1
|
Li Z, Shao Y, Liu X, Wan X, Xiong P, Wang L, Yuan J. Steap3 is a key node in regulating the phagosome escape of Listeria monocytogenes. Mol Immunol 2025; 182:96-106. [PMID: 40252499 DOI: 10.1016/j.molimm.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Listeria monocytogenes, a foodborne pathogen, poses a significant threat to human health due to its high mortality rate and increasing antibiotic resistance. This study investigates the role of Steap3 in regulating the early phagosomal escape of Listeria monocytogenes. We found that Steap3 expression is downregulated in dendritic and intestinal epithelial cells following infection, and its deficiency exacerbates bacterial proliferation both in vitro and in vivo. Mechanistically, Steap3 interacts with Gm2a and Sting to inhibit Listeria monocytogenes infection. Our results highlight Steap3 as a key regulator in dendritic and intestinal epithelial cells' defense against Listeria monocytogenes infection, suggesting the Steap3-STING/Gm2a axis is a potential therapeutic target for listeriosis. This study provides valuable insights into the molecular mechanisms underlying Listeria monocytogenes pathogenesis and host immune response, offering new directions for developing anti-Listeria monocytogenes therapies.
Collapse
Affiliation(s)
- Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Yan Shao
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiao Liu
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Xiaoyuan Wan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Pei Xiong
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China.
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China; Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Department of Infectious Diseases, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400016, China; School of Chemistry and Chemical Engineering, Jinggangshan University, Ji'an, Jiangxi 343009, China.
| |
Collapse
|
2
|
Li L, Yang Z, Liu H, He Z, Wen Z, Chen H, Zhang Z, Liu Z, Fan X, Liu L, Chen Y. Exaggerated Lung Inflammation Induced by Lung-Targeted mRNA-LNP Dampens Vaccines against Tuberculosis. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40378077 DOI: 10.1021/acsami.5c03743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
The challenges in developing a tuberculosis (TB) vaccine stem from the complex life cycle of Mycobacterium tuberculosis (Mtb) and various bacterial proteins encoded by approximately 4000 genes. mRNA is easy to design and can accommodate multiple antigens, suggesting that it may be an effective TB vaccine technology. Here, we designed an mRNA encoding Ag85B and ESAT6 that was delivered by lung targeted lipid nanoparticles (LNPlung-mRNAA-E), intending to stimulate lung immunity to combat TB. To enhance the vaccine efficacy, we further cofabricated monophosphoryl lipid A (MPLA) with mRNA to evaluate the adjuvanted mRNA vaccine (LNPlung-mRNAA-E-MPLA). Both vaccines elicited robust CD4+ T cell response, resulting in markedly locally higher production of IFN-γ, TNF-α, and IL-2. As anticipated, the addition of MPLA further enhanced the immunogenicity of LNPlung-mRNAA-E. However, the Mtb challenge experiment showed that LNPlung-mRNAA-E-MPLA neither provided effective protection nor enhanced the immune protection primed by BCG (Bacillus Calmette-Guérin). The subsequent HE staining of the lung revealed that the LNPlung-mRNAA-E-MPLA induced pulmonary inflammation, leading to tissue damage. Moreover, the inflammatory cytokines including IL-6, IL-1β, and MCP-1 were significantly increased and the MPLA additive exacerbated the inflammatory process. Therefore, the lung targeted mRNA vaccine and MPLA adjuvant synergistically induced lung inflammation and weakened protection from Mtb infection. Thus, this work provides valuable implications for developing targeted lung vaccines: Addressing chronic lung inflammation induced by vaccine systems is critical for lung-targeted mRNA vaccines.
Collapse
Affiliation(s)
- Liyan Li
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zeyu Yang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Hong Liu
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng 475004, China
- College of Chemistry and Molecular Sciences, Henan University, Zhengzhou 450046, China
| | - Zepeng He
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhenfu Wen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Haolin Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhihui Zhang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaoyong Fan
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| |
Collapse
|
3
|
Ellis JT, Kennedy PJ. Multi-criteria decision making and its application to in silico discovery of vaccine candidates for Toxoplasma gondii. Vaccine 2025; 58:127242. [PMID: 40378548 DOI: 10.1016/j.vaccine.2025.127242] [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: 01/27/2025] [Revised: 04/16/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Vaccine discovery against eukaryotic parasites is not trivial and few exist. Reverse vaccinology is an in silico vaccine discovery approach, designed to identify vaccine candidates from the thousands of protein sequences encoded by a target genome. Previously, we produced the Vacceed bioinformatics pipeline for identification of parasite membrane and excreted/secreted proteins that were likely be exposed to the hosts immune system. More recently, we improved upon machine learning as the final decision-making process to identify parasite proteins that induce a protective response in an animal model. Subsequently, we combined Vacceed with metrics on B and T cell epitope types to produce a new in silico discovery workflow. In this study we extend this in silico workflow to the developability of proteins as vaccines by the incorporation of metrics on the physicochemical properties of proteins. To demonstrate this process, every Toxoplasma gondii protein was ranked in its capacity to provide exposure to the immune system (Vacceed exposure score), presence of epitopes and solubility characteristics by several multicriteria decision making (MCDM) tools (such as TOPSIS, VIKOR and MABAC). A consensus rank was subsequently generated from the results of these tools using a variety of aggregate ranking methods. Levels of uncertainty in the aggregate protein rankings was assessed by conformal interval prediction in association with a machine learning model. Several of the top ranked proteins identified by this approach were novel, uncharacterized membrane transporters or proteins associated with RNA metabolism. In conclusion, MCDM automated the decision making using well known algorithms while conformal prediction intervals varied significantly across the 8000+ proteins of T. gondii. Highly ranked proteins (e.g. the top 100) typically generated low prediction intervals, providing high levels of confidence in their ranks.
Collapse
Affiliation(s)
- John T Ellis
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia.
| | - Paul J Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology and the Australian Artificial Intelligence Institute, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia
| |
Collapse
|
4
|
Liu X, Xiu C, He L, Zhao Y, Li B. Strong immune response and protection against Brucella abortus by Omp25 and BP26 mRNA vaccine candidates. Int Immunopharmacol 2025; 158:114765. [PMID: 40349401 DOI: 10.1016/j.intimp.2025.114765] [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: 02/09/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/14/2025]
Abstract
Brucellosis, a globally prevalent zoonotic disease caused by Brucella species, remains without safe and effective human vaccines. This study focuses on the outer membrane proteins Omp25 and BP26 of Brucella as targets, employing a multidimensional strategy to develop novel mRNA vaccines and systematically evaluate their immunoprotective efficacy. First, bioinformatics tools were used to predict the antigenicity, immunogenicity, and physicochemical properties of Omp25 and BP26, while the C-ImmSim server simulated their potential to induce innate and adaptive immune responses. Building on these predictions, we designed and synthesized lipid nanoparticle-encapsulated nucleoside-modified mRNA vaccines (Omp25-Fc and BP26-Fc mRNA-LNPs) and compared their immunogenicity with traditional alum-adjuvanted protein vaccines (alum-BP26 and alum-Omp25). Animal experiments demonstrated that three immunizations with Omp25-Fc and BP26-Fc mRNA-LNPs induced significantly stronger humoral and cellular immune responses in mice compared to conventional vaccines. Evaluation of protective efficacy through challenge experiments revealed a marked reduction in splenic bacterial load in both mRNA vaccine groups relative to the controls. Mechanistic analysis further showed that Omp25-Fc and BP26-Fc mRNA vaccines activated mixed Th1/Th2 immune responses, effectively reducing bacterial burden and inflammatory damage in systemic infection models. Notably, the mRNA vaccines exhibited more durable immune memory and broader protective coverage than the alum-adjuvanted protein vaccines. Our findings demonstrate that Omp25- and BP26-based mRNA-LNP vaccines exhibit high immunogenicity and clinical translational potential, providing innovative strategies and experimental evidence for the development of brucellosis vaccines.
Collapse
Affiliation(s)
- Xuyan Liu
- Hebei GEO University, Shijiazhuang 050031, China.
| | - Chenlin Xiu
- CSPC Megalith Biopharmaceutical Co. Ltd, Shijiazhuang 050051, China
| | - Lei He
- School of Chemistry & Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yunwang Zhao
- School of Chemistry & Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Baolin Li
- Qinhuangdao First Hospital affiliated to Hebei Medical University, Qinhuangdao 066000, China
| |
Collapse
|
5
|
Sandu AM, Chifiriuc MC, Vrancianu CO, Cristian RE, Alistar CF, Constantin M, Paun M, Alistar A, Popa LG, Popa MI, Tantu AC, Sidoroff ME, Mihai MM, Marcu A, Popescu G, Tantu MM. Healthcare-Associated Infections: The Role of Microbial and Environmental Factors in Infection Control-A Narrative Review. Infect Dis Ther 2025; 14:933-971. [PMID: 40208412 PMCID: PMC12084486 DOI: 10.1007/s40121-025-01143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025] Open
Abstract
Healthcare-associated infections (HAIs), previously known as nosocomial infections, represent a significant threat to healthcare systems worldwide, prolonging patient hospital stays and the duration of antimicrobial therapy. One of the most serious consequences of HAIs is the increase in the rate of antibiotic resistance (AR) generated by the prolonged, frequent, and sometimes incorrect use of antibiotics, which leads to the selection of resistant bacteria, making treatment difficult and expensive, with direct consequences for the safety of patients and healthcare personnel. Therefore, timely and accurate diagnosis of HAIs is mandatory to develop appropriate infection prevention and control practices (IPC) and new therapeutic strategies. This review aimed to present the prevalence, risk factors, current diagnosis, including artificial intelligence (AI) and machine learning approaches, future perspectives in combating HAIs causative bacteria (phage therapy, microbiome-based interventions, and vaccination), and HAIs surveillance strategies. Also, we discussed the latest findings regarding the relationships of AR with climate change and environmental pollution in the context of the One Health approach. Phage therapy is an emerging option that can offer an alternative to ineffective antibiotic treatments for antibiotic-resistant bacteria causing HAIs. Clinical trials dealing with vaccine development for resistant bacteria have yielded conflicting results. Two promising strategies, fecal microbiota transplantation and probiotic therapy, proved highly effective against recurrent Clostridium difficile infections and have been shown to reduce HAI incidence in hospitalized patients undergoing antibiotic therapy. Artificial intelligence and machine learning systems offer promising predictive capabilities in processing large volumes of clinical, microbiological, and patient data but require robust data integration. Our paper argues that HAIs are still a global challenge, requiring stringent IPC policies, computer vision, and AI-powered tools. Despite promising avenues like integrated One Health approaches, optimized phage therapy, microbiome-based interventions, and targeted vaccine development, several knowledge gaps in clinical efficacy, standardization, and pathogen complexity remain to be answered.
Collapse
Grants
- CNFIS-FDI-2024-F-0484 INOVEX University of Bucharest
- Pillar III Ministry of Research, Innovation and Digitalization through the National Recovery and Resilience Plan (PNRR) of Romania
- Component C9/Investment no. 8 (I8) - contract CF 68 Ministry of Research, Innovation and Digitalization through the National Recovery and Resilience Plan (PNRR) of Romania
- Project No. RO1567-IBB05/2023 Institute of Biology Bucharest of the Romanian Academy
- project no. 23020101 The core program within the National Research Development and Innovation Plan, 2022-2027', carried out with the support of the Ministry of Research, Innovation and Digitalization (MCID),
- Contract no. 7N from 3 January 2023 The core program within the National Research Development and Innovation Plan, 2022-2027', carried out with the support of the Ministry of Research, Innovation and Digitalization (MCID),
- Dezvoltarea cercetării genomice în România - ROGEN" (Development of genomic research in Romania -ROGEN). ROGEN
- The core program within the National Research Development and Innovation Plan, 2022–2027’, carried out with the support of the Ministry of Research, Innovation and Digitalization (MCID),
Collapse
Affiliation(s)
- Andreea M Sandu
- Doctoral School, Carol Davila University of Medicine and Pharmacy, Eroii Sanitari 8, District 5, 050474, Bucharest, Romania
- The County Emergency Hospital, Aleea Spitalului 36, 110283, Pitești, Romania
| | - Mariana C Chifiriuc
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, 050095, Bucharest, Romania
- Biological Sciences Division, Romanian Academy, 125 Calea Victoriei, 010071, Bucharest, Romania
| | - Corneliu O Vrancianu
- Doctoral School, Carol Davila University of Medicine and Pharmacy, Eroii Sanitari 8, District 5, 050474, Bucharest, Romania.
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania.
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania.
| | - Roxana-E Cristian
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
| | - Cristina F Alistar
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
- Institute of Biology of Romanian Academy, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
| | - Mihaela Paun
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
- Faculty of Administration and Business, University of Bucharest, Bucharest, Romania
| | - Alexandru Alistar
- The Research Institute of the University of Bucharest, ICUB, Șoseaua Panduri 90, District 5, 050663, Bucharest, Romania
| | - Loredana G Popa
- Faculty of Medicine, Microbiology Discipline II, Carol Davila University of Medicine and Pharmacy, 020021, Bucharest, Romania
| | - Mircea I Popa
- Faculty of Medicine, Microbiology Discipline II, Carol Davila University of Medicine and Pharmacy, 020021, Bucharest, Romania
- Preclinical Testing Unit, Cantacuzino National Military Medical Institute for Research and Development, 050096, Bucharest, Romania
| | - Ana C Tantu
- University of Medicine and Pharmacy of Craiova, Petru Rareș 2, 200349, Craiova, Romania
- Emergency Clinical County Hospital of Craiova, Tabaci 1, 200642, Craiova, Romania
| | - Manuela E Sidoroff
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, District 6, 060031, Bucharest, Romania
| | - Mara M Mihai
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021, Bucharest, Romania
- Department of Oncologic Dermatology, "Elias" University Emergency Hospital, 010024, Bucharest, Romania
| | - Andreea Marcu
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 020021, Bucharest, Romania
| | - George Popescu
- Department of Neurosurgery 4, Bagdasar-Arseni Emergency Clinical Hospital, Şoseaua Berceni 12, 041915, Bucharest, Romania
| | - Monica M Tantu
- Department of Medical Assistance and Physical Therapy, Pitesti University Center, Târgu din Vale 1, 110040, Piteşti, Romania
- Faculty of Science, Physical Education and Informatics, National University of Science and Technology, Politehnica, Splaiul Independenţei 313, District 6, 060042, Bucharest, Romania
| |
Collapse
|
6
|
Leddy O, Cui Y, Ahn R, Stopfer L, Choe E, Kim DH, Roerden M, Spranger S, Bryson BD, White FM. Validation and quantification of peptide antigens presented on MHCs using SureQuant. Nat Protoc 2025; 20:1196-1222. [PMID: 39438697 PMCID: PMC12012165 DOI: 10.1038/s41596-024-01076-x] [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: 12/20/2023] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Vaccines and immunotherapies that target peptide-major histocompatibility complexes (peptide-MHCs) have the potential to address multiple unmet medical needs in cancer and infectious disease. Designing vaccines and immunotherapies to target peptide-MHCs requires accurate identification of target peptides in infected or cancerous cells or tissue, and may require absolute or relative quantification to identify abundant targets and measure changes in presentation under different treatment conditions. Internal standard parallel reaction monitoring (also known as 'SureQuant') can be used to validate and/or quantify MHC peptides previously identified by using untargeted methods such as data-dependent acquisition. SureQuant MHC has three main use cases: (i) conclusive confirmation of the identities of putative MHC peptides via comparison with an internal synthetic stable isotope labeled (SIL) peptide standard; (ii) accurate relative quantification by using pre-formed heavy isotope-labeled peptide-MHC complexes (hipMHCs) containing SIL peptides as internal controls for technical variation; and (iii) absolute quantification of each target peptide by using different amounts of hipMHCs loaded with synthetic peptides containing one, two or three SIL amino acids to provide an internal standard curve. Absolute quantification can help determine whether the abundance of a peptide-MHC is sufficient for certain therapeutic modalities. SureQuant MHC therefore provides unique advantages for immunologists seeking to confidently validate antigenic targets and understand the dynamics of the MHC repertoire. After synthetic standards are ordered (3-4 weeks), this protocol can be carried out in 3-4 days and is suitable for individuals with mass spectrometry experience who are comfortable with customizing instrument methods.
Collapse
Affiliation(s)
- Owen Leddy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
| | - Yufei Cui
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
| | - Ryuhjin Ahn
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
| | - Lauren Stopfer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
- Aethon Therapeutics, New York, NY, USA
| | - Elizabeth Choe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
| | - Do Hun Kim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
| | - Malte Roerden
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
| | - Stefani Spranger
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Medicine, Cambridge, MA, USA.
- Center for Precision Cancer Medicine, Cambridge, MA, USA.
| |
Collapse
|
7
|
Van der Vreken A, Thery F, Tu C, Mwangi K, Meulewaeter S, De Beck L, Janssens E, De Veirman K, Vanderkerken K, De Bruyne E, Franceschini L, Impens F, Verbeke R, Lentacker I, Menu E, Breckpot K. Immunopeptidomics identified antigens for mRNA-lipid nanoparticle vaccines with alpha-galactosylceramide in multiple myeloma therapy. J Immunother Cancer 2025; 13:e010673. [PMID: 40300855 PMCID: PMC12049997 DOI: 10.1136/jitc-2024-010673] [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: 09/27/2024] [Accepted: 04/05/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Invariant natural killer T (iNKT) cells and CD8+ T cells are key in the immune response against multiple myeloma (MM), a largely incurable blood cancer. Immunization is a promising strategy to activate these T cell populations. To our knowledge, immunization with messenger RNA (mRNA) and the iNKT agonist, α-galactosylceramide (αGC), has not been studied in MM, as knowledge on clinically relevant antigens in preclinical MM models is lacking. METHODS Microarray data and immunopeptidomics (imPep) were used to identify candidate antigens for immunization in 5TMM models. Galsomes, lipid nanoparticles containing antigen mRNA and αGC were used to immunize 5T33MM-bearing mice. This treatment was combined with a CD40 agonist. Tumor burden and activation of iNKT cells and CD8+ T cells were studied using M-protein electrophoresis, flow cytometry and ELISA. RESULTS RNA transcripts revealed survivin as a candidate antigen. Prime-boost Galsomes therapy targeting survivin significantly reduced M-protein levels despite low survivin-specific T cell responses. Further analysis showed potential T cell fratricide. ImPep revealed HSP60, Idiotype, PICALM and EF1A1 as candidate antigens. Prime-boost therapy with Galsomes targeting these antigens reduced MM growth significantly when combined with a CD40 agonist, coinciding with significantly improved antigen presentation, costimulation and cytotoxicity of iNKT cells and CD8+ T cells. CONCLUSION These findings highlight the potential of Galsomes, an mRNA vaccine designed to activate CD8+ T cells and iNKT cells, for MM therapy, and emphasize the importance of combinatorial approaches, addressing immune anergy for effective MM immunotherapies.
Collapse
Affiliation(s)
- Arne Van der Vreken
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Chenggong Tu
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kevin Mwangi
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Sofie Meulewaeter
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Lien De Beck
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Edith Janssens
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lorenzo Franceschini
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Rein Verbeke
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Ine Lentacker
- Department of Pharmaceutics, Laboratory of Physical Pharmacy and General Biochemistry, Ghent Research Group on Nanomedicines, Universiteit Gent, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Universiteit Gent, Ghent, Belgium
| | - Eline Menu
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Department of Biomedical Sciences Brussels, Translational Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
8
|
Buzás GM. Bacterial carbonic anhydrase as a candidate vaccine target against Helicobacter pylori. Front Microbiol 2025; 16:1541387. [PMID: 40270820 PMCID: PMC12014598 DOI: 10.3389/fmicb.2025.1541387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/12/2025] [Indexed: 04/25/2025] Open
Affiliation(s)
- György M. Buzás
- Ferencváros Health Centre, Department of Gastroenterology, Budapest, Hungary
| |
Collapse
|
9
|
Willems P, Thery F, Van Moortel L, De Meyer M, Staes A, Gul A, Kovalchuke L, Declercq A, Devreese R, Bouwmeester R, Gabriels R, Martens L, Impens F. Maximizing Immunopeptidomics-Based Bacterial Epitope Discovery by Multiple Search Engines and Rescoring. J Proteome Res 2025; 24:2141-2151. [PMID: 40080147 PMCID: PMC11976845 DOI: 10.1021/acs.jproteome.4c00864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 03/15/2025]
Abstract
Mass spectrometry-based discovery of bacterial immunopeptides presented by infected cells allows untargeted discovery of bacterial antigens that can serve as vaccine candidates. However, reliable identification of bacterial epitopes is challenged by their extremely low abundance. Here, we describe an optimized bioinformatic framework to enhance the confident identification of bacterial immunopeptides. Immunopeptidomics data of cell cultures infected with Listeria monocytogenes were searched by four different search engines, PEAKS, Comet, Sage and MSFragger, followed by data-driven rescoring with MS2Rescore. Compared with individual search engine results, this integrated workflow boosted immunopeptide identification by an average of 27% and led to the high-confidence detection of 18 additional bacterial peptides (+27%) matching 15 different Listeria proteins (+36%). Despite the strong agreement between the search engines, a small number of spectra (<1%) had ambiguous matches to multiple peptides and were excluded to ensure high-confidence identifications. Finally, we demonstrate our workflow with sensitive timsTOF SCP data acquisition and find that rescoring, now with inclusion of ion mobility features, identifies 76% more peptides compared to Q Exactive HF acquisition. Together, our results demonstrate how integration of multiple search engine results along with data-driven rescoring maximizes immunopeptide identification, boosting the detection of high-confidence bacterial epitopes for vaccine development.
Collapse
Affiliation(s)
- Patrick Willems
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB-UGent
Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department
of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Fabien Thery
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Laura Van Moortel
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Margaux De Meyer
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - An Staes
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB
Proteomics Core, VIB, 9052 Ghent, Belgium
| | - Adillah Gul
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Lyudmila Kovalchuke
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Arthur Declercq
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Robbe Devreese
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Robbin Bouwmeester
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Ralf Gabriels
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
| | - Lennart Martens
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- BioOrganic
Mass Spectrometry Laboratory (LSMBO), IPHC UMR 7178, University of
Strasbourg, CNRS, ProFI FR2048, Strasbourg, France
| | - Francis Impens
- VIB-UGent
Center for Medical Biotechnology, VIB, 9052 Ghent, Belgium
- Department
of Biomolecular Medicine, Ghent University, 9052 Ghent, Belgium
- VIB-UGent
Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
- VIB
Proteomics Core, VIB, 9052 Ghent, Belgium
| |
Collapse
|
10
|
Cormican JA, Medfai L, Wawrzyniuk M, Pašen M, Afrache H, Fourny C, Khan S, Gneiße P, Soh WT, Timelli A, Nolfi E, Pannekoek Y, Cope A, Urlaub H, Sijts AJAM, Mishto M, Liepe J. PEPSeek-Mediated Identification of Novel Epitopes From Viral and Bacterial Pathogens and the Impact on Host Cell Immunopeptidomes. Mol Cell Proteomics 2025; 24:100937. [PMID: 40044041 PMCID: PMC12002930 DOI: 10.1016/j.mcpro.2025.100937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 04/07/2025] Open
Abstract
Here, we develop PEPSeek, a web-server-based software to allow higher performance in the identification of pathogen-derived epitope candidates detected via mass spectrometry in MHC class I immunopeptidomes. We apply it to human and mouse cell lines infected with SARS-CoV-2, Listeria monocytogenes, or Chlamydia trachomatis, thereby identifying a large number of novel antigens and epitopes that we prove to be recognized by CD8+ T cells. In infected cells, we identified antigenic peptide features that suggested how the processing and presentation of pathogenic antigens differ between pathogens. The quantitative tools of PEPSeek also helped to define how C. trachomatis infection cycle could impact the antigenic landscape of the host human cell system, likely reflecting metabolic changes that occurred in the infected cells.
Collapse
Affiliation(s)
- John A Cormican
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Lobna Medfai
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Magdalena Wawrzyniuk
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin Pašen
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Hassnae Afrache
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Research group of Molecular Immunology, Francis Crick Institute, London, United Kingdom
| | - Constance Fourny
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Research group of Molecular Immunology, Francis Crick Institute, London, United Kingdom
| | - Sahil Khan
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Pascal Gneiße
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Georg-August University School of Science (GAUSS), University of Göttingen, Göttingen, Germany
| | - Wai Tuck Soh
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Arianna Timelli
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Emanuele Nolfi
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Yvonne Pannekoek
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Andrew Cope
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Centre for Rheumatic Diseases, King's College London, London, UK
| | - Henning Urlaub
- Research group of Bioanalytical Mass Spectrometry, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Bioanalytics, Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany; Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Alice J A M Sijts
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; Chair T-cell Tolerance, Leibniz Institute for Immunotherapy, Regensburg, Germany.
| | - Michele Mishto
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Research group of Molecular Immunology, Francis Crick Institute, London, United Kingdom.
| | - Juliane Liepe
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Facility for Data Sciences and Biostatistics, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
11
|
Xiao W, Gao M, Mo B, Huang X, Du Z, Wang S, Chen J, Luo S, Xing H. Endoplasmic Reticulum-Targeted Phototherapy Remodels the Tumor Immunopeptidome to Enhance Immunogenic Cell Death and Adaptive Anti-Tumor Immunity. Pharmaceuticals (Basel) 2025; 18:491. [PMID: 40283929 PMCID: PMC12030737 DOI: 10.3390/ph18040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Endoplasmic reticulum (ER)-targeted phototherapy has emerged as a promising approach to amplify ER stress, induce immunogenic cell death (ICD), and enhance anti-tumor immunity. However, its impact on the antigenicity of dying tumor cells remains poorly understood. Methods: Laser activation of the ER-targeted photosensitizer ER-Cy-poNO2 was performed to investigate its effects on tumor cell antigenicity. Transcriptomic analysis was carried out to assess gene expression changes. Immunopeptidomics profiling was used to identify high-affinity major histocompatibility complex class I (MHC-I) ligands. In vitro functional studies were conducted to evaluate dendritic cell maturation and T lymphocyte activation, while in vivo experiments were performed by combining the identified peptide with poly IC to evaluate anti-tumor immunity. Results: Laser activation of ER-Cy-poNO2 significantly remodeled the antigenic landscape of 4T-1 tumor cells, enhancing their immunogenicity. Transcriptomic analysis revealed upregulation of antigen processing and presentation pathways. Immunopeptidomics profiling identified multiple high-affinity MHC-I ligands, with IF4G3986-994 (QGPKTIEQI) showing exceptional immunogenicity. In vitro, IF4G3986-994 promoted dendritic cell maturation and enhanced T lymphocytes activation. In vivo, the combination of IF4G3986-994 with poly IC elicited robust anti-tumor immunity, characterized by increased CD8+ T lymphocytes infiltration, reduced regulatory T cells (Tregs) in the tumor microenvironment, elevated systemic Interferon-gamma (IFN-γ) levels, and significant tumor growth inhibition without systemic toxicity. Conclusions: These findings establish a mechanistic link between ER stress-driven ICD, immunopeptidome remodeling, and adaptive immune activation, highlighting the potential of ER-targeted phototherapy as a platform for identifying immunogenic peptides and advancing peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Weidong Xiao
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Mingquan Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Banghui Mo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xie Huang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zaizhi Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shufeng Wang
- Department of Immunology, College of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Shenglin Luo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| |
Collapse
|
12
|
Hu W, Zhou S, Ibrahim A, Li G, Awad S, Ramos-Vivas J, Kan J, Du M. Whole Genome Analysis of Pediococcus acidilactici XJ-24 and Its Role in Preventing Listeria monocytogenes ATCC ® 19115 TM Infection in C57BL/6 Mice. Antibiotics (Basel) 2025; 14:323. [PMID: 40149133 PMCID: PMC11939717 DOI: 10.3390/antibiotics14030323] [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: 02/11/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: As probiotics gain prominence in the prevention and treatment of intestinal diseases, their protective effects against pathogens and influence on host health have drawn significant attention. This study investigates the genomic characteristics and functional potential of Pediococcus acidilactici XJ-24 (XJ-24) in the prevention of Listeria monocytogenes (LM) infection in mice. Methods/Results: Whole-genome analysis confirmed the safety and probiotic properties of XJ-24, including acid and bile salt tolerance, antimicrobial activity, and safety. In vivo, C57BL/6 mice challenges indicated that XJ-24 significantly reduced LM colonization, suppressed pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IFN-γ), alleviated colon and spleen tissue damage, and maintained intestinal barrier integrity by upregulating tight junction proteins (Occludin, Claudin-1, ZO-1). Moreover, XJ-24 modulated gut microbiota composition by increasing beneficial taxa while reducing harmful bacteria. Correlation analysis highlighted a positive association between Lachnospiraceae and tight junction proteins. Conclusions: These findings demonstrate the potential of XJ-24 as a functional probiotic for preventing LM infection and provide a basis for further clinical exploration.
Collapse
Affiliation(s)
- Weizhong Hu
- College of Food Science, Southwest University, Chongqing 400715, China; (W.H.); (S.Z.); (J.K.)
- Chinese-Hungarian Cooperative Research Center for Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
| | - Shuxin Zhou
- College of Food Science, Southwest University, Chongqing 400715, China; (W.H.); (S.Z.); (J.K.)
- Chinese-Hungarian Cooperative Research Center for Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
| | - Amel Ibrahim
- Faculty of Agriculture, Alexandria University, Alexandria 21500, Egypt; (A.I.); (S.A.)
| | - Guannan Li
- College of Sericulture, Textile and Biomass, Southwest University, Chongqing 400716, China;
| | - Sameh Awad
- Faculty of Agriculture, Alexandria University, Alexandria 21500, Egypt; (A.I.); (S.A.)
| | - José Ramos-Vivas
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain;
| | - Jianquan Kan
- College of Food Science, Southwest University, Chongqing 400715, China; (W.H.); (S.Z.); (J.K.)
- Chinese-Hungarian Cooperative Research Center for Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
| | - Muying Du
- College of Food Science, Southwest University, Chongqing 400715, China; (W.H.); (S.Z.); (J.K.)
- Chinese-Hungarian Cooperative Research Center for Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
| |
Collapse
|
13
|
De Meyst A, Van Mieghem J, Chiers K, Raemdonck K, Verbeke R, Lentacker I, Vanrompay D. mRNA Galsomes Vaccine Protects Budgerigars Against Virulent Chlamydia psittaci Challenge. Vaccines (Basel) 2025; 13:206. [PMID: 40006752 PMCID: PMC11861616 DOI: 10.3390/vaccines13020206] [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: 01/16/2025] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Chlamydia (C.) psittaci is an avian respiratory pathogen that regularly infects budgerigars (Melopsittacus undulatus) and is a known zoonosis. This study aimed to evaluate the efficacy of a nucleoside-modified mRNA vaccine formulated in lipid nanoparticles (LNPs), either with (mRNA Galsomes) or without (mRNA LNPs) the glycolipid antigen α-Galactosylceramide, in protecting budgerigars against C. psittaci genotype A infection. METHODS Three groups of eight budgerigars received two intramuscular vaccinations with PBS, mRNA LNPs or mRNA Galsomes, and were subsequently challenged via aerosol with the C. psittaci genotype A strain 90/1051. Vaccine efficacy was assessed over 14 days post challenge by monitoring clinical signs, macroscopic and microscopic lesions, pathogen excretion and chlamydial burden in organs. Antibody levels were evaluated at baseline, after vaccination and post challenge. RESULTS Both mRNA LNPs and mRNA Galsomes induced significant serum antibody responses post booster. Vaccination significantly reduced clinical signs, chlamydial burden in the lungs and macroscopic lesions in conjunctiva, conchae, lungs and thoracic airsacs, compared to controls. Additionally, mRNA Galsomes-treated birds showed a significantly reduced lung inflammation and fewer macroscopic lesions in abdominal airsacs and liver, compared to non-vaccinated animals. These animals also experienced a significantly lower chlamydial burden in the spleen, fewer clinical signs at day 11 and fewer fecal shedding at day 14 post challenge, compared to mRNA LNP-treated animals. CONCLUSIONS This study demonstrated that mRNA vaccination confers partial protection against C. psittaci in budgerigars, with mRNA Galsomes appearing to provide enhanced efficacy. However, the absence of species-specific reagents for assessing cellular immunity in Psittaciformes limits a comprehensive understanding of vaccine-induced protection. The development of psittacine-specific T cell markers and cytokine assays is necessary to further elucidate immune mechanisms and optimize vaccine formulations.
Collapse
Affiliation(s)
- Anne De Meyst
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (A.D.M.); (J.V.M.)
| | - Joeri Van Mieghem
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (A.D.M.); (J.V.M.)
| | - Koen Chiers
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium;
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (K.R.); (R.V.); (I.L.)
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (K.R.); (R.V.); (I.L.)
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium; (K.R.); (R.V.); (I.L.)
| | - Daisy Vanrompay
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (A.D.M.); (J.V.M.)
| |
Collapse
|
14
|
Hou T, Huang X, Lai J, Zhou D. Intra-tumoral bacteria in breast cancer and intervention strategies. Adv Drug Deliv Rev 2025; 217:115516. [PMID: 39828126 DOI: 10.1016/j.addr.2025.115516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/09/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
The microbiome, consisting of a wide range of both beneficial and harmful microorganisms, is vital to various physiological and pathological processes in the human body, including cancer pathogenesis. Tumor progression is often accompanied by the destruction of the vascular system, allowing bacteria to circulate into the tumor area and flourish in an immunosuppressive environment. Microbes are recognized as significant components of the tumor microenvironment. Recent research has increasingly focused on the role of intra-tumoral bacteria in the onset, progression, and treatment of breast cancer-the most prevalent cancer among women. This review elucidates the potential mechanisms by which intra-tumoral bacteria impact breast cancer and discusses different therapeutic approaches aimed at targeting these bacteria. It provides essential insights for enhancing existing treatment paradigms while paving the way for novel anticancer interventions. As our understanding of the microbiome's intricate relationship with cancer deepens, it opens avenues for groundbreaking strategies that could redefine oncology.
Collapse
Affiliation(s)
- Ting Hou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoling Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiahui Lai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Dongfang Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
15
|
Huang T, Che S, Lv Z, Hao D, Wang R, Yi Q, Mei L, Yuan Y, Zou H, Guo Y, Wang X, Chu Y, Zhao K. mRNA-LNP vaccines combined with tPA signal sequence elicit strong protective immunity against Klebsiella pneumoniae. mSphere 2025; 10:e0077524. [PMID: 39745376 PMCID: PMC11774038 DOI: 10.1128/msphere.00775-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Klebsiella pneumoniae is a prominent Gram-negative and encapsulated opportunistic pathogen that causes a multitude of infections such as severe respiratory and healthcare-associated infections. Despite the widespread anti-microbial resistance and the high mortality rate, currently, no clinically vaccine is approved for battling K. pneumoniae. To date, messenger RNA (mRNA) vaccine is one of the most advancing technologies and are extensively investigated for viral infection, while infrequently applied for prevention of bacterial infections. In the present study, we aim to construct a new mRNA vaccine encoding YidR or combining with a tissue plasminogen activator signal sequence for preventing K. pneumoniae infection. Adaptive immunity was determined in mRNA vaccines-immunized mice and the protective effects of mRNA vaccines were evaluated in K. pneumoniae infected models. The results showed that lipid nanoparticle (LNP)-YidR-mRNA vaccine was produced with good morphology, high the encapsulation efficiency, and the specific antigen was highly expressed in cells in vitro. In addition, immunization with either LNP-YidR or LNP-YidR-SP elicited a Th1-biased immune response, reduced bacterial load, and provided broad protection in the lung infection models. Importantly, the LNP-YidR-SP mRNA vaccine induced strong adaptive humoral and cellular immunity and increased the survivability of mice compared to the other groups. Our findings serve as a focal point for developing a potential mRNA vaccine against K. pneumoniae, indicating the potential of mRNA vaccines for improving next-generation bacterial vaccine.IMPORTANCEK. pneumoniae is a notorious and clinical bacterium that is evolving in community-acquired and nosocomial settings. This opportunistic pathogen causes severe infectious diseases, including urinary tract infection and pneumonia, and causes a concerning global public burden. Despite efforts having been created to develop different types of K. pneumoniae vaccines, there is no licensed vaccine for preventing K. pneumoniae infection. Therefore, to develop an effective tactic is essential to combat K. pneumoniae-caused diseases. This study provides a novel vaccine strategy against K. pneumoniae and a potent platform to elicit high levels of humoral and cell-meditated immunity.
Collapse
Affiliation(s)
- Ting Huang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Siyou Che
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Zheng Lv
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Danrui Hao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Runyu Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Qinxuan Yi
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Ling Mei
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Yang Yuan
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Hang Zou
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Yidong Guo
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Xinrong Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| | - Kelei Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| |
Collapse
|
16
|
Sastalla I, Kwon K, Huntley C, Taylor K, Brown L, Samuel T, Zou L. NIAID Workshop Report: Systematic Approaches for ESKAPE Bacteria Antigen Discovery. Vaccines (Basel) 2025; 13:87. [PMID: 39852866 PMCID: PMC11768834 DOI: 10.3390/vaccines13010087] [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: 12/11/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
On 14-15 November 2023, the National Institute of Allergy and Infectious Diseases (NIAID) organized a workshop entitled "Systematic Approaches for ESKAPE Bacteria Antigen Discovery". The goal of the workshop was to engage scientists from diverse relevant backgrounds to explore novel technologies that can be harnessed to identify and address current roadblocks impeding advances in antigen and vaccine discoveries for the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). The workshop consisted of four sessions that addressed ESKAPE infections, antigen discovery and vaccine efforts, and new technologies including systems immunology and vaccinology approaches. Each session was followed by a panel discussion. In total, there were over 260 in-person and virtual attendees, with high levels of engagement. This report provides a summary of the event and highlights challenges and opportunities in the field of ESKAPE vaccine discovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lanling Zou
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (I.S.); (K.K.); (C.H.); (K.T.); (L.B.); (T.S.)
| |
Collapse
|
17
|
Khlebnikova A, Kirshina A, Zakharova N, Ivanov R, Reshetnikov V. Current Progress in the Development of mRNA Vaccines Against Bacterial Infections. Int J Mol Sci 2024; 25:13139. [PMID: 39684849 DOI: 10.3390/ijms252313139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Bacterial infections have accompanied humanity for centuries. The discovery of the first antibiotics and the subsequent golden era of their discovery temporarily shifted the balance in this confrontation to the side of humans. Nevertheless, the excessive and improper use of antibacterial drugs and the evolution of bacteria has gotten the better of humans again. Therefore, today, the search for new antibacterial drugs or the development of alternative approaches to the prevention and treatment of bacterial infections is relevant and topical again. Vaccination is one of the most effective strategies for the prevention of bacterial infections. The success of new-generation vaccines, such as mRNA vaccines, in the fight against viral infections has prompted many researchers to design mRNA vaccines against bacterial infections. Nevertheless, the biology of bacteria and their interactions with the host's immunity are much more complex compared to viruses. In this review, we discuss structural features and key mechanisms of evasion of an immune response for nine species of bacterial pathogens against which mRNA vaccines have been developed and tested in animals. We focus on the results of experiments involving the application of mRNA vaccines against various bacterial pathogens in animal models and discuss possible options for improving the vaccines' effectiveness. This is one of the first comprehensive reviews of the use of mRNA vaccines against bacterial infections in vivo to improve our knowledge.
Collapse
Affiliation(s)
- Alina Khlebnikova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Anna Kirshina
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Natalia Zakharova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi 354340, Russia
| |
Collapse
|
18
|
Versteeg L, Adhikari R, Robinson G, Lee J, Wei J, Islam N, Keegan B, Russell WK, Poveda C, Villar MJ, Jones K, Bottazzi ME, Hotez P, Tijhaar E, Pollet J. Immunopeptidomic MHC-I profiling and immunogenicity testing identifies Tcj2 as a new Chagas disease mRNA vaccine candidate. PLoS Pathog 2024; 20:e1012764. [PMID: 39693359 DOI: 10.1371/journal.ppat.1012764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Trypanosoma cruzi is a protozoan parasite that causes Chagas disease. Globally 6 to 7 million people are infected by this parasite of which 20-30% will progress to develop Chronic Chagasic Cardiomyopathy (CCC). Despite its high disease burden, no clinically approved vaccine exists for the prevention or treatment of CCC. Developing vaccines that can stimulate T. cruzi-specific CD8+ cytotoxic T cells and eliminate infected cells requires targeting parasitic antigens presented on major histocompatibility complex-I (MHC-I) molecules. We utilized mass spectrometry-based immunopeptidomics to investigate which parasitic peptides are displayed on MHC-I of T. cruzi infected cells. Through duplicate experiments, we identified an array of unique peptides that could be traced back to 17 distinct T. cruzi proteins. Notably, six peptides were derived from Tcj2, a trypanosome chaperone protein and member of the DnaJ (heat shock protein 40) family, showcasing its potential as a viable candidate vaccine antigen with cytotoxic T cell inducing capacity. Upon testing Tcj2 as an mRNA vaccine candidate in mice, we observed a strong memory cytotoxic CD8+ T cell response along with a Th1-skewed humoral antibody response. In vitro co-cultures of T. cruzi infected cells with splenocytes of Tcj2-immunized mice restricted the replication of T. cruzi, demonstrating the protective potential of Tcj2 as a vaccine target. Moreover, antisera from Tcj2-vaccinated mice displayed no cross-reactivity with DnaJ in lysates from mouse and human indicating a decreased likelihood of triggering autoimmune reactions. Our findings highlight how immunopeptidomics can identify new vaccine targets for Chagas disease, with Tcj2 emerging as a promising new mRNA vaccine candidate.
Collapse
Affiliation(s)
- Leroy Versteeg
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Rakesh Adhikari
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Gonteria Robinson
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jungsoon Lee
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Junfei Wei
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Nelufa Islam
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Brian Keegan
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - William K Russell
- University of Texas Medical Branch, Mass Spectrometry Facility, UTMB Health, Galveston, Texas, United States of America
| | - Cristina Poveda
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Maria Jose Villar
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kathryn Jones
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Peter Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Edwin Tijhaar
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Jeroen Pollet
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
19
|
Kovalchik KA, Hamelin DJ, Kubiniok P, Bourdin B, Mostefai F, Poujol R, Paré B, Simpson SM, Sidney J, Bonneil É, Courcelles M, Saini SK, Shahbazy M, Kapoor S, Rajesh V, Weitzen M, Grenier JC, Gharsallaoui B, Maréchal L, Wu Z, Savoie C, Sette A, Thibault P, Sirois I, Smith MA, Decaluwe H, Hussin JG, Lavallée-Adam M, Caron E. Machine learning-enhanced immunopeptidomics applied to T-cell epitope discovery for COVID-19 vaccines. Nat Commun 2024; 15:10316. [PMID: 39609459 PMCID: PMC11604954 DOI: 10.1038/s41467-024-54734-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Next-generation T-cell-directed vaccines for COVID-19 focus on establishing lasting T-cell immunity against current and emerging SARS-CoV-2 variants. Precise identification of conserved T-cell epitopes is critical for designing effective vaccines. Here we introduce a comprehensive computational framework incorporating a machine learning algorithm-MHCvalidator-to enhance mass spectrometry-based immunopeptidomics sensitivity. MHCvalidator identifies unique T-cell epitopes presented by the B7 supertype, including an epitope from a + 1-frameshift in a truncated Spike antigen, supported by ribosome profiling. Analysis of 100,512 COVID-19 patient proteomes shows Spike antigen truncation in 0.85% of cases, revealing frameshifted viral antigens at the population level. Our EpiTrack pipeline tracks global mutations of MHCvalidator-identified CD8 + T-cell epitopes from the BNT162b4 vaccine. While most vaccine epitopes remain globally conserved, an immunodominant A*01-associated epitope mutates in Delta and Omicron variants. This work highlights SARS-CoV-2 antigenic features and emphasizes the importance of continuous adaptation in T-cell vaccine development.
Collapse
Affiliation(s)
- Kevin A Kovalchik
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - David J Hamelin
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
- Mila-Quebec AI Institute, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Peter Kubiniok
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Benoîte Bourdin
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Fatima Mostefai
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
- Mila-Quebec AI Institute, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Raphaël Poujol
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Bastien Paré
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Shawn M Simpson
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Éric Bonneil
- Institute of Research in Immunology and Cancer, Montreal, QC, Canada
| | | | - Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mohammad Shahbazy
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Saketh Kapoor
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Vigneshwar Rajesh
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Maya Weitzen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Bayrem Gharsallaoui
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Loïze Maréchal
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Zhaoguan Wu
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Christopher Savoie
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Pierre Thibault
- Institute of Research in Immunology and Cancer, Montreal, QC, Canada
- Department of Chemistry, Université de Montréal, Montreal, QC, Canada
| | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Martin A Smith
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Hélène Decaluwe
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Microbiology, Infectiology and Immunology Department, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Julie G Hussin
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada.
- Mila-Quebec AI Institute, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Mathieu Lavallée-Adam
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada.
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Yale Center for Immuno-Oncology, Yale Center for Systems and Engineering Immunology, Yale Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
20
|
Luo F, Xu C, Zhang C, Tan A, Lu D, Luo P, Cheng P, Zhang W, Bai L, Yu C, Sun S, Zeng H, Zou Q. mRNA-based platform for preventing and treating Staphylococcus aureus by targeted staphylococcal enterotoxin B. Front Immunol 2024; 15:1490044. [PMID: 39640268 PMCID: PMC11617584 DOI: 10.3389/fimmu.2024.1490044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Staphylococcus aureus (S. aureus) possesses numerous virulence factors, with the increasing prevalence of drug-resistant strains heightening the threat posed by this pathogen. Staphylococcal enterotoxin B (SEB), a highly conserved toxin secreted by S. aureus, is also recognized as a potential bioweapon with super-antigenic activity. SEB represents a promising target in efforts to combat infections caused by S. aureus. We developed mRNA-based vaccine and antibody targeting SEB for both prophylactic and therapeutic purposes in varying S. aureus infection conditions. The mSEB mRNA vaccine (10 μg per mouse) induces more robust and persistent immune responses, including higher antibody titers and specific cellular immune responses, compared to immunization with 30 μg of mSEB protein adjuvanted with aluminum phosphate. Additionally, the anti-SEB mRNA antibody maintains secretion of anti-SEB monoclonal antibody (mAb) with a dosage that is 10 times lower than purified protein administration. The mRNA-based antibody exhibits superior pharmacokinetic profiles compared to its protein counterparts, efficiently neutralizing SEB and clearing S. aureus from circulation. Both the mRNA vaccine and mRNA antibody demonstrate preventive and therapeutic effects by eliciting specific immune responses and generating high-affinity antibodies in mice. We have laid the groundwork for the development and evaluation of mRNA-based vaccines and antibodies targeting SEB produced by S. aureus. Our studies demonstrate that these approaches are more effective than traditional protein-based vaccines and antibodies in terms of inducing immune responses, pharmacokinetics, and their prophylactic or therapeutic efficacy against S. aureus infections.
Collapse
Affiliation(s)
- Fumei Luo
- School of Pharmacy, University of South China, Hunan, China
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chuanfei Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chengwen Zhang
- Medical Research Institute, Southwest University, Chongqing, China
| | - Aomo Tan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Lijuan Bai
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Cuiyun Yu
- School of Pharmacy, University of South China, Hunan, China
| | - Si Sun
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| |
Collapse
|
21
|
Zhu Z, Song M, Ren J, Liang L, Mao G, Chen M. Copper homeostasis and cuproptosis in central nervous system diseases. Cell Death Dis 2024; 15:850. [PMID: 39567497 PMCID: PMC11579297 DOI: 10.1038/s41419-024-07206-3] [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: 01/29/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
Copper (Cu), an indispensable micronutrient for the sustenance of living organisms, contributes significantly to a vast array of fundamental metabolic processes. The human body maintains a relatively low concentration of copper, which is mostly found in the bones, liver, and brain. Despite its low concentration, Cu plays a crucial role as an indispensable element in the progression and pathogenesis of central nervous system (CNS) diseases. Extensive studies have been conducted in recent years on copper homeostasis and copper-induced cell death in CNS disorders, including glioma, Alzheimer's disease, Amyotrophic lateral sclerosis, Huntington's disease, and stroke. Cuproptosis, a novel copper-induced cell death pathway distinct from apoptosis, necrosis, pyroptosis, and ferroptosis, has been identified as potentially intricately linked to the pathogenic mechanisms underlying various CNS diseases. Therefore, a systematic review of copper homeostasis and cuproptosis and their relationship with CNS disorders could deepen our understanding of the pathogenesis of these diseases. In addition, it may provide new insights and strategies for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Zhipeng Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, China
| | - Min Song
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Jianxun Ren
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Lirong Liang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Guohua Mao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Min Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
22
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [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: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
23
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [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] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Celis-Giraldo C, Suárez CF, Agudelo W, Ibarrola N, Degano R, Díaz J, Manzano-Román R, Patarroyo MA. Immunopeptidomics of Salmonella enterica Serovar Typhimurium-Infected Pig Macrophages Genotyped for Class II Molecules. BIOLOGY 2024; 13:832. [PMID: 39452141 PMCID: PMC11505383 DOI: 10.3390/biology13100832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024]
Abstract
Salmonellosis is a zoonotic infection that has a major impact on human health; consuming contaminated pork products is the main source of such infection. Vaccination responses to classic vaccines have been unsatisfactory; that is why peptide subunit-based vaccines represent an excellent alternative. Immunopeptidomics was used in this study as a novel approach for identifying antigens coupled to major histocompatibility complex class II molecules. Three homozygous individuals having three different haplotypes (Lr-0.23, Lr-0.12, and Lr-0.21) were thus selected as donors; peripheral blood macrophages were then obtained and stimulated with Salmonella typhimurium (MOI 1:40). Although similarities were observed regarding peptide length distribution, elution patterns varied between individuals; in total, 1990 unique peptides were identified as follows: 372 for Pig 1 (Lr-0.23), 438 for Pig 2 (Lr.0.12) and 1180 for Pig 3 (Lr.0.21). Thirty-one S. typhimurium unique peptides were identified; most of the identified peptides belonged to outer membrane protein A and chaperonin GroEL. Notably, 87% of the identified bacterial peptides were predicted in silico to be elution ligands. These results encourage further in vivo studies to assess the immunogenicity of the identified peptides, as well as their usefulness as possible protective vaccine candidates.
Collapse
Affiliation(s)
- Carmen Celis-Giraldo
- Veterinary Medicine Programme, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá 111166, Colombia; (C.C.-G.); (J.D.)
- PhD Programme in Tropical Health and Development, Doctoral School “Studii Salamantini”, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Carlos F. Suárez
- Grupo de Investigación Básica en Biología Molecular e Inmunología (GIBBMI), Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá 111321, Colombia; (C.F.S.); (W.A.)
| | - William Agudelo
- Grupo de Investigación Básica en Biología Molecular e Inmunología (GIBBMI), Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá 111321, Colombia; (C.F.S.); (W.A.)
| | - Nieves Ibarrola
- Centro de Investigación del Cáncer e Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, 37007 Salamanca, Spain; (N.I.); (R.D.)
| | - Rosa Degano
- Centro de Investigación del Cáncer e Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, 37007 Salamanca, Spain; (N.I.); (R.D.)
| | - Jaime Díaz
- Veterinary Medicine Programme, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá 111166, Colombia; (C.C.-G.); (J.D.)
| | - Raúl Manzano-Román
- Infectious and Tropical Diseases Group (e-INTRO), IBSAL-CIETUS (Instituto de Investigación Biomédica de Salamanca—Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca), Pharmacy Faculty, Universidad de Salamanca, 37007 Salamanca, Spain;
| | - Manuel A. Patarroyo
- Grupo de Investigación Básica en Biología Molecular e Inmunología (GIBBMI), Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá 111321, Colombia; (C.F.S.); (W.A.)
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| |
Collapse
|
25
|
Saadat M, Zare-Mirakabad F, Masoudi-Nejad A, Farahanchi Baradaran M, Hosseinkhan N. HLAPepBinder: An Ensemble Model for The Prediction Of HLA-Peptide Binding. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3927. [PMID: 40225296 PMCID: PMC11993240 DOI: 10.30498/ijb.2024.459448.3927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 12/03/2024] [Indexed: 04/15/2025]
Abstract
Background Human leukocyte antigens (HLAs) play a pivotal role in orchestrating the host's immune response, offering a promising avenue with reduced adverse effects compared to conventional treatments. Cancer immunotherapies use HLA class I molecules for T cells to recognize tumor antigens, emphasizing the importance of identifying peptides that bind effectively to HLAs. Computer modeling of HLA-peptide binding speeds up the search for immunogenic epitopes, which enhances the prospect of personalized medicine and targeted therapies. The Immune Epitope Database (IEDB) is a vital repository, housing curated immune epitope data and prediction tools for HLA-peptide binding. It can be challenging for immunologists to choose the best tool from the IEDB for predicting HLA-peptide binding. This has led to the creation of consensus-based methods that combine the results of several predictors. One of the major challenges in these methods is how to effectively integrate the results from multiple predictors. Objectives Previous consensus-based methods integrate at most three tools by relying on simple strategies, such as selecting prediction methods based on their proximity to HLA in training data. In this study, we introduce HLAPepBinder, a novel consensus approach using ensemble machine learning methods to predict HLA-peptide binding, addressing the challenges biologists face in model selection. Materials and Methods The key contribution is the development of an automatic pipeline named HLAPepBinder that integrates the predictions of multiple models using a random forest approach. Unlike previous approaches, HLAPepBinder seamlessly integrates results from all nine predictors, providing a comprehensive and accurate predictive framework. By combining the strengths of these models, HLAPepBinder eliminates the need for manual model selection, providing a streamlined and reliable solution for biologists. Results HLAPepBinder offers a practical and high-performing alternative for HLA-peptide binding predictions, outperforming both traditional methods and complex deep learning models. Compared to the recently introduced transformer-based model, TranspHLA, which requires substantial computational resources, HLAPepBinder demonstrates superior performance in both prediction accuracy and resource efficiency. Notably, it operates effectively in limited computational environments, making it accessible to researchers with minimal resources. The codes are available online at https://github.com/CBRC-lab/HLAPepBinder. Conclusion Our study introduces a novel ensemble-learning model designed to enhance the accuracy and efficiency of HLA-peptide binding predictions. Due to the lack of reliable negative data and the typical assumption of unknown interactions being negative, we focus on analyzing the unknown HLA-peptide bindings in the test set that our model predicts with 100% certainty as positive bindings. Using HLAPepBinder, we identify 26 HLA-peptide pairs with absolute prediction confidence. These predictions are validated through a multi-step pipeline involving literature review, BLAST sequence similarity analysis, and molecular docking studies. This comprehensive validation process highlights HLAPepBinder's ability to make accurate and reliable predictions, contributing significantly to advancements in immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Mahsa Saadat
- Computational Biology Research Center (CBRC), Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | - Fatemeh Zare-Mirakabad
- Computational Biology Research Center (CBRC), Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mohammad Farahanchi Baradaran
- Computational Biology Research Center (CBRC), Department of Mathematics and Computer Science, Amirkabir University of Technology, Tehran, Iran
| | - Nazanin Hosseinkhan
- Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Lokras AG, Bobak TR, Baghel SS, Sebastiani F, Foged C. Advances in the design and delivery of RNA vaccines for infectious diseases. Adv Drug Deliv Rev 2024; 213:115419. [PMID: 39111358 DOI: 10.1016/j.addr.2024.115419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
RNA medicines represent a paradigm shift in treatment and prevention of critical diseases of global significance, e.g., infectious diseases. The highly successful messenger RNA (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were developed at record speed during the coronavirus disease 2019 pandemic. A consequence of this is exceptionally shortened vaccine development times, which in combination with adaptability makes the RNA vaccine technology highly attractive against infectious diseases and for pandemic preparedness. Here, we review state of the art in the design and delivery of RNA vaccines for infectious diseases based on different RNA modalities, including linear mRNA, self-amplifying RNA, trans-amplifying RNA, and circular RNA. We provide an overview of the clinical pipeline of RNA vaccines for infectious diseases, and present analytical procedures, which are paramount for characterizing quality attributes and guaranteeing their quality, and we discuss future perspectives for using RNA vaccines to combat pathogens beyond SARS-CoV-2.
Collapse
Affiliation(s)
- Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Thomas Rønnemoes Bobak
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Saahil Sandeep Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Federica Sebastiani
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Division of Physical Chemistry, Department of Chemistry, Lund University, 22100, Lund, Sweden
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
27
|
Zhao N, Li JX, Han YJ, Lv LP, Deng J, Zhang YY. A promising strategy to improve the stability and immunogenicity of killed but metabolically active vaccines: low-temperature preparation and coating of nanoparticles. NANOSCALE 2024; 16:17118-17125. [PMID: 39189698 DOI: 10.1039/d4nr02323d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Bacteria are becoming an increasingly serious threat to human health. The emergence of super bacteria makes clinical treatment more difficult. Vaccines are one of the most effective means of preventing and treating bacterial infections. As a new class of vaccines, killed but metabolically active (KBMA) vaccines provide the immunogenicity of live vaccines and the safety of inactivated vaccines. Herein, a promising strategy is proposed to improve the stability and immunogenicity of KBMA vaccines. KBMA vaccines were produced at low temperature (4 °C), and the bacterial surface was engineered using mesoporous silica nanoparticle (MSN) coating. Compared to vaccines prepared at room temperature, the metabolic activity of KBMA vaccines prepared at 4 °C remarkably improved. Benefiting from the induction of MSNs, the stability of KBMA vaccines was increased and the preservation time was prolonged at 4 °C. Meanwhile, metabolomics analysis showed that the metabolite spectrum of live bacteria changed after photochemical treatment and MSN coating, which interfered with organic acid metabolism pathways, lipid metabolism and biosynthesis of secondary metabolites. Furthermore, the immune response in the mice treated with KBMA/MSN vaccines was similar to that in those treated with live vaccines and stronger than that in those treated with inactivated vaccines. In comparison with the control group, bacteria tissue burdens of KBMA/MSN group were significantly reduced. CD4+ T cells dominated immune responses for the protection of mice. Thus, the current work promotes the application of KBMA vaccines, providing an alternative choice for treating bacterial infections.
Collapse
Affiliation(s)
- Ning Zhao
- Academy of Military Medical Sciences, Beijing 100850, China.
| | - Jia-Xv Li
- Academy of Military Medical Sciences, Beijing 100850, China.
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yong-Jiao Han
- Academy of Military Medical Sciences, Beijing 100850, China.
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Li-Ping Lv
- Academy of Military Medical Sciences, Beijing 100850, China.
| | - Jiang Deng
- Academy of Military Medical Sciences, Beijing 100850, China.
| | - Yan-Yu Zhang
- Academy of Military Medical Sciences, Beijing 100850, China.
| |
Collapse
|
28
|
Leddy O, Yuki Y, Carrington M, Bryson BD, White FM. PathMHC: a workflow to selectively target pathogen-derived MHC peptides in discovery immunopeptidomics experiments for vaccine target identification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612454. [PMID: 39314426 PMCID: PMC11419027 DOI: 10.1101/2024.09.11.612454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Vaccine-elicited T cell responses can contribute to immune protection against emerging infectious disease risks such as antimicrobials-resistant (AMR) microbial pathogens and viruses with pandemic potential, but rapidly identifying appropriate targets for T cell priming vaccines remains challenging. Mass spectrometry (MS) analysis of peptides presented on major histocompatibility complexes (MHCs) can identify potential targets for protective T cell responses in a proteome-wide manner. However, pathogen-derived peptides are outnumbered by self peptides in the MHC repertoire and may be missed in untargeted MS analyses. Here we present a novel approach, termed PathMHC, that uses computational analysis of untargeted MS data followed by targeted MS to discover novel pathogen-derived MHC peptides more efficiently than untargeted methods alone. We applied this workflow to identify MHC peptides derived from multiple microbes, including potential vaccine targets presented on MHC-I by human dendritic cells infected with Mycobacterium tuberculosis . PathMHC will facilitate antigen discovery campaigns for vaccine development.
Collapse
|
29
|
Gul A, Pewe LL, Willems P, Mayer R, Thery F, Asselman C, Aernout I, Verbeke R, Eggermont D, Van Moortel L, Upton E, Zhang Y, Boucher K, Miret-Casals L, Demol H, De Smedt SC, Lentacker I, Radoshevich L, Harty JT, Impens F. Immunopeptidomics Mapping of Listeria monocytogenes T Cell Epitopes in Mice. Mol Cell Proteomics 2024; 23:100829. [PMID: 39147027 PMCID: PMC11414675 DOI: 10.1016/j.mcpro.2024.100829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes, we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6000 mouse self-peptides presented on MHC class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in the mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification.
Collapse
Affiliation(s)
- Adillah Gul
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lecia L Pewe
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Patrick Willems
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Rupert Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laura Van Moortel
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ellen Upton
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Yifeng Zhang
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Laia Miret-Casals
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Hans Demol
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.
| | - John T Harty
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
30
|
Bergstrom C, Fischer NO, Kubicek-Sutherland JZ, Stromberg ZR. mRNA vaccine platforms to prevent bacterial infections. Trends Mol Med 2024; 30:524-526. [PMID: 38485647 DOI: 10.1016/j.molmed.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 06/15/2024]
Abstract
Bacterial infections are an urgent public health priority. The application of mRNA vaccine technology to prevent bacterial infections is a promising therapeutic strategy undergoing active development. This article discusses recent advances and limitations of mRNA vaccines to prevent bacterial diseases and provides perspectives on future research directions.
Collapse
Affiliation(s)
- Carson Bergstrom
- Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, WA, USA; Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Nicholas O Fischer
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Zachary R Stromberg
- Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
31
|
Meulewaeter S, Aernout I, Deprez J, Engelen Y, De Velder M, Franceschini L, Breckpot K, Van Calenbergh S, Asselman C, Boucher K, Impens F, De Smedt SC, Verbeke R, Lentacker I. Alpha-galactosylceramide improves the potency of mRNA LNP vaccines against cancer and intracellular bacteria. J Control Release 2024; 370:379-391. [PMID: 38697317 DOI: 10.1016/j.jconrel.2024.04.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Although various types of mRNA-based vaccines have been explored, the optimal conditions for induction of both humoral and cellular immunity remain rather unknown. In this study, mRNA vaccines of nucleoside-modified mRNA in lipoplexes (LPXs) or lipid nanoparticles (LNPs) were evaluated after administration in mice through different routes, assessing mRNA delivery, tolerability and immunogenicity. In addition, we investigated whether mRNA vaccines could benefit from the inclusion of the adjuvant alpha-galactosylceramide (αGC), an invariant Natural Killer T (iNKT) cell ligand. Intramuscular (IM) vaccination with ovalbumin (OVA)-encoding mRNA encapsulated in LNPs adjuvanted with αGC showed the highest antibody- and CD8+ T cell responses. Furthermore, we observed that addition of signal peptides and endocytic sorting signals of either LAMP1 or HLA-B7 in the OVA-encoding mRNA sequence further enhanced CD8+ T cell activation although reducing the induction of IgG antibody responses. Moreover, mRNA LNPs with the ionizable lipidoid C12-200 exhibited higher pro-inflammatory- and reactogenic activity compared to mRNA LNPs with SM-102, correlating with increased T cell activation and antitumor potential. We also observed that αGC could further enhance the cellular immunity of clinically relevant mRNA LNP vaccines, thereby promoting therapeutic antitumor potential. Finally, a Listeria monocytogenes mRNA LNP vaccine supplemented with αGC showed synergistic protective effects against listeriosis, highlighting a key advantage of co-activating iNKT cells in antibacterial mRNA vaccines. Taken together, our study offers multiple insights for optimizing the design of mRNA vaccines for disease applications, such as cancer and intracellular bacterial infections.
Collapse
Affiliation(s)
- Sofie Meulewaeter
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium
| | - Joke Deprez
- Inflammation Research Center, VIB-UGent, Zwijnaarde, Belgium
| | - Yanou Engelen
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium
| | - Margo De Velder
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium
| | - Lorenzo Franceschini
- Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center, Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Serge Van Calenbergh
- Laboratory of Medicinal Chemistry, Faculty of Pharmacy, Ghent University, Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium.
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
32
|
Zhang W, Sun S, Zhu W, Meng D, Hu W, Yang S, Gao M, Yao P, Wang Y, Wang Q, Ji J. Birinapant Reshapes the Tumor Immunopeptidome and Enhances Antigen Presentation. Int J Mol Sci 2024; 25:3660. [PMID: 38612472 PMCID: PMC11011986 DOI: 10.3390/ijms25073660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Birinapant, an antagonist of the inhibitor of apoptosis proteins, upregulates MHCs in tumor cells and displays a better tumoricidal effect when used in combination with immune checkpoint inhibitors, indicating that Birinapant may affect the antigen presentation pathway; however, the mechanism remains elusive. Based on high-resolution mass spectrometry and in vitro and in vivo models, we adopted integrated genomics, proteomics, and immunopeptidomics strategies to study the mechanism underlying the regulation of tumor immunity by Birinapant from the perspective of antigen presentation. Firstly, in HT29 and MCF7 cells, Birinapant increased the number and abundance of immunopeptides and source proteins. Secondly, a greater number of cancer/testis antigen peptides with increased abundance and more neoantigens were identified following Birinapant treatment. Moreover, we demonstrate the existence and immunogenicity of a neoantigen derived from insertion/deletion mutation. Thirdly, in HT29 cell-derived xenograft models, Birinapant administration also reshaped the immunopeptidome, and the tumor exhibited better immunogenicity. These data suggest that Birinapant can reshape the tumor immunopeptidome with respect to quality and quantity, which improves the presentation of CTA peptides and neoantigens, thus enhancing the immunogenicity of tumor cells. Such changes may be vital to the effectiveness of combination therapy, which can be further transferred to the clinic or aid in the development of new immunotherapeutic strategies to improve the anti-tumor immune response.
Collapse
Affiliation(s)
- Weiyan Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Shenghuan Sun
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94143, USA;
| | - Wenyuan Zhu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Delan Meng
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Weiyi Hu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Siqi Yang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Mingjie Gao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Pengju Yao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Yuhao Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| |
Collapse
|
33
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
34
|
Zhao Y, Fan B, Song X, Gao J, Guo R, Yi C, He Z, Hu H, Jiang J, Zhao L, Zhong T, Li B. PEDV-spike-protein-expressing mRNA vaccine protects piglets against PEDV challenge. mBio 2024; 15:e0295823. [PMID: 38231557 PMCID: PMC10865985 DOI: 10.1128/mbio.02958-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/06/2023] [Indexed: 01/18/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), a swine enteropathogenic coronavirus, causes severe diarrhea in neonatal piglets, which is associated with a high mortality rate. Thus, developing effective and safe vaccines remains a top priority for controlling PEDV infection. Here, we designed two lipid nanoparticle (LNP)-encapsulated mRNA (mRNA-LNP) vaccines encoding either the full-length PEDV spike (S) protein or a multiepitope chimeric spike (Sm) protein. We found that the S mRNA-LNP vaccine was superior to the Sm mRNA-LNP vaccine at inducing antibody and cellular immune responses in mice. Evaluation of the immunogenicity and efficacy of the S mRNA vaccine in piglets confirmed that it induced robust PEDV-specific humoral and cellular immune responses in vivo. Importantly, the S mRNA-LNP vaccine not only protected actively immunized piglets against PEDV but also equipped neonatal piglets with effective passive anti-PEDV immunity in the form of colostrum-derived antibodies after the immunization of sows. Our findings suggest that the PEDV-S mRNA-LNP vaccine is a promising candidate for combating PEDV infection.IMPORTANCEPorcine epidemic diarrhea virus (PEDV) continues to harm the global swine industry. It is important to develop a highly effective vaccine to control PEDV infection. Here, we report a PEDV spike (S) mRNA vaccine that primes a potent antibody response and antigen-specific T-cell responses in immunized piglets. Active and passive immunization can protect piglets against PED following the virus challenge. This study highlights the efficiency of the PEDV-S mRNA vaccine and represents a viable approach for developing an efficient PEDV vaccine.
Collapse
Affiliation(s)
- Yongxiang Zhao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Xu Song
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
| | - Jie Gao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Cheng Yi
- Suzhou Huiliao Biomedical Technology Co. Ltd., Suzhou, China
| | - Zhaoming He
- Suzhou Huiliao Biomedical Technology Co. Ltd., Suzhou, China
| | - Hongpeng Hu
- Suzhou Huiliao Biomedical Technology Co. Ltd., Suzhou, China
| | - Jianhao Jiang
- Suzhou Huiliao Biomedical Technology Co. Ltd., Suzhou, China
| | - Lixiang Zhao
- Suzhou Huiliao Biomedical Technology Co. Ltd., Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Tianyi Zhong
- Suzhou Huiliao Biomedical Technology Co. Ltd., Suzhou, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| |
Collapse
|
35
|
ElAbd H, Franke A. Mass Spectrometry-Based Immunopeptidomics of Peptides Presented on Human Leukocyte Antigen Proteins. Methods Mol Biol 2024; 2758:425-443. [PMID: 38549028 DOI: 10.1007/978-1-0716-3646-6_23] [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: 04/02/2024]
Abstract
Human leukocyte antigen (HLA) proteins are a group of glycoproteins that are expressed at the cell surface, where they present peptides to T cells through physical interactions with T-cell receptors (TCRs). Hence, characterizing the set of peptides presented by HLA proteins, referred to hereafter as the immunopeptidome, is fundamental for neoantigen identification, immunotherapy, and vaccine development. As a result, different methods have been used over the years to identify peptides presented by HLA proteins, including competition assays, peptide microarrays, and yeast display systems. Nonetheless, over the last decade, mass spectrometry-based immunopeptidomics (MS-immunopeptidomics) has emerged as the gold-standard method for identifying peptides presented by HLA proteins. MS-immunopeptidomics enables the direct identification of the immunopeptidome in different tissues and cell types in different physiological and pathological states, for example, solid tumors or virally infected cells. Despite its advantages, it is still an experimentally and computationally challenging technique with different aspects that need to be considered before planning an MS-immunopeptidomics experiment, while conducting the experiment and with analyzing and interpreting the results. Hence, we aim in this chapter to provide an overview of this method and discuss different practical considerations at different stages starting from sample collection until data analysis. These points should aid different groups aiming at utilizing MS-immunopeptidomics, as well as, identifying future research directions to improve the method.
Collapse
Affiliation(s)
- Hesham ElAbd
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany.
| |
Collapse
|
36
|
Kola NS, Patel D, Thakur A. RNA-Based Vaccines and Therapeutics Against Intracellular Pathogens. Methods Mol Biol 2024; 2813:321-370. [PMID: 38888787 DOI: 10.1007/978-1-0716-3890-3_21] [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: 06/20/2024]
Abstract
RNA-based vaccines have sparked a paradigm shift in the treatment and prevention of diseases by nucleic acid medicines. There has been a notable surge in the development of nucleic acid therapeutics and vaccines following the global approval of the two messenger RNA-based COVID-19 vaccines. This growth is fueled by the exploration of numerous RNA products in preclinical stages, offering several advantages over conventional methods, i.e., safety, efficacy, scalability, and cost-effectiveness. In this chapter, we provide an overview of various types of RNA and their mechanisms of action for stimulating immune responses and inducing therapeutic effects. Furthermore, this chapter delves into the varying delivery systems, particularly emphasizing the use of nanoparticles to deliver RNA. The choice of delivery system is an intricate process involved in developing nucleic acid medicines that significantly enhances their stability, biocompatibility, and site-specificity. Additionally, this chapter sheds light on the current landscape of clinical trials of RNA therapeutics and vaccines against intracellular pathogens.
Collapse
Affiliation(s)
- Naga Suresh Kola
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Dhruv Patel
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Aneesh Thakur
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
37
|
Mayer RL, Mechtler K. Immunopeptidomics in the Era of Single-Cell Proteomics. BIOLOGY 2023; 12:1514. [PMID: 38132340 PMCID: PMC10740491 DOI: 10.3390/biology12121514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
Immunopeptidomics, as the analysis of antigen peptides being presented to the immune system via major histocompatibility complexes (MHC), is being seen as an imperative tool for identifying epitopes for vaccine development to treat cancer and viral and bacterial infections as well as parasites. The field has made tremendous strides over the last 25 years but currently still faces challenges in sensitivity and throughput for widespread applications in personalized medicine and large vaccine development studies. Cutting-edge technological advancements in sample preparation, liquid chromatography as well as mass spectrometry, and data analysis, however, are currently transforming the field. This perspective showcases how the advent of single-cell proteomics has accelerated this transformation of immunopeptidomics in recent years and will pave the way for even more sensitive and higher-throughput immunopeptidomics analyses.
Collapse
Affiliation(s)
- Rupert L. Mayer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, 1030 Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, 1030 Vienna, Austria
- Gregor Mendel Institute of Molecular Plant Biology (GMI), Austrian Academy of Sciences, Vienna BioCenter (VBC), 1030 Vienna, Austria
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter (VBC), 1030 Vienna, Austria
| |
Collapse
|
38
|
Otálora-Otálora BA, López-Rivera JJ, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Host Transcriptional Regulatory Genes and Microbiome Networks Crosstalk through Immune Receptors Establishing Normal and Tumor Multiomics Metafirm of the Oral-Gut-Lung Axis. Int J Mol Sci 2023; 24:16638. [PMID: 38068961 PMCID: PMC10706695 DOI: 10.3390/ijms242316638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
The microbiome has shown a correlation with the diet and lifestyle of each population in health and disease, the ability to communicate at the cellular level with the host through innate and adaptative immune receptors, and therefore an important role in modulating inflammatory process related to the establishment and progression of cancer. The oral cavity is one of the most important interaction windows between the human body and the environment, allowing the entry of an important number of microorganisms and their passage across the gastrointestinal tract and lungs. In this review, the contribution of the microbiome network to the establishment of systemic diseases like cancer is analyzed through their synergistic interactions and bidirectional crosstalk in the oral-gut-lung axis as well as its communication with the host cells. Moreover, the impact of the characteristic microbiota of each population in the formation of the multiomics molecular metafirm of the oral-gut-lung axis is also analyzed through state-of-the-art sequencing techniques, which allow a global study of the molecular processes involved of the flow of the microbiota environmental signals through cancer-related cells and its relationship with the establishment of the transcription factor network responsible for the control of regulatory processes involved with tumorigenesis.
Collapse
Affiliation(s)
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Carlos Arturo Álvarez-Moreno
- Infectious Diseases Department, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| |
Collapse
|
39
|
Zhang S, Yang H, Wang M, Mantovani D, Yang K, Witte F, Tan L, Yue B, Qu X. Immunomodulatory biomaterials against bacterial infections: Progress, challenges, and future perspectives. Innovation (N Y) 2023; 4:100503. [PMID: 37732016 PMCID: PMC10507240 DOI: 10.1016/j.xinn.2023.100503] [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] [Received: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Bacterial infectious diseases are one of the leading causes of death worldwide. Even with the use of multiple antibiotic treatment strategies, 4.95 million people died from drug-resistant bacterial infections in 2019. By 2050, the number of deaths will reach 10 million annually. The increasing mortality may be partly due to bacterial heterogeneity in the infection microenvironment, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants. In addition, the complexity of the immune microenvironment at different stages of infection makes biomaterials with direct antimicrobial activity unsatisfactory for the long-term treatment of chronic bacterial infections. The increasing mortality may be partly attributed to the biomaterials failing to modulate the active antimicrobial action of immune cells. Therefore, there is an urgent need for effective alternatives to treat bacterial infections. Accordingly, the development of immunomodulatory antimicrobial biomaterials has recently received considerable interest; however, a comprehensive review of their research progress is lacking. In this review, we focus mainly on the research progress and future perspectives of immunomodulatory antimicrobial biomaterials used at different stages of infection. First, we describe the characteristics of the immune microenvironment in the acute and chronic phases of bacterial infections. Then, we highlight the immunomodulatory strategies for antimicrobial biomaterials at different stages of infection and their corresponding advantages and disadvantages. Moreover, we discuss biomaterial-mediated bacterial vaccines' potential applications and challenges for activating innate and adaptive immune memory. This review will serve as a reference for future studies to develop next-generation immunomodulatory biomaterials and accelerate their translation into clinical practice.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Frank Witte
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, Charite Medical University, Assmannshauser Strasse 4–6, 14197 Berlin, Germany
| | - Lili Tan
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
40
|
Wang C, Shen Y, Ma Y. Bifidobacterium infantis-Mediated Herpes Simplex Virus-TK/Ganciclovir Treatment Inhibits Cancer Metastasis in Mouse Model. Int J Mol Sci 2023; 24:11721. [PMID: 37511481 PMCID: PMC10380465 DOI: 10.3390/ijms241411721] [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: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Previous studies have found that Bifidobacterium infantis-mediated herpes simplex virus-TK/ganciclovir (BF-TK/GCV) reduces the expression of VEGF and CD146, implying tumor metastasis inhibition. However, the mechanism by which BF-TK/GCV inhibits tumor metastasis is not fully studied. Here, we comprehensively identified and quantified protein expression profiling for the first time in gastric cancer (GC) cells MKN-45 upon BF-TK/GCV treatment using quantitative proteomics. A total of 159 and 72 differential expression proteins (DEPs) were significantly changed in the BF-TK/GCV/BF-TK and BF-TK/GCV/BF/GCV comparative analysis. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis enriched some metastasis-related pathways such as gap junction and cell adhesion molecules pathways. Moreover, the transwell assay proved that BF-TK/GCV inhibited the invasion and migration of tumor cells. Furthermore, immunohistochemistry (IHC) demonstrated that BF-TK/GCV reduced the expression of HIF-1α, mTOR, NF-κB1-p105, VCAM1, MMP13, CXCL12, ATG16, and CEBPB, which were associated with tumor metastasis. In summary, BF-TK/GCV inhibited tumor metastasis, which deepened and expanded the understanding of the antitumor mechanism of BF-TK/GCV.
Collapse
Affiliation(s)
- Changdong Wang
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
41
|
Song C, Wang B, Wang Y, Liu J, Wang D. Detection of Listeria monocytogenes in Food Using the Proofman-LMTIA Assay. Molecules 2023; 28:5457. [PMID: 37513329 PMCID: PMC10385859 DOI: 10.3390/molecules28145457] [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: 06/27/2023] [Revised: 07/09/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Microbial factors, including bacteria, viruses, and other pathogens, are significant contributors to foodborne illnesses, posing serious food safety risks due to their potential for rapid growth and contamination. Listeria monocytogenes is one of the most common types of foodborne bacteria that can cause serious foodborne diseases or even fatalities. In this study, a novel nucleic acid amplification method called Proofman-LMTIA was employed to detect Listeria monocytogenes contamination in food. This method combines proofreading enzyme-mediated probe cleavage with ladder-shape melting temperature isothermal amplification. A positive recombinant plasmid was used as a control to ensure the accuracy of the detection results, and primers and Proofman probes were specifically designed for the LMTIA. Genomic DNA was extracted, the reaction temperature was optimized, and the primers' specificity was verified using foodborne pathogens like Staphylococcus aureus, Escherichia coli O157:H7, and Salmonella. The sensitivity was assessed by testing serial dilutions of genomic DNA, and the method's applicability was confirmed by detecting artificially contaminated fresh pork. The established LMTIA method exhibited both high specificity and sensitivity. At the optimal reaction temperature of 63 °C, the primers specifically identified Listeria monocytogenes contamination in pork at a concentration of 8.0 ± 0.7 colony-forming units (CFUs) per 25 g. Furthermore, the Proofman-LMTIA method was applied to test Listeria monocytogenes DNA in 30 food samples purchased from a Chinese retail market, and reassuringly, all results indicated no contamination. Proofman-LMTIA can serve as a reliable and rapid method for detecting Listeria monocytogenes in food, contributing to public health by safeguarding consumers from foodborne illnesses, and strengthening food safety regulations.
Collapse
Affiliation(s)
- Chunmei Song
- Key Laboratory of Biomarker Based Rapid-Detection Technology for Food Safety of Henan Province, Xuchang University, Xuchang 461000, China
| | - Borui Wang
- School of Food and Biological Engineering, Henan University of Science and Technology, Luoyang 471000, China
| | - Yongzhen Wang
- Key Laboratory of Biomarker Based Rapid-Detection Technology for Food Safety of Henan Province, Xuchang University, Xuchang 461000, China
| | - Jinxin Liu
- Key Laboratory of Biomarker Based Rapid-Detection Technology for Food Safety of Henan Province, Xuchang University, Xuchang 461000, China
| | - Deguo Wang
- Key Laboratory of Biomarker Based Rapid-Detection Technology for Food Safety of Henan Province, Xuchang University, Xuchang 461000, China
| |
Collapse
|
42
|
Wang K, Zhou W, Jin X, Shang X, Wu X, Wen L, Li S, Hong Y, Ke J, Xu Y, Yuan H, Hu F. Enhanced brain delivery of hypoxia-sensitive liposomes by hydroxyurea for rescue therapy of hyperacute ischemic stroke. NANOSCALE 2023. [PMID: 37377137 DOI: 10.1039/d3nr01071f] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Ischemic stroke is characterized by high morbidity, disability, and mortality. Unfortunately, the only FDA-approved pharmacological thrombolytic, alteplase, has a narrow therapeutic window of only 4.5 h. Other drugs like neuroprotective agents have not been clinically used because of their low efficacy. To improve the efficacy of neuroprotective agents and the effectiveness of rescue therapies for hyperacute ischemic stroke, we investigated and verified the variation trends of the blood-brain barrier (BBB) permeability and regional cerebral blood flow over 24 h in rats that had ischemic strokes. Hypoperfusion and the biphasic increase of BBB permeability are still the main limiting factors for lesion-specific drug distribution and drug brain penetration. Herein, the nitric oxide donor hydroxyurea (HYD) was reported to downregulate the expression of tight junction proteins and upregulate intracellular nitric oxide content in the brain microvascular endothelial cells subjected to oxygen-glucose deprivation, which was shown to facilitate the transport of liposomes across brain endothelial monolayer in an in vitro model. HYD also increased the BBB permeability and promoted microcirculation in the hyperacute phase of stroke. The neutrophil-like cell-membrane-fusogenic hypoxia-sensitive liposomes exhibited excellent performance in targeting the inflamed brain microvascular endothelial cells, enhancing cell association, and promoting rapid hypoxic-responsive release in the hypoxic microenvironment. Overall, the combined HYD and hypoxia-sensitive liposome dosing regimen effectively decreased the cerebral infarction volume and relieved neurological dysfunction in rats that had ischemic strokes; these therapies were involved in the anti-oxidative stress effect and the neurotrophic effect mediated by macrophage migration inhibitory factor.
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Wentao Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiangyu Jin
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xuwei Shang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Xiaomei Wu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Lijuan Wen
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Sufen Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Yiling Hong
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Yichong Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
43
|
Lim Kam Sian TCC, Goncalves G, Steele JR, Shamekhi T, Bramberger L, Jin D, Shahbazy M, Purcell AW, Ramarathinam S, Stoychev S, Faridi P. SAPrIm, a semi-automated protocol for mid-throughput immunopeptidomics. Front Immunol 2023; 14:1107576. [PMID: 37334365 PMCID: PMC10272402 DOI: 10.3389/fimmu.2023.1107576] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Human leukocyte antigen (HLA) molecules play a crucial role in directing adaptive immune responses based on the nature of their peptide ligands, collectively coined the immunopeptidome. As such, the study of HLA molecules has been of major interest in the development of cancer immunotherapies such as vaccines and T-cell therapies. Hence, a comprehensive understanding and profiling of the immunopeptidome is required to foster the growth of these personalised solutions. We herein describe SAPrIm, an Immunopeptidomics tool for the Mid-Throughput era. This is a semi-automated workflow involving the KingFisher platform to isolate immunopeptidomes using anti-HLA antibodies coupled to a hyper-porous magnetic protein A microbead, a variable window data independent acquisition (DIA) method and the ability to run up to 12 samples in parallel. Using this workflow, we were able to concordantly identify and quantify ~400 - 13000 unique peptides from 5e5 - 5e7 cells, respectively. Overall, we propose that the application of this workflow will be crucial for the future of immunopeptidome profiling, especially for mid-size cohorts and comparative immunopeptidomics studies.
Collapse
Affiliation(s)
- Terry C. C. Lim Kam Sian
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Gabriel Goncalves
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Joel R. Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tima Shamekhi
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
| | - Liesl Bramberger
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
| | - Dongbin Jin
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
| | - Mohammad Shahbazy
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sri Ramarathinam
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
44
|
Montero DA, Vidal RM, Velasco J, George S, Lucero Y, Gómez LA, Carreño LJ, García-Betancourt R, O’Ryan M. Vibrio cholerae, classification, pathogenesis, immune response, and trends in vaccine development. Front Med (Lausanne) 2023; 10:1155751. [PMID: 37215733 PMCID: PMC10196187 DOI: 10.3389/fmed.2023.1155751] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/14/2023] [Indexed: 05/24/2023] Open
Abstract
Vibrio cholerae is the causative agent of cholera, a highly contagious diarrheal disease affecting millions worldwide each year. Cholera is a major public health problem, primarily in countries with poor sanitary conditions and regions affected by natural disasters, where access to safe drinking water is limited. In this narrative review, we aim to summarize the current understanding of the evolution of virulence and pathogenesis of V. cholerae as well as provide an overview of the immune response against this pathogen. We highlight that V. cholerae has a remarkable ability to adapt and evolve, which is a global concern because it increases the risk of cholera outbreaks and the spread of the disease to new regions, making its control even more challenging. Furthermore, we show that this pathogen expresses several virulence factors enabling it to efficiently colonize the human intestine and cause cholera. A cumulative body of work also shows that V. cholerae infection triggers an inflammatory response that influences the development of immune memory against cholera. Lastly, we reviewed the status of licensed cholera vaccines, those undergoing clinical evaluation, and recent progress in developing next-generation vaccines. This review offers a comprehensive view of V. cholerae and identifies knowledge gaps that must be addressed to develop more effective cholera vaccines.
Collapse
Affiliation(s)
- David A. Montero
- Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto M. Vidal
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juliana Velasco
- Unidad de Paciente Crítico, Clínica Hospital del Profesor, Santiago, Chile
- Programa de Formación de Especialista en Medicina de Urgencia, Universidad Andrés Bello, Santiago, Chile
| | - Sergio George
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Yalda Lucero
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Pediatría y Cirugía Infantil, Hospital Dr. Roberto del Rio, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Leonardo A. Gómez
- Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Leandro J. Carreño
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Richard García-Betancourt
- Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Miguel O’Ryan
- Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
45
|
Feola S, Chiaro J, Cerullo V. Integrating immunopeptidome analysis for the design and development of cancer vaccines. Semin Immunol 2023; 67:101750. [PMID: 37003057 DOI: 10.1016/j.smim.2023.101750] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
The repertoire of naturally presented peptides within the MHC (major histocompatibility complex) or HLA (human leukocyte antigens) system on the cellular surface of every mammalian cell is referred to as ligandome or immunopeptidome. This later gained momentum upon the discovery of CD8 + T cells able to recognize and kill cancer cells in an MHC-I antigen-restricted manner. Indeed, cancer immune surveillance relies on T cell recognition of MHC-I-restricted peptides, making the identification of those peptides the core for designing T cell-based cancer vaccines. Moreover, the breakthrough of antibodies targeting immune checkpoint molecules has led to a new and strong interest in discovering suitable targets for CD8 +T cells. Therapeutic cancer vaccines are designed for the artificial generation and/or stimulation of CD8 +T cells; thus, their combination with ICIs to unleash the breaks of the immune system comes as a natural consequence to enhance anti-tumor efficacy. In this context, the identification and knowledge of peptide candidates take advantage of the fast technology updates in immunopeptidome and mass spectrometric methodologies, paying the way to the rational design of vaccines for immunotherapeutic approaches. In this review, we discuss mainly the role of immunopeptidome analysis and its application for the generation of therapeutic cancer vaccines with main focus on HLA-I peptides. Here, we review cancer vaccine platforms based on two different preparation methods: pathogens (viruses and bacteria) and not (VLPs, nanoparticles, subunits vaccines) that exploit discoveries in the ligandome field to generate and/or enhance anti-tumor specific response. Finally, we discuss possible drawbacks and future challenges in the field that remain still to be addressed.
Collapse
Affiliation(s)
- Sara Feola
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy Helsinki University, Viikinkaari 5E, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Fabianinkatu 33, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, Haartmaninkatu 8, Finland
| | - Jacopo Chiaro
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy Helsinki University, Viikinkaari 5E, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Fabianinkatu 33, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, Haartmaninkatu 8, Finland
| | - Vincenzo Cerullo
- Drug Research Program (DRP) ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy Helsinki University, Viikinkaari 5E, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Fabianinkatu 33, Finland; Translational Immunology Program (TRIMM), Faculty of Medicine Helsinki University, Haartmaninkatu 8, Finland; Department of Molecular Medicine and Medical Biotechnology, Naples University "Federico II", S. Pansini 5, Italy.
| |
Collapse
|
46
|
ElAbd H, Bacher P, Tholey A, Lenz TL, Franke A. Challenges and opportunities in analyzing and modeling peptide presentation by HLA-II proteins. Front Immunol 2023; 14:1107266. [PMID: 37063883 PMCID: PMC10090296 DOI: 10.3389/fimmu.2023.1107266] [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: 11/24/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
The human leukocyte antigen (HLA) proteins are an indispensable component of adaptive immunity because of their role in presenting self and foreign peptides to T cells. Further, many complex diseases are associated with genetic variation in the HLA region, implying an important role for specific HLA-presented peptides in the etiology of these diseases. Identifying the specific set of peptides presented by an individual’s HLA proteins in vivo, as a whole being referred to as the immunopeptidome, has therefore gathered increasing attention for different reasons. For example, identifying neoepitopes for cancer immunotherapy, vaccine development against infectious pathogens, or elucidating the role of HLA in autoimmunity. Despite the tremendous progress made during the last decade in these areas, several questions remain unanswered. In this perspective, we highlight five remaining key challenges in the analysis of peptide presentation and T cell immunogenicity and discuss potential solutions to these problems. We believe that addressing these questions would not only improve our understanding of disease etiology but will also have a direct translational impact in terms of engineering better vaccines and in developing more potent immunotherapies.
Collapse
Affiliation(s)
- Hesham ElAbd
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Petra Bacher
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Andreas Tholey
- Proteomics & Bioanalytics, Institute for Experimental Medicine, University of Kiel, Kiel, Germany
| | - Tobias L. Lenz
- Research Unit for Evolutionary Immunogenomics, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
- *Correspondence: Andre Franke,
| |
Collapse
|
47
|
Kon E, Levy Y, Elia U, Cohen H, Hazan-Halevy I, Aftalion M, Ezra A, Bar-Haim E, Naidu GS, Diesendruck Y, Rotem S, Ad-El N, Goldsmith M, Mamroud E, Peer D, Cohen O. A single-dose F1-based mRNA-LNP vaccine provides protection against the lethal plague bacterium. SCIENCE ADVANCES 2023; 9:eadg1036. [PMID: 36888708 PMCID: PMC9995031 DOI: 10.1126/sciadv.adg1036] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 05/28/2023]
Abstract
Messenger RNA (mRNA) lipid nanoparticle (LNP) vaccines have emerged as an effective vaccination strategy. Although currently applied toward viral pathogens, data concerning the platform's effectiveness against bacterial pathogens are limited. Here, we developed an effective mRNA-LNP vaccine against a lethal bacterial pathogen by optimizing mRNA payload guanine and cytosine content and antigen design. We designed a nucleoside-modified mRNA-LNP vaccine based on the bacterial F1 capsule antigen, a major protective component of Yersinia pestis, the etiological agent of plague. Plague is a rapidly deteriorating contagious disease that has killed millions of people during the history of humankind. Now, the disease is treated effectively with antibiotics; however, in the case of a multiple-antibiotic-resistant strain outbreak, alternative countermeasures are required. Our mRNA-LNP vaccine elicited humoral and cellular immunological responses in C57BL/6 mice and conferred rapid, full protection against lethal Y. pestis infection after a single dose. These data open avenues for urgently needed effective antibacterial vaccines.
Collapse
Affiliation(s)
- Edo Kon
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yinon Levy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Uri Elia
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Hila Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Assaf Ezra
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Gonna Somu Naidu
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yael Diesendruck
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shahar Rotem
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Nitay Ad-El
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Meir Goldsmith
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Emanuelle Mamroud
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ofer Cohen
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| |
Collapse
|
48
|
Pharmaceuticals and Personal Care Products in the Environment with Emphasis on Horizontal Transfer of Antibiotic Resistance Genes. CHEMISTRY-DIDACTICS-ECOLOGY-METROLOGY 2022. [DOI: 10.2478/cdem-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Abstract
Pharmaceuticals and personal care products (PPCPs) discharged into environment has several adverse impacts. PPCPs are widely utilised for veterinary as well as cosmetic and personal health reasons. These are members of the expanding class of substances known as Contaminants of Emerging Concern (CECs). Antibiotic resistance in the environment and garbage generated by PPCP endanger life. The World Health Organisation (WHO) now recognises antibiotic resistance as a significant global health problem due to the expected increase in mortality caused by it. In the past ten years, mounting data has led experts to believe that the environment has a significant impact on the development of resistance. For human diseases, the external environment serves as a source of resistance genes. It also serves as a major pathway for the spread of resistant bacteria among various habitats and human populations. Large-scale DNA sequencing methods are employed in this thesis to better comprehend the dangers posed by environmental antibiotic resistance. The quantification of the number is an important step in this process. Metagenomic measurement of the number of antibiotic resistance genes in various contexts is a crucial step in this process. However, it’s also crucial to put this data into a broader context by integrating things like taxonomic information, antibiotic concentrations, and the genomic locations of found resistance genes.
Collapse
|