD8+ T cell response, as measured by in vivo antigen-specific CD8+ T cell proliferation, target cell elimination, and cytokine production. Furthermore, Casp-1/11 and Casp-1/11/RIPK3 combined deficiencies restrict the early initiation of antigen-specific CD8+ T cell memory response. Taken together, our findings demonstrate that RIPK3 and Casp-1/11 influence the quality of CD8+ T cell responses induced by recombinant L. monocytogenes vectors.
Collapse
Affiliation(s)
- Aamir Rana
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Felipe Campos de Almeida
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | | | | | - Karina R Bortoluci
- Departamento de Ciências Biológicas, Centro de Terapia Celular e Molecular (CTC-Mol), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Gustavo P Amarante-Mendes
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| |
Collapse
|
14
|
Sorgi S, Bonezi V, Dominguez MR, Gimenez AM, Dobrescu I, Boscardin S, Nakaya HI, Bargieri DY, Soares IS, Silveira ELV. São Paulo School of Advanced Sciences on Vaccines: an overview. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190061. [PMID: 32362926 PMCID: PMC7187638 DOI: 10.1590/1678-9199-jvatitd-2019-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/21/2020] [Indexed: 01/08/2023] Open
Abstract
Two years ago, we held an exciting event entitled the São Paulo School of Advanced Sciences on Vaccines (SPSASV). Sixty-eight Ph.D. students, postdoctoral fellows and independent researchers from 37 different countries met at the Mendes Plaza Hotel located in the city of Santos, SP - Brazil to discuss the challenges and the new frontiers of vaccinology. The SPSASV provided a critical and comprehensive view of vaccine research from basics to the current state-of-the-art techniques performed worldwide. For 10 days, we discussed all the aspects of vaccine development in 36 lectures, 53 oral presentations and 2 poster sessions. At the end of the course, participants were further encouraged to present a model of a grant proposal related to vaccine development against individual pathogens. Among the targeted pathogens were viruses (Chikungunya, HIV, RSV, and Influenza), bacteria (Mycobacterium tuberculosis and Streptococcus pyogenes), parasites (Plasmodium falciparum or Plasmodium vivax), and the worm Strongyloides stercoralis. This report highlights some of the knowledge shared at the SPSASV.
Collapse
Affiliation(s)
- Sara Sorgi
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Dipartimento di Biotecnologie Mediche, Universita’ degli Studi di Siena, Siena, Italia
| | - Vivian Bonezi
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Mariana R. Dominguez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Alba Marina Gimenez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Irina Dobrescu
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Silvia Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Daniel Y. Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Irene S. Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Eduardo L. V. Silveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
15
|
Belizário J, Destro Rodrigues MF. Checkpoint inhibitor blockade and epigenetic reprogrammability in CD8 + T-cell activation and exhaustion. Ther Adv Vaccines Immunother 2020; 8:2515135520904238. [PMID: 32206744 PMCID: PMC7074507 DOI: 10.1177/2515135520904238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 12/19/2019] [Indexed: 11/17/2022] Open
Abstract
CD8+ T-cell exhaustion is a dysfunctional state that is regulated through the expression of inhibitory checkpoint receptor genes including the cytotoxic T-lymphocyte–associated antigen 4, programmed death 1, and DNA methylation of effector genes interferon-γ, perforin, and granzyme B. Different strategies have been used to reverse T-cell exhaustion, which is an adverse event of checkpoint inhibitor blockade. Here, we present the mechanisms by which DNA methyltransferase inhibitors and Simian virus 40 large T antigen through viral mimicry can promote the reversion of exhausted CD8+ T cells. We examine how these pharmacological strategies can work together to improve the clinical efficacy of immunotherapies.
Collapse
Affiliation(s)
- José Belizário
- Department of Pharmacology, Institute Biomedical Sciences of the University of Sao Paulo, Avenida Lineu Prestes, 1524, São Paulo, CEP 05508-900, Brazil
| | | |
Collapse
|
16
|
Immune-orthogonal orthologues of AAV capsids and of Cas9 circumvent the immune response to the administration of gene therapy. Nat Biomed Eng 2019; 3:806-816. [PMID: 31332341 PMCID: PMC6783354 DOI: 10.1038/s41551-019-0431-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 06/16/2019] [Indexed: 12/19/2022]
Abstract
Protein-based therapeutics can activate the adaptive immune system and lead to the production of neutralizing antibodies and to cytotoxic-T-cell-mediated clearance of the treated cells. Here, we show that the sequential use of immune-orthogonal orthologues of the CRISPR-associated protein 9 (Cas9) and of adeno-associated viruses (AAVs) eludes adaptive immune responses and enables effective gene editing from repeated dosing. We compared total sequence similarities and predicted binding strengths to class-I and class-II major-histocompatibility-complex proteins for 284 DNA-targeting and 84 RNA-targeting CRISPR effectors, and for 167 AAV VP1-capsid-protein orthologues. We predict the absence of cross-reactive immune responses for 79% of the DNA-targeting Cas orthologs, which we validate for three Cas9 orthologs in mice, yet anticipate broad immune cross-reactivity among the AAV serotypes. We also show that efficacious in vivo gene editing is uncompromised when using multiple dosing with orthologues of AAVs and Cas9 in mice previously immunized against the AAV vector and the Cas9 payload. Multiple dosing with protein orthologues may allow for sequential regimens of protein therapeutics that circumvent pre-existing immunity or induced immunity.
Collapse
|
17
|
Xia Y, Xie Y, Yu Z, Xiao H, Jiang G, Zhou X, Yang Y, Li X, Zhao M, Li L, Zheng M, Han S, Zong Z, Meng X, Deng H, Ye H, Fa Y, Wu H, Oldfield E, Hu X, Liu W, Shi Y, Zhang Y. The Mevalonate Pathway Is a Druggable Target for Vaccine Adjuvant Discovery. Cell 2018; 175:1059-1073.e21. [PMID: 30270039 DOI: 10.1016/j.cell.2018.08.070] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 01/02/2023]
Abstract
Motivated by the clinical observation that interruption of the mevalonate pathway stimulates immune responses, we hypothesized that this pathway may function as a druggable target for vaccine adjuvant discovery. We found that lipophilic statin drugs and rationally designed bisphosphonates that target three distinct enzymes in the mevalonate pathway have potent adjuvant activities in mice and cynomolgus monkeys. These inhibitors function independently of conventional "danger sensing." Instead, they inhibit the geranylgeranylation of small GTPases, including Rab5 in antigen-presenting cells, resulting in arrested endosomal maturation, prolonged antigen retention, enhanced antigen presentation, and T cell activation. Additionally, inhibiting the mevalonate pathway enhances antigen-specific anti-tumor immunity, inducing both Th1 and cytolytic T cell responses. As demonstrated in multiple mouse cancer models, the mevalonate pathway inhibitors are robust for cancer vaccinations and synergize with anti-PD-1 antibodies. Our research thus defines the mevalonate pathway as a druggable target for vaccine adjuvants and cancer immunotherapies.
Collapse
Affiliation(s)
- Yun Xia
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Yonghua Xie
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Zhengsen Yu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Hongying Xiao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Guimei Jiang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Xiaoying Zhou
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Yunyun Yang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Xin Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Meng Zhao
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Liping Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Mingke Zheng
- Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Shuai Han
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China
| | - Zhaoyun Zong
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Huahu Ye
- Laboratory Animal Center, Academy of Military Medical Sciences, 100071 Beijing, China
| | - Yunzhi Fa
- Laboratory Animal Center, Academy of Military Medical Sciences, 100071 Beijing, China
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, 100850 Beijing, China
| | - Eric Oldfield
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiaoyu Hu
- Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China; Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China.
| | - Yan Shi
- Institute for Immunology and School of Medicine, Tsinghua University, 100084 Beijing, China; Institute Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, AB, Canada.
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, 100084 Beijing, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, 100084 Beijing, China; Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041 Sichuan, China.
| |
Collapse
|
18
|
Proteomic and functional analysis identifies galectin-1 as a novel regulatory component of the cytotoxic granule machinery. Cell Death Dis 2017; 8:e3176. [PMID: 29215607 PMCID: PMC5827204 DOI: 10.1038/cddis.2017.506] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 11/25/2022]
Abstract
Secretory granules released by cytotoxic T lymphocytes (CTLs) are powerful weapons against intracellular microbes and tumor cells. Despite significant progress, there is still limited information on the molecular mechanisms implicated in target-driven degranulation, effector cell survival and composition and structure of the lytic granules. Here, using a proteomic approach we identified a panel of putative cytotoxic granule proteins, including some already known granule constituents and novel proteins that contribute to regulate the CTL lytic machinery. Particularly, we identified galectin-1 (Gal1), an endogenous immune regulatory lectin, as an integral component of the secretory granule machinery and unveil the unexpected function of this lectin in regulating CTL killing activity. Mechanistic studies revealed the ability of Gal1 to control the non-secretory lytic pathway by influencing Fas–Fas ligand interactions. This study offers new insights on the composition of the cytotoxic granule machinery, highlighting the dynamic cross talk between secretory and non-secretory pathways in controlling CTL lytic function.
Collapse
|
19
|
Wang N, Zhen Y, Jin Y, Wang X, Li N, Jiang S, Wang T. Combining different types of multifunctional liposomes loaded with ammonium bicarbonate to fabricate microneedle arrays as a vaginal mucosal vaccine adjuvant-dual delivery system (VADDS). J Control Release 2016; 246:12-29. [PMID: 27986552 DOI: 10.1016/j.jconrel.2016.12.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 12/01/2016] [Accepted: 12/09/2016] [Indexed: 01/09/2023]
Abstract
To develop effective mucosal vaccines, two types of multifunctional liposomes, the mannosylated lipid A-liposomes (MLLs) with a size of 200nm and the stealth lipid A-liposomes (SLLs) of 50nm, both loaded with a model antigen and NH4HCO3, were fabricated together into microneedles, forming the proSLL/MLL-constituted microneedle array (proSMMA), which upon rehydration dissolved rapidly recovering the initial MLLs and SLLs. Mice vaccinated with proSMMAs by vaginal mucosa patching other than conventional intradermal administration established robust antigen-specific humoral and cellular immunity at both systemic and mucosal levels, especially, in the reproductive and intestinal ducts. Further exploration demonstrated that the MLLs reconstituted from the administered proSMMAs were mostly taken up by vaginal mucosal dendritic cells, whereas the recovered SLLs trafficked directly to draining lymph nodes wherein to be picked up by macrophages. Moreover, the antigens delivered by either liposomes were also cross-presented for MHC-I displaying by APCs thanks to lysosome escape and ROS (reactive oxygen species) stimulation, both of which occurred when lysosomal acidifying the liposome-released NH4HCO3 into CO2 and NH4+/NH3 to rupture lysosomes by gas expansion and to cause ROS production by excessive ammonia induction, resulting in a mixed Th1/Th2 type response which was also promoted by liposomal lipid A via activation of TLR4. In addition, vaginal vaccination of the engineered HSV2 antigen gD-loaded proSMMAs successfully protected mice from the virus challenge. Thus, the proSMMAs are in fact a vaccine adjuvant-dual delivery system capable of eliciting robust humoral and cellular immunity against the invading pathogens, especially, the sexually transmitted ones.
Collapse
Affiliation(s)
- Ning Wang
- School of Biological and Medical Engineering, Hefei University of Technology, 193 Tun Brook Road, Hefei, Anhui Province 230009, China
| | - Yuanyuan Zhen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Xueting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ning Li
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Shaohong Jiang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
20
|
Teixeira LH, Tararam CA, Lasaro MO, Camacho AGA, Ersching J, Leal MT, Herrera S, Bruna-Romero O, Soares IS, Nussenzweig RS, Ertl HCJ, Nussenzweig V, Rodrigues MM. Immunogenicity of a prime-boost vaccine containing the circumsporozoite proteins of Plasmodium vivax in rodents. Infect Immun 2014; 82:793-807. [PMID: 24478093 PMCID: PMC3911365 DOI: 10.1128/iai.01410-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 11/30/2013] [Indexed: 02/03/2023] Open
Abstract
Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.
Collapse
Affiliation(s)
- Lais H. Teixeira
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Cibele A. Tararam
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
| | | | - Ariane G. A. Camacho
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Jonatan Ersching
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Monica T. Leal
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
| | | | - Oscar Bruna-Romero
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Irene S. Soares
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ruth S. Nussenzweig
- Michael Heidelberger Division, Department of Pathology, New York University School of Medicine, New York, New York, USA
| | | | - Victor Nussenzweig
- Michael Heidelberger Division, Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Mauricio M. Rodrigues
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil
| |
Collapse
|
21
|
Martin SJ, Henry CM. Distinguishing between apoptosis, necrosis, necroptosis and other cell death modalities. Methods 2014; 61:87-9. [PMID: 23768793 DOI: 10.1016/j.ymeth.2013.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|