1
|
Sun N, Su Z, Zheng X. Research progress of mosquito-borne virus mRNA vaccines. Mol Ther Methods Clin Dev 2025; 33:101398. [PMID: 39834558 PMCID: PMC11743085 DOI: 10.1016/j.omtm.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
In recent years, mRNA vaccines have emerged as a leading technology for preventing infectious diseases due to their rapid development and high immunogenicity. These vaccines encode viral antigens, which are translated into antigenic proteins within host cells, inducing both humoral and cellular immune responses. This review systematically examines the progress in mRNA vaccine research for major mosquito-borne viruses, including dengue virus, Zika virus, Japanese encephalitis virus, Chikungunya virus, yellow fever virus, Rift Valley fever virus, and Venezuelan equine encephalitis virus. Enhancements in mRNA vaccine design, such as improvements to the 5' cap structure, 5'UTR, open reading frame, 3'UTR, and polyadenylation tail, have significantly increased mRNA stability and translation efficiency. Additionally, the use of lipid nanoparticles and polymer nanoparticles has greatly improved the delivery efficiency of mRNA vaccines. Currently, mRNA vaccines against mosquito-borne viruses are under development and clinical trials, showing promising protective effects. Future research should continue to optimize vaccine design and delivery systems to achieve broad-spectrum and long-lasting protection against various mosquito-borne virus infections.
Collapse
Affiliation(s)
- Ningze Sun
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Zhiwei Su
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Xiaoyan Zheng
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| |
Collapse
|
2
|
Cui J, Hu Z, Jiang Y, Wang Y, Li C, Zhang S, Chen L, Zhang Z, Yang D, Shen H, Zheng P, Qiu L, Lu Z. Jiawei Yanghe Decoction alleviates pulmonary sarcoidosis by upregulating NR1D1/2 and suppressing Th17 cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119372. [PMID: 39826790 DOI: 10.1016/j.jep.2025.119372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/03/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei Yanghe Decoction (JWYHD) is a modified version traditional Chinese medicine formula Yanghe Decoction which has been used to treat various autoimmune diseases. However, the effect of JWYHD on pulmonary sarcoidosis remains unclear. AIM OF THE STUDY This study aimed to determine the therapeutic efficacy and potential mechanism of action of JWYHD in pulmonary sarcoidosis. MATERIALS AND METHODS A murine model of sarcoidosis was established by intravenous injection of inactivated Propionibacterium acnes and mature dendritic cells to assess the efficacy of JWYHD. Lung tissue mRNA sequencing was conducted to identify the targets of JWYHD's action. Molecular docking verified of the interaction between identified compounds and key targets. RESULTS JWYHD treatment alleviated the formation of granulomas in the lung tissue of sarcoidosis model mice. JWYHD significantly attenuated the pulmonary accumulation of macrophages and CD4+T lymphocytes in sarcoidosis mice, and effectively suppressed the proportion of Th17 cells and the levels of IL-17A and TNF-α in BALF, which are pivotal in the pathogenesis of granuloma formation and progression. The therapeutic efficacy of JWYHD was found to be equivalent to that of prednisone. RNA-seq revealed that JWYHD upregulated Nr1d1/2 expression in the lung tissue. Nr1d1/2 is highly expressed in Th17 cells and regulates their differentiation. The NR1D1/2 agonist SR9009 could inhibit Th17 cell proportion and reduce the formation of pulmonary granuloma, exhibiting effects similar to those of JWYHD. Molecular docking result showed that Cyclocephaloside II, Epimedin B, Glycyrrhetic acid, Glycyrrhizic acid, Uralsaponin B, and Uralsaponin U may be key compounds in JWYHD for the treatment of pulmonary sarcoidosis, which had a strong binding ability for NR1D1/2. CONCLUSIONS JWYHD might exert a therapeutic benefit in pulmonary sarcoidosis through upregulating NR1D1/2 and suppressing Th17 cells. NR1D1/2 might serve as a therapeutic target for the treatment of pulmonary sarcoidosis.
Collapse
Affiliation(s)
- Jie Cui
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhuannan Hu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuwei Jiang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yu Wang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Cui Li
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shaoyan Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Linjin Chen
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhengyi Zhang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Di Yang
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Huimin Shen
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Peiyong Zheng
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lei Qiu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Zhenhui Lu
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
3
|
Cebrian I, Dinamarca S, Pena Rodríguez MJ, Priego E, Brouwers N, Barends M, Brunnberg J, Tampé R, Blanchard N, Sancho D, Malhotra V. Dendritic cell phagosomes recruit GRASP55 for export of antigen-loaded MHC molecules. Cell Rep 2025; 44:115333. [PMID: 39955774 DOI: 10.1016/j.celrep.2025.115333] [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: 03/13/2024] [Revised: 10/14/2024] [Accepted: 01/28/2025] [Indexed: 02/18/2025] Open
Abstract
Dendritic cells (DCs) present exogenous antigens via major histocompatibility complex class I (MHC-I) and MHC class II (MHC-II) molecules, activating CD8+ and CD4+ T cells. A critical but poorly understood step in this process is the trafficking of peptide-loaded MHC molecules from the endocytic system to the cell surface. In this study, we demonstrate that the Golgi reassembly-stacking protein of 55 kDa (GRASP55), which has been shown to have no role in stacking, is essential for antigen presentation. Using soluble, bead-coated, and bacterial-bound antigens, we found significantly impaired exogenous antigen presentation in GRASP55-deficient bone-marrow-derived DCs (BMDCs). Notably, GRASP55 was recruited to late phagosomes, and our data suggest that it is crucial for sorting MHC-I and MHC-II molecules, facilitating their trafficking to the plasma membrane. Our findings highlight the vital role of GRASP55 in the intracellular transport of MHC molecules bound to their respective peptides during exogenous antigen presentation.
Collapse
Affiliation(s)
- Ignacio Cebrian
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina.
| | - Sofía Dinamarca
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina
| | - María Jesús Pena Rodríguez
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Elena Priego
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Nathalie Brouwers
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Martina Barends
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Jamina Brunnberg
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Nicolas Blanchard
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM/CNRS/Université Toulouse 3, 31300 Toulouse, France
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Vivek Malhotra
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
4
|
Gachpazan M, Alashti AA, Jahantigh HR, Moghbeli M, Faezi S, Hosseini SY, Eftekharian MM, Nasimi M, Khiavi FM, Rahimi A, Mianroodi RA, Pakjoo M, Taghizadeh M, Tempesta M, Mahdavi M. Immunization with recombinant HPV16-E7d in fusion with Flagellin as a cancer vaccine: Effect of antigen-adjuvant orientation on the immune response pattern. Immunol Res 2025; 73:50. [PMID: 39939497 DOI: 10.1007/s12026-025-09598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/19/2025] [Indexed: 02/14/2025]
Abstract
Human papillomavirus (HPV) is the leading cause of cervical cancer worldwide. The pathogenesis of HPV is mainly dependent on its E7 and E6 proteins. Up to now, different adjuvants have been used to enhance the efficacy of the immune response against these two proteins. In this study, Flagellin (FLA) was used as adjuvant to test adjuvant activity and also see whether its orientation of attachment can affect the immune response pattern. The E7d-FLA and FLA-E7d in pET28a vector were constructed and then the recombinant proteins were expressed in E. coli BL21 (DE3) bacteria under IPTG induction. The expression of recombinant E7d-FLA and FLA-E7d proteins is confirmed by SDS-PAGE and western blot. Then, recombinant fusion proteins were purified using a nickel-nitrilotriacetic acid (Ni-NTA) column. The recombinant proteins were checked for endotoxin contamination and then quantified by Bradford. Eight-to-ten-week-old male Balb/C mice were immunized subcutaneously with 10 µg recombinant E7d-FLA, FLA-E7d and HPV16E7d vaccine on days 0, 14 and 28. In addition, PBS and FLA groups were considered as control group. Then, spleen cells were harvested to assess lymphocyte proliferation and IFN-γ, IL-4 and IL-17 cytokines. In addition, mice sera were used for specific total IgG and IgG1, IgG2a, IgG2b and IgM antibodies assessment by ELISA. The results show that E7d-FLA is more potent in the induction of lymphocyte proliferation, CTL response and specific total IgG, IgG2a and IgG2b response, while the FLA-E7d vaccine was associated with more IFN-γ, and IL-17 cytokine response. The results of this study proved the ability of FLA as an adjuvant in fusion with E7d in the induction of cellular and humoral immune responses. In addition, it also emphasizes that antigen-adjuvant orientation can affect the immune response strength and polarization against HPV E7d vaccine candidate. HIGHLIGHTS: Flagellin is attached to HPV-16 E7d at the C- or N-terminus to create E7d-FLA and FLA-E7d candidate vaccines. The E7d-FLA vaccine showed a significant increase in lymphocyte proliferation, CTL response and IgG response versus FLA-E7d vaccine. The FLA-E7d vaccine is associated with a significant increase in IFN-γ and IL-17 cytokines response versus E7d-FLA vaccine. It seems that that antigen-adjuvant orientation is an important parameter in the strength and polarization of immune response in HPV E7d vaccine candidate.
Collapse
Affiliation(s)
- Meysam Gachpazan
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran
- Department of Biology, Islamic Azad University of Damghan Branch, Damghan, Iran
| | - Ali Ahmadnia Alashti
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran
| | - Hamid Reza Jahantigh
- Department of Pathology, Faculty of Medicine, Emory University, Atlanta, GA, 30033, USA
- Interdisciplinary Department of Medicine - Section of Occupational Medicine, University of Bari, Bari, Italy
| | - Majid Moghbeli
- Department of Biology, Islamic Azad University of Damghan Branch, Damghan, Iran
| | - Sobhan Faezi
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Maryam Nasimi
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Motavalli Khiavi
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Etemad Zadeh Street, Fatemi-Gharbi Street, Tehran, Iran
| | - Alireza Rahimi
- Department of Recombinant Products, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Arabi Mianroodi
- Department of Research and Development, Research and Production Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Pakjoo
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran
| | - Morteza Taghizadeh
- Department of Medical Vaccine, Agricultural Research, Education and Extension Organization (AREEO), Razi Vaccine and Serum Research Institute, Karaj, Iran.
| | - Maria Tempesta
- Department of Veterinary Medicine, Animal Health and Zoonosis PhD Course, University of Bari, Bari, Italy
| | - Mehdi Mahdavi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center Academic Center for Education, Culture and Research (ACECR)Vanak Sq, Motamed Cancer Institute, South Gandi Ave, P.O. BOX, Tehran, 15179/64311, NO.146, Iran.
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, 16 Azar St, P. O. Box: 1316943551, Tehran, 14174, Iran.
| |
Collapse
|
5
|
Charette M, Rosenblum C, Shade O, Deiters A. Optogenetics with Atomic Precision─A Comprehensive Review of Optical Control of Protein Function through Genetic Code Expansion. Chem Rev 2025. [PMID: 39928721 DOI: 10.1021/acs.chemrev.4c00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Conditional control of protein activity is important in order to elucidate the particular functions and interactions of proteins, their regulators, and their substrates, as well as their impact on the behavior of a cell or organism. Optical control provides a perhaps optimal means of introducing spatiotemporal control over protein function as it allows for tunable, rapid, and noninvasive activation of protein activity in its native environment. One method of introducing optical control over protein activity is through the introduction of photocaged and photoswitchable noncanonical amino acids (ncAAs) through genetic code expansion in cells and animals. Genetic incorporation of photoactive ncAAs at key residues in a protein provides a tool for optical activation, or sometimes deactivation, of protein activity. Importantly, the incorporation site can typically be rationally selected based on structural, mechanistic, or computational information. In this review, we comprehensively summarize the applications of photocaged lysine, tyrosine, cysteine, serine, histidine, glutamate, and aspartate derivatives, as well as photoswitchable phenylalanine analogues. The extensive and diverse list of proteins that have been placed under optical control demonstrates the broad applicability of this methodology.
Collapse
Affiliation(s)
- Maura Charette
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Carolyn Rosenblum
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Olivia Shade
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
6
|
Zhou Y, Li Z, Wang G, Yu H, Zhou Y, Li Y, Chen W, Dai H, He Y, Li L. Understanding the relationship between silicone implants, tumor antigens, and breast cancer risk: An immunological study in rats. Int Immunopharmacol 2025; 147:113991. [PMID: 39805174 DOI: 10.1016/j.intimp.2024.113991] [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: 10/29/2024] [Revised: 12/29/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025]
Abstract
This study aimed to investigate the effects of silicone implants on the incidence of breast cancer in rats, as well as their impact on immune surveillance mechanisms. Female SD rats were divided into three groups: a Placebo Surgery Group (PSG), a Thoracic Implant Group (TIG), and a Back Implant Group (BIG). Following the corresponding surgical procedures, we measured Secretoglobin Family 2A, Member 2(SCGB2A2) and Mucin-1 (MUC1) antigen levels using ELISA, and statistical analyses were conducted to evaluate immune responses. The N-Methyl-N-Nitrosourea(MNU)-induced breast cancer model and pathological analyses indicated that the incidence of breast cancer in the thoracic implant group was lower, suggesting that silicone implants may reduce the risk of breast cancer. Additionally, laser speckle blood flow imaging and immunohistochemical analysis revealed blood perfusion in the implant capsule area and an active response of immune cells, indicating that immune surveillance may exert local effects. These findings provide the first evidence of a relationship between tumor antigens, silicone implants, and breast cancer incidence, offering a new immunological perspective on the safety of silicone implants.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Zihao Li
- Department of First Clinical Medical School, Wenzhou Medical University, Wenzhou, PR China
| | - Gaoyi Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Hua Yu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Yaqin Zhou
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Yijun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Wanying Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Hao Dai
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China; National Key Clinical Specialty (Wound Healing), The First Affiliate d Hospital Of Wenzhou Medical University, PR China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China; National Key Clinical Specialty (Wound Healing), The First Affiliate d Hospital Of Wenzhou Medical University, PR China.
| |
Collapse
|
7
|
Günther M, Sticht J, Freund C, Höfer T. Antigen presentation by MHC-II is shaped by competitive and cooperative allosteric mechanisms of peptide exchange. Structure 2025; 33:389-400.e13. [PMID: 39708815 DOI: 10.1016/j.str.2024.11.014] [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: 04/22/2024] [Revised: 08/12/2024] [Accepted: 11/20/2024] [Indexed: 12/23/2024]
Abstract
Major histocompatibility complex class II (MHC-II) presents antigens to T helper cells. The spectrum of presented peptides is regulated by the exchange catalyst human leukocyte antigen DM (HLA-DM), which dissociates peptide-MHC-II complexes in the endosome. How susceptible a peptide is to HLA-DM is mechanistically not understood. Here, we present a data-driven mathematical model for the conformational landscape of MHC-II that explains the wide range of measured HLA-DM susceptibilities and predicts why some peptides are largely HLA-DM-resistant. We find that the conformational plasticity of MHC-II mediates both allosteric competition and cooperation between peptide and HLA-DM. Competition causes HLA-DM susceptibility to be proportional to the intrinsic peptide off-rate. Remarkably, diverse MHC-II allotypes with conserved HLA-DM interactions show a universal linear susceptibility function. However, HLA-DM-resistant peptides deviate from this susceptibility function; we predict resistance to be caused by fast peptide association with MHC-II. Thus, our study provides quantitative insight into peptide and MHC-II allotype parameters that shape class-II antigen presentation.
Collapse
Affiliation(s)
- Matthias Günther
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Jana Sticht
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Christian Freund
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
8
|
Szabo A, Akkouh I, Osete JR, de Assis DR, Kondratskaya E, Hughes T, Ueland T, Andreassen OA, Djurovic S. NLRP3 inflammasome mediates astroglial dysregulation of innate and adaptive immune responses in schizophrenia. Brain Behav Immun 2025; 124:144-156. [PMID: 39617069 DOI: 10.1016/j.bbi.2024.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Mounting evidence indicates the involvement of neuroinflammation in the development of schizophrenia (SCZ), but the potential role of astroglia in this phenomenon remains poorly understood. We assessed the molecular and functional consequences of inflammasome activation using induced pluripotent stem cell (iPSC)-derived astrocytes generated from SCZ patients and healthy controls (CTRL). Screening protein levels in astrocytes at baseline identified lower expression of the NLRP3-ASC complex in SCZ, but increased Caspase-1 activity upon specific NLRP3 stimulation compared to CTRL. Using transcriptional profiling, we found corresponding downregulations of NLRP3 and ASC/PYCARD in both iPSC-derived astrocytes, and in a large (n = 429) brain postmortem case-control sample. Functional analyses following NLRP3 activation revealed an inflammatory phenotype characterized by elevated production of IL-1β/IL-18 and skewed priming of helper T lymphocytes (Th1/Th17) by SCZ astrocytes. This phenotype was rescued by specific inhibition of NLRP3 activation, demonstrating its dependence on the NLRP3 inflammasome. Taken together, SCZ iPSC-astrocytes display unique, NLRP3-dependent inflammatory characteristics that are manifested via various cellular functions, as well as via dysregulated innate and adaptive immune responses.
Collapse
Affiliation(s)
- Attila Szabo
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway.
| | - Ibrahim Akkouh
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jordi Requena Osete
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Denis Reis de Assis
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Elena Kondratskaya
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Timothy Hughes
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Centre, University of Tromsø, Tromsø, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
9
|
Zhao Y, Song D, Wang Z, Huang Q, Huang F, Ye Z, Wich D, Chen M, Khirallah J, Gao S, Liu Y, Xu Q. Antitumour vaccination via the targeted proteolysis of antigens isolated from tumour lysates. Nat Biomed Eng 2025; 9:234-248. [PMID: 39609559 DOI: 10.1038/s41551-024-01285-5] [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: 11/13/2023] [Accepted: 10/17/2024] [Indexed: 11/30/2024]
Abstract
The activation of cytotoxic T cells against tumour cells typically requires the cross-presentation, by antigen-presenting cells (and via major histocompatibility complex class I molecules), of an epitope derived from a tumour antigen. A critical step in antigen processing is the proteolysis of tumour antigens mediated by the ubiquitin-proteasome pathway. Here we describe a tumour vaccine leveraging targeted antigen degradation to augment antigen processing and cross-presentation. Analogous to proteolysis-targeting chimaeras, the vaccine consists of lymph-node-targeting lipid nanoparticles encapsulated with tumour antigens pre-conjugated with ligands that can bind to E3 ubiquitin ligases. In mice with subcutaneous human melanoma or triple-negative breast cancer, or with orthotopic mouse Lewis lung carcinoma or clinically inoperable mouse ovarian cancer, subcutaneously delivered vaccines prepared using tumour lysate proteins elicited antigen-specific adaptive immunity and immunological memory, and inhibited tumour growth, metastasis and recurrence, particularly when combined with immune checkpoint inhibition.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Donghui Song
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Zeyu Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Qingqing Huang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, China
| | - Fan Huang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Douglas Wich
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Mengting Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Jennifer Khirallah
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Shuliang Gao
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Yang Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, China
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| |
Collapse
|
10
|
Gu X, Chen C, Chen Y, Zeng C, Lin Y, Guo R, Xu S, Lin C. Bioinformatics approach reveals the critical role of inflammation-related genes in age-related hearing loss. Sci Rep 2025; 15:2687. [PMID: 39837906 PMCID: PMC11751394 DOI: 10.1038/s41598-024-83428-x] [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: 06/14/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory impairment in the elderly. However, the pathogenesis of ARHL remains unclear. This study was aimed to explore the potential inflammation-related genes of ARHL and suggest novel therapeutic targets for this condition. Initially, a total of 105 Inflammatory related differentially expressed genes (IRDEGs) were obtained by overlapping the differentially expressed genes from the GSE49522 and GSE49543 datasets with Inflammatory related genes. The IRDEGs were mainly enriched in MAPK, PI3K-Akt, Hippo and JAK-STAT pathways by analysis of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. We then identified 10 key IRDEGs including Alox5ap, Chil1, Clec7a, Dysf, Fcgr3, etc. using Least absolute shrinkage and selection operator regression analysis and converted them into human genes. The ROC curve indicated that Alox5ap expression presented a high accuracy in distinguishing between different groups. By CIBERSORT algorithm, 8 humanized key IRDEGs were correlated with the infiltration abundance of 3 immune cells. Finally, it showed that the Alox5ap expression was significantly more effective compared to other variables in the diagnostic model of ARHL. This study suggests that inflammation might play a role in the development of ARHL, providing a deeper understanding of the underlying causes of this disease.
Collapse
Affiliation(s)
- Xi Gu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chenyu Chen
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Yuqing Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chaojun Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yanchun Lin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruosi Guo
- Fujian Medical University, Fuzhou, China
| | - Shujin Xu
- Fujian Medical University, Fuzhou, China
| | - Chang Lin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Institute of Otolaryngology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
11
|
James LM, Georgopoulos AP. Negative Association of Gulf War Illness Symptomatology with Predicted Binding Affinity of Anthrax Vaccine Antigen to Human Leukocyte (HLA) Class II Molecules. Vaccines (Basel) 2025; 13:88. [PMID: 39852867 PMCID: PMC11768865 DOI: 10.3390/vaccines13010088] [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: 12/16/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Anthrax is a serious disease caused by Bacillus anthracis (B. anthracis) with a very high mortality when the spores of B. anthracis are inhaled (inhalational anthrax). Aerosolized B. anthracis spores can be used as a deadly bioweapon. Vaccination against anthrax is the only effective preventive measure and, hence, the anthrax vaccine was administered to United States (and other) troops during the 1990-91 Gulf War. However, the anthrax vaccine is not harmless, and the anthrax vaccination has been linked to the occurrence and severity of Gulf War Illness (GWI), a debilitating Chronic Multisymptom Illness (CMI). We hypothesized that this is partly due to the combination of two factors, namely (a) the cytotoxicity of the antigen (anthrax Protective Antigen, PA) contained in the vaccine, and (b) the Human Leukocyte Antigen (HLA) genotype of susceptible vaccinees, reducing their ability to make antibodies against the cytotoxic PA. METHOD Here, we tested this hypothesis by determining the association between severity of GWI symptoms in 458 GW veterans and the overall strength of the binding affinity of the PA epitopes to the specific six Human Leukocyte Antigen (HLA) Class II alleles carried by each individual (two of each of the HLA-II genes: DPB1, DQB1, DRB1), responsible for initiating the process of antibody production in otherwise immunocompetent individuals, estimated in silico. RESULTS We found that the severity of GWI symptomatology was negatively and significantly correlated with the strength of the predicted binding affinity of PA peptides to HLA-II molecules (r=-0.356, p<0.001); the stronger the overall binding affinity, the weaker the symptoms. Since the binding of a peptide to an HLA-II molecule is the first and necessary step in initiating the production of antibodies, the findings above support our hypothesis that the severity of GWI symptomatology is partly due to a lack of HLA-II protection. CONCLUSIONS Reduced HLA protection against the toxic anthrax vaccine may underlie GWI.
Collapse
Affiliation(s)
- Lisa M. James
- The GWI and HLA Research Groups, Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA;
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Apostolos P. Georgopoulos
- The GWI and HLA Research Groups, Brain Sciences Center, Department of Veterans Affairs Health Care System, Minneapolis, MN 55417, USA;
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
12
|
Wang M, Yu F, Zhang Y. Present and future of cancer nano-immunotherapy: opportunities, obstacles and challenges. Mol Cancer 2025; 24:26. [PMID: 39827147 PMCID: PMC11748575 DOI: 10.1186/s12943-024-02214-5] [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: 09/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
Clinically, multimodal therapies are adopted worldwide for the management of cancer, which continues to be a leading cause of death. In recent years, immunotherapy has firmly established itself as a new paradigm in cancer care that activates the body's immune defense to cope with cancer. Immunotherapy has resulted in significant breakthroughs in the treatment of stubborn tumors, dramatically improving the clinical outcome of cancer patients. Multiple forms of cancer immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapy and cancer vaccines, have become widely available. However, the effectiveness of these immunotherapies is not much satisfying. Many cancer patients do not respond to immunotherapy, and disease recurrence appears to be unavoidable because of the rapidly evolving resistance. Moreover, immunotherapies can give rise to severe off-target immune-related adverse events. Strategies to remove these hindrances mainly focus on the development of combinatorial therapies or the exploitation of novel immunotherapeutic mediations. Nanomaterials carrying anticancer agents to the target site are considered as practical approaches for cancer treatment. Nanomedicine combined with immunotherapies offers the possibility to potentiate systemic antitumor immunity and to facilitate selective cytotoxicity against cancer cells in an effective and safe manner. A myriad of nano-enabled cancer immunotherapies are currently under clinical investigation. Owing to gaps between preclinical and clinical studies, nano-immunotherapy faces multiple challenges, including the biosafety of nanomaterials and clinical trial design. In this review, we provide an overview of cancer immunotherapy and summarize the evidence indicating how nanomedicine-based approaches increase the efficacy of immunotherapies. We also discuss the key challenges that have emerged in the era of nanotechnology-based cancer immunotherapy. Taken together, combination nano-immunotherapy is drawing increasing attention, and it is anticipated that the combined treatment will achieve the desired success in clinical cancer therapy.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China.
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| |
Collapse
|
13
|
Dodd GK, Keşmir C. In silico functional analysis of the human, chimpanzee, and gorilla MHC-A repertoires. Immunogenetics 2025; 77:12. [PMID: 39821345 PMCID: PMC11741996 DOI: 10.1007/s00251-024-01369-1] [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: 09/17/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
T cells recognize peptides displayed on the surface of cells on MHC molecules. Genetic variation in MHC genes alters their peptide-binding repertoire and thus influences the potential immune response generated against pathogens. Both gorillas and chimpanzees show reduced diversity at their MHC class I A (MHC-A) locus compared to humans, which has been suggested to be the result of a pathogen-mediated selective sweep. More specifically, gorillas lack A3 lineage alleles while chimpanzees seem to have lost the A2 lineage. While previous studies showed this using phylogenetic analysis, here, we take an in silico functional approach and use the peptide-MHC binding prediction software NetMHCpan to examine the peptide-binding repertoires of common human, chimpanzee, and gorilla MHC-A molecules. We find that both gorillas and chimpanzees lack the A02 peptide binding specificity (supertype) despite gorillas being expected to have this specificity since they kept the A2 lineage. Additionally, we show that human MHC molecules with the A02 specificity bind fewer virus-derived peptides than other MHC molecules. We also do not find differential presentation of self-peptides by the A02 supertype, making the purpose of maintaining this specificity in high frequencies in the human population unclear. Taken together, we hypothesize that poor presentation of viral peptides by A02 supertype MHC molecules could have resulted in a selective sweep in chimpanzees and/or gorillas, though we could not identify a specific virus that may have caused this sweep.
Collapse
Affiliation(s)
- Griffin Kutler Dodd
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Can Keşmir
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Arshad S, Cameron B, Joglekar AV. Immunopeptidomics for autoimmunity: unlocking the chamber of immune secrets. NPJ Syst Biol Appl 2025; 11:10. [PMID: 39833247 PMCID: PMC11747513 DOI: 10.1038/s41540-024-00482-x] [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: 07/14/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
T cells mediate pathogenesis of several autoimmune disorders by recognizing self-epitopes presented on Major Histocompatibility Complex (MHC) or Human Leukocyte Antigen (HLA) complex. The majority of autoantigens presented to T cells in various autoimmune disorders are not known, which has impeded autoantigen identification. Recent advances in immunopeptidomics have started to unravel the repertoire of antigenic epitopes presented on MHC. In several autoimmune diseases, immunopeptidomics has led to the identification of novel autoantigens and has enhanced our understanding of the mechanisms behind autoimmunity. Especially, immunopeptidomics has provided key evidence to explain the genetic risk posed by HLA alleles. In this review, we shed light on how immunopeptidomics can be leveraged to discover potential autoantigens. We highlight the application of immunopeptidomics in Type 1 Diabetes (T1D), Systemic Lupus Erythematosus (SLE), and Rheumatoid Arthritis (RA). Finally, we highlight the practical considerations of implementing immunopeptidomics successfully and the technical challenges that need to be addressed. Overall, this review will provide an important context for using immunopeptidomics for understanding autoimmunity.
Collapse
Affiliation(s)
- Sanya Arshad
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin Cameron
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alok V Joglekar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Zhang C, Wang J, Dang P, Wei Y, Wang X, Brothwell J, Sun Y, Zhu H, So K, Liu J, Wang Y, Lu X, Spinola S, Zhang X, Cao S. A physics informed neural network approach to quantify antigen presentation activities at single cell level using omics data. RESEARCH SQUARE 2025:rs.3.rs-5629379. [PMID: 39877095 PMCID: PMC11774464 DOI: 10.21203/rs.3.rs-5629379/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Antigen processing and presentation via major histocompatibility complex (MHC) molecules are central to immune surveillance. Yet, quantifying the dynamic activity of MHC class I and II antigen presentation remains a critical challenge, particularly in diseases like cancer, infection and autoimmunity where these pathways are often disrupted. Current methods fall short in providing precise, sample-specific insights into antigen presentation, limiting our understanding of immune evasion and therapeutic responses. Here, we present PSAA (PINN-empowered Systems Biology Analysis of Antigen Presentation Activity), which is designed to estimate sample-wise MHC class I and class II antigen presentation activity using bulk, single-cell, and spatially resolved transcriptomics or proteomics data. By reconstructing MHC pathways and employing pathway flux estimation, PSAA offers a detailed, stepwise quantification of MHC pathway activity, enabling predictions of gene-specific impacts and their downstream effects on immune interactions. Benchmarked across diverse omics datasets and experimental validations, PSAA demonstrates a robust prediction accuracy and utility across various disease contexts. In conclusion, PSAA and its downstream functions provide a comprehensive framework for analyzing the dynamics of MHC antigen presentation using omics data. By linking antigen presentation to immune cell activity and clinical outcomes, PSAA both elucidates key mechanisms driving disease progression and uncovers potential therapeutic targets.
Collapse
Affiliation(s)
- Chi Zhang
- Indiana University School of Medicine
| | | | | | | | | | | | - Yifan Sun
- Indiana University School of Medicine
| | | | - Kaman So
- Indiana University School of Medicine
| | | | - Yijie Wang
- Computer Science Department, Indiana University
| | | | | | | | - Sha Cao
- Oregon Health & Science University
| |
Collapse
|
16
|
Sun Y, Pumroy RA, Mallik L, Chaudhuri A, Wang C, Hwang D, Danon JN, Dasteh Goli K, Moiseenkova-Bell VY, Sgourakis NG. CryoEM structure of an MHC-I/TAPBPR peptide-bound intermediate reveals the mechanism of antigen proofreading. Proc Natl Acad Sci U S A 2025; 122:e2416992122. [PMID: 39786927 PMCID: PMC11745410 DOI: 10.1073/pnas.2416992122] [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: 08/21/2024] [Accepted: 12/05/2024] [Indexed: 01/30/2025] Open
Abstract
Class I major histocompatibility complex (MHC-I) proteins play a pivotal role in adaptive immunity by displaying epitopic peptides to CD8+ T cells. The chaperones tapasin and TAPBPR promote the selection of immunogenic antigens from a large pool of intracellular peptides. Interactions of chaperoned MHC-I molecules with incoming peptides are transient in nature, and as a result, the precise antigen proofreading mechanism remains elusive. Here, we leverage a high-fidelity TAPBPR variant and conformationally stabilized MHC-I, to determine the solution structure of the human antigen editing complex bound to a peptide decoy by cryogenic electron microscopy (cryo-EM) at an average resolution of 3.0 Å. Antigen proofreading is mediated by transient interactions formed between the nascent peptide binding groove with the P2/P3 peptide anchors, where conserved MHC-I residues stabilize incoming peptides through backbone-focused contacts. Finally, using our high-fidelity chaperone, we demonstrate robust peptide exchange on the cell surface across multiple clinically relevant human MHC-I allomorphs. Our work has important ramifications for understanding the selection of immunogenic epitopes for T cell screening and vaccine design applications.
Collapse
Affiliation(s)
- Yi Sun
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Ruth A. Pumroy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Leena Mallik
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Apala Chaudhuri
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Chloe Wang
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Immunology Graduate Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
| | - Daniel Hwang
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Julia N. Danon
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Kimia Dasteh Goli
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Vera Y. Moiseenkova-Bell
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Nikolaos G. Sgourakis
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Center for Computational and Genomic Medicine and Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| |
Collapse
|
17
|
Georgopoulos AP, James LM, Peterson PK. Human Leukocyte Antigen (HLA) at the Root of Persistent Antigens and Long COVID. JOURNAL OF IMMUNOLOGICAL SCIENCES 2025; 9:1-3. [PMID: 39949480 PMCID: PMC11823424 DOI: 10.29245/2578-3009/2025/1.1257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/16/2025]
Abstract
Here we offer a perspective on recent findings of persistent SARS-CoV-2 antigens in Long COVID1 through the lens of immunogenetic risk and protection, namely in the context of the fundamental role of Human Leukocyte Antigen (HLA) in eliminating viral infections. Specifically, we attribute the persistence of viral antigens to the lack or weak protection conferred by HLA against SARS-CoV-2 in individuals carrying HLA alleles with low binding affinities to the virus. We suggest that determining the HLA Class I and II makeup of Long COVID patients will provide valuable new information in elucidating the cause for antigen persistence underlying the development of Long COVID and pave the way for successful interventions.
Collapse
Affiliation(s)
- Apostolos P. Georgopoulos
- Brain Sciences Center, Minneapolis Veterans Affairs Health
Care System, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical
School, Minneapolis, MN, USA
| | - Lisa M. James
- Brain Sciences Center, Minneapolis Veterans Affairs Health
Care System, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota Medical
School, Minneapolis, MN, USA
- Department of Psychiatry, University of Minnesota Medical
School, Minneapolis, MN, USA
| | - Phillip K. Peterson
- Department of Medicine, University of Minnesota Medical
School, Minneapolis, MN, USA
| |
Collapse
|
18
|
Maccari G, Robinson J, Barker D, Yates A, Hammond J, Marsh SE. The 2024 IPD-MHC database update: a comprehensive resource for major histocompatibility complex studies. Nucleic Acids Res 2025; 53:D457-D461. [PMID: 39436012 PMCID: PMC11701557 DOI: 10.1093/nar/gkae932] [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: 09/16/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
The IPD-MHC Database project (http://www.ebi.ac.uk/ipd/mhc/) serves as a comprehensive and expertly curated repository for major histocompatibility complex (MHC) sequences from non-human species, providing the necessary infrastructure and tools to study the function and evolution of this highly polymorphic genomic region. In its latest version, the IPD-MHC database has expanded both in content and in the tools for data visualization and comparison. The database now hosts over 18 000 MHC alleles from 125 species, organized into eleven taxonomic groups, all manually curated and named by the Comparative MHC Nomenclature Committee. A cetacean section has recently been included, offering researchers valuable data to study the immune system of whales, dolphins, and porpoises, as well establishing the official nomenclature platform for the Cetacea Leukocyte Antigens (CeLA). In response to user demand and reflecting broader trends in bioinformatics and immunogenetics, IPD-MHC now includes the predicted tertiary structure of over 8000 alleles and allows comparison and visualisation of allele variation within and between species at single residue resolution. These latest developments maintain the critically important link between official nomenclature of curated alleles and the ability to analyse this complex polymorphism using the most up to date methods within a single repository.
Collapse
Affiliation(s)
- Giuseppe Maccari
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- The Pirbright Institute, Pirbright, Woking, Surrey GU24 0NF, UK
| | - James Robinson
- Anthony Nolan Research Institute, Royal Free Hospital, Pond Street, London NW3 2QG, UK
- UCL Cancer Institute, University College London (UCL), Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Dominic J Barker
- Anthony Nolan Research Institute, Royal Free Hospital, Pond Street, London NW3 2QG, UK
- UCL Cancer Institute, University College London (UCL), Royal Free Campus, Pond Street, London NW3 2QG, UK
| | - Andrew D Yates
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, Cambridgeshire CB10 1SD, UK
| | - John A Hammond
- The Pirbright Institute, Pirbright, Woking, Surrey GU24 0NF, UK
| | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, Pond Street, London NW3 2QG, UK
- UCL Cancer Institute, University College London (UCL), Royal Free Campus, Pond Street, London NW3 2QG, UK
| |
Collapse
|
19
|
Mallis RJ, Brazin KN, Duke‐Cohan JS, Akitsu A, Stephens HM, Chang‐Gonzalez AC, Masi DJ, Kirkpatrick EH, Holliday EL, Feng Y, Zienkiewicz KJ, Lee JJ, Cinella V, Uberoy KI, Tan K, Wagner G, Arthanari H, Hwang W, Lang MJ, Reinherz EL. Biophysical and Structural Features of αβT-Cell Receptor Mechanosensing: A Paradigmatic Shift in Understanding T-Cell Activation. Immunol Rev 2025; 329:e13432. [PMID: 39745432 PMCID: PMC11744257 DOI: 10.1111/imr.13432] [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: 11/08/2024] [Accepted: 12/10/2024] [Indexed: 01/21/2025]
Abstract
αβT cells protect vertebrates against many diseases, optimizing surveillance using mechanical force to distinguish between pathophysiologic cellular alterations and normal self-constituents. The multi-subunit αβT-cell receptor (TCR) operates outside of thermal equilibrium, harvesting energy via physical forces generated by T-cell motility and actin-myosin machinery. When a peptide-bound major histocompatibility complex molecule (pMHC) on an antigen presenting cell is ligated, the αβTCR on the T cell leverages force to form a catch bond, prolonging bond lifetime, and enhancing antigen discrimination. Under load, the αβTCR undergoes reversible structural transitions involving partial unfolding of its clonotypic immunoglobulin-like (Ig) domains and coupled rearrangements of associated CD3 subunits and structural elements. We postulate that transitions provide critical energy to initiate the signaling cascade via induction of αβTCR quaternary structural rearrangements, associated membrane perturbations, exposure of CD3 ITAMs to phosphorylation by non-receptor tyrosine kinases, and phase separation of signaling molecules. Understanding force-mediated signaling by the αβTCR clarifies long-standing questions regarding αβTCR antigen recognition, specificity and affinity, providing a basis for continued investigation. Future directions include examining atomistic mechanisms of αβTCR signal initiation, performance quality, tissue compliance adaptability, and T-cell memory fate. The mechanotransduction paradigm will foster improved rational design of T-cell based vaccines, CAR-Ts, and adoptive therapies.
Collapse
MESH Headings
- Humans
- Animals
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Lymphocyte Activation/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Mechanotransduction, Cellular
- Signal Transduction
Collapse
Affiliation(s)
- Robert J. Mallis
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
- Department of DermatologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Kristine N. Brazin
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Jonathan S. Duke‐Cohan
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Aoi Akitsu
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Hanna M. Stephens
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | | | - Daniel J. Masi
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Evan H. Kirkpatrick
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Elizabeth L. Holliday
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Yinnian Feng
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | | | - Jonathan J. Lee
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Vincenzo Cinella
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Kaveri I. Uberoy
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Kemin Tan
- Structural Biology Center, X‐Ray Science Division, Advanced Photon SourceArgonne National LaboratoryLemontIllinoisUSA
| | - Gerhard Wagner
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Haribabu Arthanari
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Wonmuk Hwang
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTexasUSA
- Department of Materials Science and EngineeringTexas A&M UniversityCollege StationTexasUSA
- Department of Physics and AstronomyTexas A&M UniversityCollege StationTexasUSA
- Center for AI and Natural SciencesKorea Institute for Advanced StudySeoulRepublic of Korea
| | - Matthew J. Lang
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTennesseeUSA
- Department of Molecular Physiology and BiophysicsVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Ellis L. Reinherz
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
20
|
Gillespie GM, Quastel MN, McMichael AJ. HLA-E: Immune Receptor Functional Mechanisms Revealed by Structural Studies. Immunol Rev 2025; 329:e13434. [PMID: 39753525 PMCID: PMC11698700 DOI: 10.1111/imr.13434] [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: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025]
Abstract
HLA-E is a nonclassical, nonpolymorphic, class Ib HLA molecule. Its primary function is to present a conserved nonamer peptide, termed VL9, derived from the signal sequence of classical MHC molecules to the NKG2x-CD94 receptors on NK cells and a subset of T lymphocytes. These receptors regulate the function of NK cells, and the importance of this role, which is conserved across mammalian species, probably accounts for the lack of genetic polymorphism. A second minor function is to present other, weaker binding, pathogen-derived peptides to T lymphocytes. Most of these peptides bind suboptimally to HLA-E, but this binding appears to be enabled by the relative stability of peptide-free, but receptive, HLA-E-β2m complexes. This, in turn, may favor nonclassical antigen processing that may be associated with bacteria infected cells. This review explores how the structure of HLA-E, bound to different peptides and then to NKG2-CD94 or T-cell receptors, relates to HLA-E cell biology and immunology. A detailed understanding of this molecule could open up opportunities for development of universal T-cell and NK-cell-based immunotherapies.
Collapse
MESH Headings
- Humans
- Histocompatibility Antigens Class I/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/chemistry
- Animals
- HLA-E Antigens
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Protein Binding
- Antigen Presentation
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/chemistry
- NK Cell Lectin-Like Receptor Subfamily C/metabolism
- Structure-Activity Relationship
- Peptides/chemistry
- Peptides/immunology
- Peptides/metabolism
- NK Cell Lectin-Like Receptor Subfamily D/metabolism
- NK Cell Lectin-Like Receptor Subfamily D/chemistry
- NK Cell Lectin-Like Receptor Subfamily D/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/chemistry
- Protein Conformation
Collapse
Affiliation(s)
| | - Max N. Quastel
- Nuffield Department of Medicine, Center for Immuno‐OncologyUniversity of OxfordOxfordUK
| | - Andrew J. McMichael
- Nuffield Department of Medicine, Center for Immuno‐OncologyUniversity of OxfordOxfordUK
| |
Collapse
|
21
|
Wang H, Li W, Ma Z, Zhang N, Wang Y, Gao L, Li X, Cao H, Zheng SJ. Identification of novel T-cell epitopes on viral protein VP4 of Infectious Bursal Disease Virus (IBDV) that play critical roles in eliciting cellular immune response. Int J Biol Macromol 2025; 284:137942. [PMID: 39579808 DOI: 10.1016/j.ijbiomac.2024.137942] [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: 10/21/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Infectious bursal disease virus (IBDV) is a highly infectious RNA virus that causes severe damage to the bursa of Fabricius (BF), resulting in immunosuppression. Currently, IBDV vaccines mainly rely on the induction of neutralizing antibodies against VP2 for protection, and the role of cellular immunity against IBDV infection is unclear. Here, we show that IBDV VP4, a serine protease of the virion, is responsible for inducing specific T cell immunity against IBDV infection. Furthermore, we identified three specific T cell epitope peptides on VP4, among which, two epitopes (4/10 and 4/12) could be recognized by CD8+T cells, and the other one (4/27) by both CD4+T cells and CD8+T cells. Importantly, infection of SPF chickens with rFAdV-4-ON1-VP4, which is generated with the backbone of an avirulent fowl adenovirus strain (FAdV-4-ON1) without causing any clinical symptoms in chickens, induced IBDV-specific cellular immunity, providing effective protection for chickens against IBDV infection. This study demonstrates for the first time that the specific cellular immunity induced by IBDV VP4 specific-T cell epitopes plays a protective role in host response against IBDV infection, providing a new insight into the development of novel vaccines for the control of viral diseases.
Collapse
Affiliation(s)
- Hongnuan Wang
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Wei Li
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Ziyue Ma
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Nianzhi Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yongqiang Wang
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Li Gao
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaoqi Li
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hong Cao
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shijun J Zheng
- National Key Laboratory of Veterinary Public Health Security, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
22
|
Wekerle H, Kawakami N. Fragments of the brain's myelin proteins train T cells to ward off autoimmune attacks. Nature 2025; 637:41-42. [PMID: 39633120 DOI: 10.1038/d41586-024-03846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
|
23
|
Matsumoto S, Tsujikawa T, Tokita S, Mohamed Bedeir M, Matsuo K, Hata F, Hirohashi Y, Kanaseki T, Torigoe T. HLA class II neoantigen presentation for CD4 + T cell surveillance in HLA class II-negative colorectal cancer. Oncoimmunology 2024; 13:2404665. [PMID: 39508845 PMCID: PMC11542397 DOI: 10.1080/2162402x.2024.2404665] [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: 08/09/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 11/15/2024] Open
Abstract
Neoantigen-reactive CD4+ T cells play a key role in the anti-tumor immune response. However, the majority of epithelial tumors are negative for HLA class II (HLA-II) surface expression, and less is known about the processing of HLA-II antigens. Here, we directly identified naturally presented HLA-II neoantigens in HLA-II negative colorectal cancer (CRC) tissue using a proteogenomic approach. The neoantigens were immunogenic and induced patient CD4+ T cells with a Th1-like memory phenotype that produced IFN-γ, IL2 and TNF-α. Multiplex immunohistochemistry (IHC) demonstrated an interaction between Th cells and HLA-II-positive antigen-presenting cells (APCs) at the invasive margin and within the tertiary lymphoid structures (TLS). In our CRC cohort, the density of stromal APCs was associated with HLA-II antigen presentation in the tumor microenvironment (TME), and the number of TLS was positively correlated with the number of somatic mutations in the tumors. These results demonstrate the presence of neoantigen-specific CD4+ surveillance in HLA-II-negative CRC and suggest a potential role for macrophages and dendritic cells (DCs) at the invasive margin and in TLS for antigen presentation. Stromal APCs in the TME can potentially be used as a source for HLA-II neoantigen identification.
Collapse
Affiliation(s)
- Satoru Matsumoto
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Department of Surgery, IMS Sapporo Digestive Disease Center General Hospital, Sapporo, Japan
| | - Takahiro Tsujikawa
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Serina Tokita
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | - Mai Mohamed Bedeir
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Fumitake Hata
- Department of Surgery, Sapporo Dohto Hospital, Sapporo, Japan
| | | | | | | |
Collapse
|
24
|
Santiago-Sánchez GS, Fabian KP, Hodge JW. A landscape of checkpoint blockade resistance in cancer: underlying mechanisms and current strategies to overcome resistance. Cancer Biol Ther 2024; 25:2308097. [PMID: 38306161 PMCID: PMC10841019 DOI: 10.1080/15384047.2024.2308097] [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: 08/31/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
The discovery of immune checkpoints and the development of immune checkpoint inhibitors (ICI) have achieved a durable response in advanced-stage cancer patients. However, there is still a high proportion of patients who do not benefit from ICI therapy due to a lack of response when first treated (primary resistance) or detection of disease progression months after objective response is observed (acquired resistance). Here, we review the current FDA-approved ICI for the treatment of certain solid malignancies, evaluate the contrasting responses to checkpoint blockade in different cancer types, explore the known mechanisms associated with checkpoint blockade resistance (CBR), and assess current strategies in the field that seek to overcome these mechanisms. In order to improve current therapies and develop new ones, the immunotherapy field still has an unmet need in identifying other molecules that act as immune checkpoints, and uncovering other mechanisms that promote CBR.
Collapse
Affiliation(s)
- Ginette S. Santiago-Sánchez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kellsye P. Fabian
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Hodge
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Marzella DF, Crocioni G, Radusinović T, Lepikhov D, Severin H, Bodor DL, Rademaker DT, Lin C, Georgievska S, Renaud N, Kessler AL, Lopez-Tarifa P, Buschow SI, Bekkers E, Xue LC. Geometric deep learning improves generalizability of MHC-bound peptide predictions. Commun Biol 2024; 7:1661. [PMID: 39702482 DOI: 10.1038/s42003-024-07292-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
The interaction between peptides and major histocompatibility complex (MHC) molecules is pivotal in autoimmunity, pathogen recognition and tumor immunity. Recent advances in cancer immunotherapies demand for more accurate computational prediction of MHC-bound peptides. We address the generalizability challenge of MHC-bound peptide predictions, revealing limitations in current sequence-based approaches. Our structure-based methods leveraging geometric deep learning (GDL) demonstrate promising improvement in generalizability across unseen MHC alleles. Further, we tackle data efficiency by introducing a self-supervised learning approach on structures (3D-SSL). Without being exposed to any binding affinity data, our 3D-SSL outperforms sequence-based methods trained on ~90 times more data points. Finally, we demonstrate the resilience of structure-based GDL methods to biases in binding data on an Hepatitis B virus vaccine immunopeptidomics case study. This proof-of-concept study highlights structure-based methods' potential to enhance generalizability and data efficiency, with possible implications for data-intensive fields like T-cell receptor specificity predictions.
Collapse
Affiliation(s)
- Dario F Marzella
- Medical BioSciences department, Radboudumc, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | | | | | - Daniil Lepikhov
- Medical BioSciences department, Radboudumc, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Heleen Severin
- Medical BioSciences department, Radboudumc, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Dani L Bodor
- Netherlands eScience Center, Amsterdam, The Netherlands
| | - Daniel T Rademaker
- Medical BioSciences department, Radboudumc, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - ChiaYu Lin
- Netherlands eScience Center, Amsterdam, The Netherlands
| | | | | | - Amy L Kessler
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | | | - Sonja I Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | - Erik Bekkers
- University of Amsterdam, Amsterdam, The Netherlands
| | - Li C Xue
- Medical BioSciences department, Radboudumc, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Banerjee A, Dass D, Mukherjee S, Kaul M, Harshithkumar R, Bagchi P, Mukherjee A. The 'Oma's of the Gammas-Cancerogenesis by γ-Herpesviruses. Viruses 2024; 16:1928. [PMID: 39772235 PMCID: PMC11680331 DOI: 10.3390/v16121928] [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: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/03/2025] Open
Abstract
Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV), which are the only members of the gamma(γ) herpesviruses, are oncogenic viruses that significantly contribute to the development of various human cancers, such as Burkitt's lymphoma, nasopharyngeal carcinoma, Hodgkin's lymphoma, Kaposi's sarcoma, and primary effusion lymphoma. Oncogenesis triggered by γ-herpesviruses involves complex interactions between viral genetics, host cellular mechanisms, and immune evasion strategies. At the genetic level, crucial viral oncogenes participate in the disruption of cell signaling, leading to uncontrolled proliferation and inhibition of apoptosis. These viral proteins can modulate several cellular pathways, including the NF-κB and JAK/STAT pathways, which play essential roles in cell survival and inflammation. Epigenetic modifications further contribute to EBV- and KSHV-mediated cancerogenesis. Both EBV and KSHV manipulate host cell DNA methylation, histone modification, and chromatin remodeling, the interplay of which contribute to the elevation of oncogene expression and the silencing of the tumor suppressor genes. Immune factors also play a pivotal role in the development of cancer. The γ-herpesviruses have evolved intricate immune evasion strategies, including the manipulation of the major histocompatibility complex (MHC) and the release of cytokines, allowing infected cells to evade immune detection and destruction. In addition, a compromised immune system, such as in HIV/AIDS patients, significantly increases the risk of cancers associated with EBV and KSHV. This review aims to provide a comprehensive overview of the genetic, epigenetic, and immune mechanisms by which γ-herpesviruses drive cancerogenesis, highlighting key molecular pathways and potential therapeutic targets.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Debashree Dass
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Soumik Mukherjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Mollina Kaul
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - R. Harshithkumar
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
| | - Parikshit Bagchi
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anupam Mukherjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, MH, India; (A.B.); (D.D.); (S.M.); (M.K.); (R.H.)
- AcSIR—Academy of Scientific & Innovative Research, Ghaziabad 201002, UP, India
| |
Collapse
|
27
|
López D, Zumárraga J. Bioinformatic Tools for Studying the Cellular Immune Response to SARS-CoV-2, Vaccine Efficacy, and Future Pandemics at the Global Population Level. Int J Mol Sci 2024; 25:13477. [PMID: 39769240 PMCID: PMC11678114 DOI: 10.3390/ijms252413477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Antigen recognition by human leukocyte antigen (HLA) restriction is critical for an adequate antiviral response in both natural infection and vaccination. However, the overwhelming polymorphism of HLA, with nearly 40,000 alleles identified, is an important limitation for the global analysis of cellular immune responses and vaccine efficacy. In this narrative review, we included several immunoinformatics studies performed in our laboratory to circumvent this limitation. These analyses focused on studying the cellular immune responses restricted by the most common HLA alleles, and their role in vaccine efficacy. Computational studies validated experimentally, such as our laboratory has carried out, represent a useful, rapid, and cost-effective strategy to combat future pandemics.
Collapse
Affiliation(s)
- Daniel López
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain;
| | | |
Collapse
|
28
|
Flatby HM, Ravi A, Liyanarachi KV, Afset JE, Rasheed H, Brumpton BM, Hveem K, Åsvold BO, DeWan AT, Solligård E, Damås JK, Rogne T. A Genome-wide Association Study of Susceptibility to Upper Urinary Tract Infections. J Infect Dis 2024; 230:e1334-e1343. [PMID: 38713594 PMCID: PMC11646603 DOI: 10.1093/infdis/jiae231] [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: 09/18/2023] [Revised: 03/21/2024] [Accepted: 05/06/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Our goal was to identify genetic and modifiable risk factors for upper urinary tract infections (UTIs). METHODS We used data from UK Biobank, the Trøndelag Health Study, and the Michigan Genomics Initiative to conduct genome-wide association studies and sex-stratified analyses on upper UTI. Mendelian randomization (MR) analyses were conducted to examine potential causal relationships between cardiometabolic risk factors and upper UTIs. RESULTS One genome-wide significant (P ≤ 5E-08) locus was associated with the susceptibility to upper UTI, located near TSN in the female-only analysis. Additionally, we identified suggestive (P ≤ 5E-06) loci near DNAI3 for females, SCAMP1-AS1 for males, and near TSN, LINC00603, and HLA-DQA2 for both sexes. In MR analyses, higher genetically predicted lifetime smoking scores were associated with an increased risk of developing upper UTI for females and both sexes (odds ratio [OR], 4.84, P = 4.50E-06 and OR, 2.79, P = 3.02E-05, respectively). CONCLUSIONS We found that genetic variants near TSN was associated with the risk of upper UTIs among females. In addition, we found several genetic loci with suggestive associations with the risk of upper UTIs. Finally, MR analyses found smoking to be a potential causal risk factor for upper UTIs.
Collapse
Affiliation(s)
- Helene M Flatby
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Clinic of Anaesthesia and Intensive Care
| | - Anuradha Ravi
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Medical Genetics
| | - Kristin V Liyanarachi
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Infectious Diseases, St Olavs Hospital, Trondheim University Hospital
| | - Jan E Afset
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology
- Department of Medical Microbiology, St Olavs Hospital, Trondheim University Hospital
| | - Humaira Rasheed
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology
- Clinic of Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim
| | - Ben M Brumpton
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology
- Clinic of Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim
- The Trøndelag Health Study Research Center, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger
| | - Kristian Hveem
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology
- Department of Research, Innovation, and Education
| | - Bjørn O Åsvold
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology
- The Trøndelag Health Study Research Center, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger
- Department of Endocrinology, Clinic of Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Andrew T DeWan
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Chronic Disease Epidemiology and Center for Perinatal, Pediatric and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Erik Solligård
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Innovation, Education and Health Sciences, Helse Møre og Romsdal Hospital Trust, Ålesund, Norway
| | - Jan K Damås
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Infectious Diseases, St Olavs Hospital, Trondheim University Hospital
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tormod Rogne
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology
- Department of Chronic Disease Epidemiology and Center for Perinatal, Pediatric and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut
| |
Collapse
|
29
|
Ma X, Li J, Liu N, Banerjee S, Hu X, Wang X, Dong J, Liu K, Yang C, Dong Z. Insights into the distinct membrane targeting mechanisms of WDR91 family proteins. Structure 2024; 32:2287-2300.e4. [PMID: 39426373 DOI: 10.1016/j.str.2024.09.023] [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: 11/27/2023] [Revised: 07/15/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
WDR91 and SORF1, members of the WD repeat-containing protein 91 family, control phosphoinositide conversion by inhibiting phosphatidylinositol 3-kinase activity on endosomes, which promotes endosome maturation. Here, we report the crystal structure of the human WDR91 WD40 domain complexed with Rab7 that has an unusual interface at the C-terminus of the Rab7 switch II region. WDR91 is highly selective for Rab7 among the tested GTPases. A LIS1 homology (LisH) motif within the WDR91 N-terminal domain (NTD) mediates self-association and may contribute partly to the augmented interaction between full-length WDR91 and Rab7. Both the Rab7 binding site and the LisH motif are indispensable for WDR91 function in endocytic trafficking. For the WDR91 orthologue SORF1 lacking the C-terminal WD40 domain, a C-terminal amphipathic helix (AH) mediates strong interactions with liposomes containing acidic lipids. During evolution the human WDR91 ancestor gene might have acquired a WD40 domain to replace the AH for endosomal membrane targeting.
Collapse
Affiliation(s)
- Xinli Ma
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, China
| | - Jian Li
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, China
| | - Nan Liu
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Surajit Banerjee
- Northeastern Collaborative Access Team, Argonne National Laboratory, Argonne, Illinois 60439, USA
| | - Xiaotong Hu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, China
| | - Xiaoyu Wang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, China
| | - Jianshu Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, China; College of Medicine, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chonglin Yang
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450003, China; College of Medicine, Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
30
|
Shahsavari S, Rad MB, Hajiaghajani A, Rostami M, Hakimian F, Jafarzadeh S, Hasany M, Collingwood JF, Aliakbari F, Fouladiha H, Bardania H, Otzen DE, Morshedi D. Magnetoresponsive liposomes applications in nanomedicine: A comprehensive review. Biomed Pharmacother 2024; 181:117665. [PMID: 39541790 DOI: 10.1016/j.biopha.2024.117665] [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: 07/09/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Safe and effective cancer therapy requires a suitable nanocarrier that can target particular sites, such as cancer cells, in a selective manner. With the tremendous growth in nanotechnology, liposomes, among various competing nanocarriers, have shown promising advances in cancer therapy. Magnetic nanoparticles and metal ions are wide-reaching candidates for conferring magnetic properties and for incorporation into liposomes. Combining liposomes with magnetic structures enables construction of magnetoresponsive liposomes, allowing stimuli-responsiveness to an alternating magnetic field, magnetic targeting, and tracking by magnetic resonance imaging, which could all occur in parallel. This review presents a comprehensive analysis of the practical advances and novel aspects of design, synthesis and engineering magnetoresponsive liposomes, emphasizing their diverse properties for various applications. Our work explores the innovative uses of these structures, extending beyond drug delivery to include smart contrast agents, cell labeling, biosensing, separation, and filtering. By comparing new findings with earlier studies, we showcase significant improvements in efficiency and uncover new potentials, setting a new benchmark for future research in the field of magnetoresponsive liposomes.
Collapse
Affiliation(s)
- Shayan Shahsavari
- Iran Nanotechnology Innovation Council, Nanoclub Elites Association, Tehran, Iran
| | - Mohammad Behnam Rad
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, Tehran, Iran
| | - Amirhossein Hajiaghajani
- School of Electrical Engineering, Iran University of Science and Technology, Tehran 1684613114, Iran
| | | | - Fatemeh Hakimian
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384, Tehran, Iran
| | - Sina Jafarzadeh
- Department of Energy Conversion and Storage, Technical University of Denmark, Anker Engelunds Vej, Lyngby 2800 Kgs, Denmark
| | - Masoud Hasany
- Department of Civil and Mechanical Engineering, Technical University of Denmark, Lyngby 2800 Kgs, Denmark
| | | | - Farhang Aliakbari
- National Institute of Genetic Engineering and Biotechnology, Shahrak-e Pajoohesh, km 15 Tehran - Karaj Highway, P.O.Box:14965/161, Tehran, Iran; Molecular Medicine Research Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Hamideh Fouladiha
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Centre (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, Aarhus C DK-8000, Denmark.
| | - Dina Morshedi
- National Institute of Genetic Engineering and Biotechnology, Shahrak-e Pajoohesh, km 15 Tehran - Karaj Highway, P.O.Box:14965/161, Tehran, Iran.
| |
Collapse
|
31
|
Galdino Andrade TE, Scavassini Peña M, Fiorotti J, de Souza Bin R, Rodrigues Caetano A, Connelley T, Ferreira de Miranda Santos IK. Graduate Student Literature Review: The DRB3 gene of the bovine major histocompatibility complex-Discovery, diversity, and distribution of alleles in commercial breeds of cattle and applications for development of vaccines. J Dairy Sci 2024; 107:11324-11341. [PMID: 39004123 DOI: 10.3168/jds.2023-24628] [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: 12/28/2023] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
The bovine major histocompatibility complex (MHC), also known as the bovine leukocyte antigen (BoLA) complex, is the genomic region that encodes the most important molecules for antigen presentation to initiate immune responses. The first evidence of MHC in bovines pointed to a locus containing 2 antigens, one detected by cytotoxic antiserum (MHC class I) and another studied by mixed lymphocyte culture tests (MHC class II). The most studied gene in the BoLA region is the highly polymorphic BoLA-DRB3, which encodes a β chain with a peptide groove domain involved in antigen presentation for T cells that will develop and co-stimulate cellular and humoral effector responses. The BoLA-DRB3 alleles have been associated with outcomes in infectious diseases such as mastitis, trypanosomiasis, and tick loads, and with production traits. To catalog these alleles, 2 nomenclature methods were proposed, and the current use of both systems makes it difficult to list, comprehend and apply these data effectively. In this review we have organized the knowledge available in all of the reports on the frequencies of BoLA-DRB3 alleles. It covers information from studies made in at least 26 countries on more than 30 breeds; studies are lacking in countries that are important producers of cattle livestock. We highlight practical applications of BoLA studies for identification of markers associated with resistance to infectious and parasitic diseases, increased production traits and T cell epitope mapping, in addition to genetic diversity and conservation studies of commercial and Creole and locally adapted breeds. Finally, we provide support for the need of studies to discover new BoLA alleles and uncover unknown roles of this locus in production traits.
Collapse
Affiliation(s)
| | - Maurício Scavassini Peña
- Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil, 14049-900
| | - Jéssica Fiorotti
- Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil, 14049-900
| | - Renan de Souza Bin
- Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil, 14049-900
| | | | - Timothy Connelley
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom, EH25 9RG
| | | |
Collapse
|
32
|
Wang CY, Lin SC, Chang KJ, Cheong HP, Wu SR, Lee CH, Chuang MW, Chiou SH, Hsu CH, Ko PS. Immunoediting in acute myeloid leukemia: Reappraising T cell exhaustion and the aberrant antigen processing machinery in leukemogenesis. Heliyon 2024; 10:e39731. [PMID: 39568858 PMCID: PMC11577197 DOI: 10.1016/j.heliyon.2024.e39731] [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: 02/24/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Acute myeloid leukemia (AML) establishes an immunosuppressive microenvironment that favors leukemic proliferation. The immune-suppressive cytokines altered antigen processing, and presentation collectively assist AML cells in escaping cytotoxic T-cell surveillance. These CD8+ T cell dysfunction features are emerging therapeutic targets in relapsed/refractory AML patients. Besides, CD8+ T cell exhaustion is a hotspot in recent clinical oncology studies, but its pathophysiology has yet to be elucidated in AML. In this review, we summarize high-quality original studies encompassing the phenotypic and genomic characteristics of T cell exhaustion events in the leukemia progression, emphasize the surface immuno-peptidome that dynamically tunes the fate of T cells to function or dysfunction states, and revisit the biochemical and biophysical properties of type 1 MHC antigen processing mechanism (APM) that pivots in the phenomenon of leukemia antigen dampening.
Collapse
Affiliation(s)
- Ching-Yun Wang
- Department of Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shiuan-Chen Lin
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kao-Jung Chang
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Han-Ping Cheong
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Sin-Rong Wu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Hao Lee
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Wei Chuang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Chih-Hung Hsu
- Department of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Po-Shen Ko
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Division of Hematology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
33
|
Zhang X, Goedegebuure SP, Chen MY, Mishra R, Zhang F, Yu YY, Singhal K, Li L, Gao F, Myers NB, Vickery T, Hundal J, McLellan MD, Sturmoski MA, Kim SW, Chen I, Davidson JT, Sankpal NV, Myles S, Suresh R, Ma CX, Foluso A, Wang-Gillam A, Davies S, Hagemann IS, Mardis ER, Griffith O, Griffith M, Miller CA, Hansen TH, Fleming TP, Schreiber RD, Gillanders WE. Neoantigen DNA vaccines are safe, feasible, and induce neoantigen-specific immune responses in triple-negative breast cancer patients. Genome Med 2024; 16:131. [PMID: 39538331 PMCID: PMC11562513 DOI: 10.1186/s13073-024-01388-3] [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: 11/26/2023] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Neoantigen vaccines can induce or enhance highly specific antitumor immune responses with minimal risk of autoimmunity. We have developed a neoantigen DNA vaccine platform capable of efficiently presenting both HLA class I and II epitopes and performed a phase 1 clinical trial in triple-negative breast cancer patients with persistent disease on surgical pathology following neoadjuvant chemotherapy, a patient population at high risk of disease recurrence. METHODS Expressed somatic mutations were identified by tumor/normal exome sequencing and tumor RNA sequencing. The pVACtools software suite of neoantigen prediction algorithms was used to identify and prioritize cancer neoantigens and facilitate vaccine design for manufacture in an academic GMP facility. Neoantigen DNA vaccines were administered via electroporation in the adjuvant setting (i.e., following surgical removal of the primary tumor and completion of standard of care therapy). Vaccines were monitored for safety and immune responses via ELISpot, intracellular cytokine production via flow cytometry, and TCR sequencing. RESULTS Eighteen subjects received three doses of a neoantigen DNA vaccine encoding on average 11 neoantigens per patient (range 4-20). The vaccinations were well tolerated with relatively few adverse events. Neoantigen-specific T cell responses were induced in 14/18 patients as measured by ELISpot and flow cytometry. At a median follow-up of 36 months, recurrence-free survival was 87.5% (95% CI: 72.7-100%) in the cohort of vaccinated patients. CONCLUSION Our study demonstrates neoantigen DNA vaccines are safe, feasible, and capable of inducing neoantigen-specific immune responses. CLINICAL TRIAL REGISTRATION NUMBER NCT02348320.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael Y Chen
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rashmi Mishra
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Felicia Zhang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yik Yeung Yu
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kartik Singhal
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Feng Gao
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nancy B Myers
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammi Vickery
- Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jasreet Hundal
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael D McLellan
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mark A Sturmoski
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Samuel W Kim
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ina Chen
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse T Davidson
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Narendra V Sankpal
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie Myles
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Rama Suresh
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Cynthia X Ma
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Ademuyiwa Foluso
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Andrea Wang-Gillam
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Sherri Davies
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
- Current Affiliation: Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Obi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Malachi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Christopher A Miller
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ted H Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert D Schreiber
- Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA.
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
34
|
Magnusen AF, Pandey MK. Complement System and Adhesion Molecule Skirmishes in Fabry Disease: Insights into Pathogenesis and Disease Mechanisms. Int J Mol Sci 2024; 25:12252. [PMID: 39596318 PMCID: PMC11594573 DOI: 10.3390/ijms252212252] [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] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Fabry disease is a rare X-linked lysosomal storage disorder caused by mutations in the galactosidase alpha (GLA) gene, resulting in the accumulation of globotriaosylceramide (Gb3) and its deacetylated form, globotriaosylsphingosine (Lyso-Gb3) in various tissues and fluids throughout the body. This pathological accumulation triggers a cascade of processes involving immune dysregulation and complement system activation. Elevated levels of complement 3a (C3a), C5a, and their precursor C3 are observed in the plasma, serum, and tissues of patients with Fabry disease, correlating with significant endothelial cell abnormalities and vascular dysfunction. This review elucidates how the complement system, particularly through the activation of C3a and C5a, exacerbates disease pathology. The activation of these pathways leads to the upregulation of adhesion molecules, including vascular cell adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1), platelet and endothelial cell adhesion molecule 1 (PECAM1), and complement receptor 3 (CR3) on leukocytes and endothelial cells. This upregulation promotes the excessive recruitment of leukocytes, which in turn exacerbates disease pathology. Targeting complement components C3a, C5a, or their respective receptors, C3aR (C3a receptor) and C5aR1 (C5a receptor 1), could potentially reduce inflammation, mitigate tissue damage, and improve clinical outcomes for individuals with Fabry disease.
Collapse
Affiliation(s)
- Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
35
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Tokita S, Kanaseki T, Torigoe T. Neoantigen prioritization based on antigen processing and presentation. Front Immunol 2024; 15:1487378. [PMID: 39569190 PMCID: PMC11576432 DOI: 10.3389/fimmu.2024.1487378] [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: 08/28/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Somatic mutations in tumor cells give rise to mutant proteins, fragments of which are often presented by MHC and serve as neoantigens. Neoantigens are tumor-specific and not expressed in healthy tissues, making them attractive targets for T-cell-based cancer immunotherapy. On the other hand, since most somatic mutations differ from patient to patient, neoantigen-targeted immunotherapy is personalized medicine and requires their identification in each patient. Computational algorithms and machine learning methods have been developed to prioritize neoantigen candidates. In fact, since the number of clinically relevant neoantigens present in a patient is generally limited, this process is like finding a needle in a haystack. Nevertheless, MHC presentation of neoantigens is not random but follows certain rules, and the efficiency of neoantigen detection may be further improved with technological innovations. In this review, we discuss current approaches to the detection of clinically relevant neoantigens, with a focus on antigen processing and presentation.
Collapse
Affiliation(s)
- Serina Tokita
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University, Sapporo, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
37
|
Wang Q, Su T, Cheng F, Zhou S, Liu X, Wang M, Xu Y, Tang R, Liao S, Dailey J, Xiao G, Yang C, Wen H, Zheng W, Wen B, Tyc KM, Liu J, Sun D, Wang S, Zhu G. Proteolysis-targeting vaccines (PROTAVs) for robust combination immunotherapy of melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.01.616069. [PMID: 39574605 PMCID: PMC11580958 DOI: 10.1101/2024.10.01.616069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Protein/peptide subunit vaccines are promising to promote the tumor therapeutic efficacy of immune checkpoint blockade (ICB). However, current protein/peptide vaccines elicit limited antitumor T cell responses, leading to suboptimal therapeutic efficacy. Here, we present proteolysis-targeting vaccines (PROTAVs) that facilitate antigen proteolytic processing and cross-presentation to potentiate T cell responses for robust ICB combination immunotherapy of melanoma. PROTAVs are modular conjugates of protein/peptide antigens, E3 ligase-binding ligands, and linkers. In antigen-presenting cells (APCs), PROTAVs bind to E3 ligases to rapidly ubiquitinate PROTAV antigens, facilitating antigen proteolytic processing by proteasome, and thereby promoting antigen cross-presentation to T cells and potentiating CD8+ T cell responses. We developed a melanoma PROTAV using a tandem peptide of trivalent melanoma-associated antigens. Co-delivered by lipid nanoparticles (LNPs) with bivalent immunostimulant adjuvants, this PROTAV promotes the quantity and quality of melanoma-specific CD8+ T cells in mice. Further, combining PROTAV and ICB ameliorates the immunosuppressive melanoma microenvironment. As a result, PROTAV and ICB combination enhances melanoma complete regression rates and eradicated 100% large Braf V600E melanoma without recurrence in syngeneic mice. PROTAVs hold the potential for robust tumor combination immunotherapy.
Collapse
Affiliation(s)
- Qiyan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ting Su
- Department of Pharmaceutics and Center for Pharmaceutical Engineering, College of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Furong Cheng
- Department of Pharmaceutics and Center for Pharmaceutical Engineering, College of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shurong Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiang Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mi Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - You Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ri Tang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shimiao Liao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jordan Dailey
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guolan Xiao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chunpeng Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hanning Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weijia Zheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katarzyna M Tyc
- Department of Biostatistics, School of Public Health; Bioinformatics Shared Resource, Massey Comprehensive Cancer Center; Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jinze Liu
- Department of Biostatistics, School of Public Health; Bioinformatics Shared Resource, Massey Comprehensive Cancer Center; Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Bioinnovations in Brain Cancer, Biointerfaces Institute; Center for RNA Biomedicine. University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
38
|
Gonzales GA, Huang S, Wilkinson L, Nguyen JA, Sikdar S, Piot C, Naumenko V, Rajwani J, Wood CM, Dinh I, Moore M, Cedeño E, McKenna N, Polyak MJ, Amidian S, Ebacher V, Rosin NL, Carneiro MB, Surewaard B, Peters NC, Mody CH, Biernaskie J, Yates RM, Mahoney DJ, Canton J. The pore-forming apolipoprotein APOL7C drives phagosomal rupture and antigen cross-presentation by dendritic cells. Sci Immunol 2024; 9:eadn2168. [PMID: 39485861 DOI: 10.1126/sciimmunol.adn2168] [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: 11/28/2023] [Revised: 07/23/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Conventional dendritic cells (cDCs) generate protective cytotoxic T lymphocyte (CTL) responses against extracellular pathogens and tumors. This is achieved through a process known as cross-presentation (XP), and, despite its biological importance, the mechanism(s) driving XP remains unclear. Here, we show that a cDC-specific pore-forming protein called apolipoprotein L 7C (APOL7C) is up-regulated in response to innate immune stimuli and is recruited to phagosomes. Association of APOL7C with phagosomes led to phagosomal rupture and escape of engulfed antigens to the cytosol, where they could be processed via the endogenous MHC class I antigen processing pathway. Accordingly, mice deficient in APOL7C did not efficiently prime CD8+ T cells in response to immunization with bead-bound and cell-associated antigens. Together, our data indicate the presence of dedicated apolipoproteins that mediate the delivery of phagocytosed proteins to the cytosol of activated cDCs to facilitate XP.
Collapse
Affiliation(s)
- Gerone A Gonzales
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Song Huang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Liam Wilkinson
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jenny A Nguyen
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Saif Sikdar
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cécile Piot
- Immunobiology Laboratory, Francis Crick Institute, London, UK
| | - Victor Naumenko
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Jahanara Rajwani
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Cassandra M Wood
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Irene Dinh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melanie Moore
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eymi Cedeño
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Neil McKenna
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maria J Polyak
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Sara Amidian
- Cell Imaging Core, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Nicole L Rosin
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matheus B Carneiro
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Bas Surewaard
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Nathan C Peters
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Christopher H Mody
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
| | - Douglas J Mahoney
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Research Institute, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
| | - Johnathan Canton
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Riddell Centre for Cancer Immunotherapy, Calgary, Alberta, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Disease, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Gestal-Mato U, Herhaus L. Autophagy-dependent regulation of MHC-I molecule presentation. J Cell Biochem 2024; 125:e30416. [PMID: 37126231 DOI: 10.1002/jcb.30416] [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: 01/19/2023] [Revised: 04/03/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023]
Abstract
The major histocompatibility complex (MHC) class I molecules present peptide antigens to MHC class I-restricted CD8+ T lymphocytes to elicit an effective immune response. The conventional antigen-processing pathway for MHC-I presentation depends on proteasome-mediated peptide generation and peptide loading in the endoplasmic reticulum by members of the peptide loading complex. Recent discoveries in this field highlight the role of alternative MHC-I peptide loading and presentation pathways, one of them being autophagy. Autophagy is a cell-intrinsic degradative pathway that ensures cellular homoeostasis and plays critical roles in cellular immunity. In this review article, we discuss the role of autophagy in MHC class I-restricted antigen presentation, elucidating new findings on the crosstalk of autophagy and ER-mediated MHC-I peptide presentation, dendritic cell-mediated cross-presentation and also mechanisms governing immune evasion. A detailed molecular understanding of the key drivers of autophagy-mediated MHC-I modulation holds promising targets to devise effective measures to improve T cell immunotherapies.
Collapse
Affiliation(s)
- Uxia Gestal-Mato
- Goethe University School of Medicine, Institute of Biochemistry II, Frankfurt am Main, Germany
| | - Lina Herhaus
- Goethe University School of Medicine, Institute of Biochemistry II, Frankfurt am Main, Germany
| |
Collapse
|
40
|
Frederico SC, Raphael I, Nisnboym M, Huq S, Schlegel BT, Sneiderman CT, Jackson SA, Jain A, Olin MR, Rood BR, Pollack IF, Hwang EI, Rajasundaram D, Kohanbash G. Transcriptomic observations of intra and extracellular immunotherapy targets for pediatric brain tumors. Expert Rev Clin Immunol 2024; 20:1411-1420. [PMID: 39114885 DOI: 10.1080/1744666x.2024.2390023] [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: 05/14/2024] [Accepted: 08/04/2024] [Indexed: 08/20/2024]
Abstract
OBJECTIVES Despite surgical resection, chemoradiation, and targeted therapy, brain tumors remain a leading cause of cancer-related death in children. Immunotherapy has shown some promise and is actively being investigated for treating childhood brain tumors. However, a critical step in advancing immunotherapy for these patients is to uncover targets that can be effectively translated into therapeutic interventions. METHODS In this study, our team performed a transcriptomic analysis across pediatric brain tumor types to identify potential targets for immunotherapy. Additionally, we assessed components that may impact patient response to immunotherapy, including the expression of genes essential for antigen processing and presentation, inhibitory ligands and receptors, interferon signature, and overall predicted T cell infiltration. RESULTS We observed distinct expression patterns across tumor types. These included elevated expression of antigen genes and antigen processing machinery in some tumor types while other tumors had elevated inhibitory checkpoint receptors, known to be associated with response to checkpoint inhibitor immunotherapy. CONCLUSION These findings suggest that pediatric brain tumors exhibit distinct potential for specific immunotherapies. We believe our findings can guide investigators in their assessment of appropriate immunotherapy classes and targets in pediatric brain tumors.
Collapse
Affiliation(s)
- Stephen C Frederico
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michal Nisnboym
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Sakibul Huq
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brent T Schlegel
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chaim T Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sydney A Jackson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anya Jain
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R Olin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Brian R Rood
- Division of Oncology, Children's National Medical Center, Washington, DC, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugene I Hwang
- Division of Oncology, Children's National Medical Center, Washington, DC, USA
| | | | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Jiang D, Ma Z, Zhang J, Sun Y, Bai T, Liu R, Wang Y, Guan L, Fu S, Sun Y, Li Y, Zhou B, Yang Y, Yang S, Chang Y, Sun B, Yang K. Immunoreactivity Analysis of MHC-I Epitopes Derived from the Nucleocapsid Protein of SARS-CoV-2 via Computation and Vaccination. Vaccines (Basel) 2024; 12:1214. [PMID: 39591116 PMCID: PMC11598499 DOI: 10.3390/vaccines12111214] [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: 09/21/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Since 2019, the SARS-CoV-2 virus has been responsible for the global spread of respiratory illness. As of 1 September 2024, the cumulative number of infections worldwide exceeded 776 million. There are many structural proteins of the virus, among which the SARS-CoV-2 nucleocapsid (N) protein plays a pivotal role in the viral life cycle, participating in a multitude of essential activities following viral invasion. An important antiviral immune response is the major histocompatibility complex (MHC)-restricted differentiation cluster 8 (CD8+) T cell cytotoxicity. Therefore, understanding the immunogenicity of SARS-CoV-2 NP-specific MHC-I-restricted epitopes is highly important. Methods: MHC-I molecules from 11 human leukocyte antigen I (HLA-I) superfamilies with 98% population coverage and 6 mouse H2 alleles were selected. The affinity were screened by IEDB, NetMHCpan, SYFPEITHI, SMMPMBEC and Rankpep. Further immunogenicity and conservative analyses were performed using VaxiJen and BLASTp, respectively. EpiDock was used to simulate molecular docking. Cluster analysis was performed. Selective epitopes were validated by enzyme-linked immunospot (ELISpot) assay and flow cytometry in the mice with pVAX-NPSARS-CoV-2 immunization. Enzyme-Linked Immunosorbent Assay (ELISA) was used to detect whether the preferred epitope induced humoral immunity. Results: There were 64 dominant epitopes for the H-2 haplotype and 238 dominant epitopes for the HLA-I haplotype. Further analysis of immunogenicity and conservation yielded 8 preferred epitopes, and docking simulations were conducted with corresponding MHC-I alleles. The relationships between the NP peptides and MHC-I haplotypes were then determined via two-way hierarchical clustering. ELISA, ELISpot assay, and flow cytometry revealed that the preferred epitope stimulated both humoral and cellular immunity and enhanced cytokine secretion in mice. Conclusions: our study revealed the general patterns among multiple haplotypes within the humans and mice superfamily, providing a comprehensive assessment of the pan-MHC-I immunoreactivity of SARS-CoV-2 NP. Our findings would render prospects for the development and application of epitope-based immunotherapy in lasting viral epidemics.
Collapse
Affiliation(s)
- Dongbo Jiang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Zilu Ma
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Junqi Zhang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yubo Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Tianyuan Bai
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Ruibo Liu
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yongkai Wang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Liang Guan
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Shuaishuai Fu
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yuanjie Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yuanzhe Li
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Bingquan Zhou
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yulin Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Shuya Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Yuanhang Chang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| | - Baozeng Sun
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
- Yingtan Detachment, Jiangxi General Hospital, Chinese People’s Armed Police Force, Nanchang 330001, China
- General Practice Medicine Base of Shanghai Changzheng Hospital, Shanghai 200041, China
| | - Kun Yang
- Department of Immunology, The Key Laboratory of Bio-Hazard Damage and Prevention Medicine, Basic Medicine School, Air Force Medical University (In Former The Fourth Military Medical University), Xi’an 710032, China; (D.J.); (Z.M.); (J.Z.); (Y.S.); (T.B.); (R.L.); (Y.W.); (L.G.); (S.F.); (Y.S.); (Y.L.); (B.Z.); (Y.Y.); (S.Y.); (Y.C.)
| |
Collapse
|
42
|
Wan L, Xie B, Shuda M, Delgoffe G, Chang Y, Moore PS. Engineered protein destabilization reverses intrinsic immune evasion for candidate vaccine pan-strain KSHV and SARS-CoV-2 antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619692. [PMID: 39484438 PMCID: PMC11526888 DOI: 10.1101/2024.10.22.619692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Both Kaposi sarcoma herpesvirus LANA and SARS coronavirus 2 RdRp/nsp12 are highly conserved replication proteins that evade immune processing. By deleting the LANA central repeat 1 domain (LANA ΔCR1 ) or by dividing RdRp into two separated fragments (RdRp Frag ) to maximize nascent protein mis-folding, cis peptide presentation was increased. Native LANA or RdRp SIINFEKL fusion proteins expressed in MC38 cancer cells were not recognized by activated OT-1 CD8 + cells against SIINFEKL but cytotoxic recognition was restored by expression of the corresponding modified proteins. Immunocompetent syngeneic mice injected with LANA- or RdRp-SIINFEKL MC38 cells developed rapidly-growing tumors with short median survival times. Mice injected with LANA ΔCR1 - or RdRp Frag -SIINFEKL had partial tumor regression, slower tumor growth, longer median survival, as well as increased effector-specific tumor-infiltrating lymphocytes. These mice developed robust T cell responses lasting at least 90 days post-injection that recognized native viral protein epitopes. Engineered vaccine candidate antigens can unmask virus-specific CTL responses that are typically suppressed during native viral infection.
Collapse
|
43
|
Song X, Li Y, Wu H, Qiu H, Sun Y. T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases. Vaccines (Basel) 2024; 12:1181. [PMID: 39460347 PMCID: PMC11511246 DOI: 10.3390/vaccines12101181] [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: 08/26/2024] [Revised: 10/06/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
With the development of novel vaccine strategies, T-cell epitope-based vaccines have become promising prophylactic and therapeutic tools against infectious diseases that cannot be controlled via traditional vaccines. T-cell epitope-based vaccines leverage specific immunogenic peptides to elicit protective T-cell responses against infectious pathogens. Compared to traditional vaccines, they provide superior efficacy and safety, minimizing the risk of adverse side effects. In this review, we summarized and compared the prediction and identification methods of T-cell epitopes. By integrating bioinformatic prediction and experimental validation, efficient and precise screening of T-cell epitopes can be achieved. Importantly, we delved into the development approaches to diverse T-cell epitope-based vaccines, comparing their merits and demerits, as well as discussing the prevalent challenges and perspectives in their applications. This review offers fresh perspectives for the formulation of safe and efficacious epitope-based vaccines for the devastating diseases against which no vaccines are currently available.
Collapse
Affiliation(s)
| | | | | | - Huaji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.S.); (Y.L.); (H.W.)
| | - Yuan Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.S.); (Y.L.); (H.W.)
| |
Collapse
|
44
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [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: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
45
|
Montauti E, Oh DY, Fong L. CD4 + T cells in antitumor immunity. Trends Cancer 2024; 10:969-985. [PMID: 39242276 PMCID: PMC11464182 DOI: 10.1016/j.trecan.2024.07.009] [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: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Advances in cancer immunotherapy have transformed cancer care and realized unprecedented responses in many patients. The growing arsenal of novel therapeutics - including immune checkpoint inhibition (ICI), adoptive T cell therapies (ACTs), and cancer vaccines - reflects the success of cancer immunotherapy. The therapeutic benefits of these treatment modalities are generally attributed to the enhanced quantity and quality of antitumor CD8+ T cell responses. Nevertheless, CD4+ T cells are now recognized to play key roles in both the priming and effector phases of the antitumor immune response. In addition to providing T cell help through co-stimulation and cytokine production, CD4+ T cells can also possess cytotoxicity either directly on MHC class II-expressing tumor cells or to other cells within the tumor microenvironment (TME). The presence of specific populations of CD4+ T cells, and their intrinsic plasticity, within the TME can represent an important determinant of clinical response to immune checkpoint inhibitors, vaccines, and chimeric antigen receptor (CAR) T cell therapies. Understanding how the antitumor functions of specific CD4+ T cell types are induced while limiting their protumorigenic attributes will enable more successful immunotherapies.
Collapse
Affiliation(s)
- Elena Montauti
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Immunotherapy Integrated Research Center, Fred Hutchison Cancer Center, Seattle, WA, USA.
| |
Collapse
|
46
|
Löffler M, Frühschulz S, Rockel Z, Pečak M, Tampé R, Wieneke R. Antigen Delivery Controlled by an On-Demand Photorelease. Angew Chem Int Ed Engl 2024; 63:e202405035. [PMID: 38818622 DOI: 10.1002/anie.202405035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/18/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
To eliminate infected and cancerous cells, antigen processing and presentation play a pivotal role through the recognition of antigenic peptides displayed on Major Histocompatibility Complex class I (MHC I) molecules. Here, we developed a photostimulated antigen release system that enables the temporal inception of antigen flux. Simple and effective photocaging of the human immunodeficiency virus (HIV)-Nef73-derived epitope, a representative high-affinity MHC I ligand, was provided by steric hindrance to block the recognition by the transporter associated with antigen processing (TAP) in the peptide loading complex (PLC). In response to light, a heteronomous release of antigens and subsequent translocation in various scenarios is demonstrated, including a TAP-related ATP-binding cassette (ABC) transporter reconstituted in liposomes and the native PLC in the endoplasmic reticulum (ER) membrane of human cells. The photochemically induced 'burst' of antigens opens new opportunities for a mechanistic analysis of the antigen translocation machinery and will help to provide insights into antigen processing pathways via an on-demand, subcellular pulse-chase release of antigens.
Collapse
Affiliation(s)
- Max Löffler
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Stefan Frühschulz
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Zoe Rockel
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Matija Pečak
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| |
Collapse
|
47
|
Elsayed A, von Hardenberg S, Atschekzei F, Graalmann T, Jänke C, Witte T, Ringshausen FC, Sogkas G. Phenotypic and pathomechanistic overlap between tapasin and TAP deficiencies. J Allergy Clin Immunol 2024; 154:1069-1075. [PMID: 38866210 DOI: 10.1016/j.jaci.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Human tapasin deficiency is reported to cause an autosomal-recessive inborn error of immunity characterized by substantially reduced cell surface expression of major histocompatibility complex class I (MHC-I). OBJECTIVE We evaluated the immunologic and clinical consequences of tapasin deficiency. METHODS A novel homozygous variant in TAPBP was identified by means of whole genome sequencing. The expression of tapasin and both subunits of the transporter associated with antigen presentation (TAP) were evaluated by Western blot analysis. Cell surface and intracellular expression of MHC-I were evaluated by flow cytometry. Small interfering RNAs were used for silencing TAPBP expression in HEK293T cells. RESULTS We identified a deletion in TAPBP (c.312del, p.(K104Nfs∗6)) causing tapasin deficiency in a patient with bronchiectasis and recurrent respiratory tract infections as well as herpes zoster. Besides substantial reduction in TAP1 and TAP2 expression, peripheral blood mononuclear cells from this patient and TAPBP-knockdown HEK293T cells, displayed reduced cell surface expression of MHC-I, while reduction in intracellular expression of MHC-I was less prominent, suggesting a defect in MHC-I trafficking to the plasma membrane. IFN-α improved cell surface expression of MHC-I in tapasin deficient lymphocytes and TAPBP-knockdown HEK293T cells, representing a possible therapeutic approach for tapasin deficiency. CONCLUSION Tapasin deficiency is a very rare inborn error of immunity, the pathomechanism and clinical spectrum of which overlaps with TAP deficiencies.
Collapse
Affiliation(s)
- Abdulwahab Elsayed
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | | | - Faranaz Atschekzei
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Theresa Graalmann
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Junior Research Group for Translational Immunology, TWINCORE, Center for Infection Research and the Hannover Medical School, Hannover, Germany; Biomedical Research in End-Stage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Christine Jänke
- Junior Research Group for Translational Immunology, TWINCORE, Center for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Felix C Ringshausen
- Biomedical Research in End-Stage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany; Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; European Reference Network on Rare and Complex Respiratory Diseases (ERN-LUNG), Frankfurt, Germany
| | - Georgios Sogkas
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
48
|
van de Weijer ML, Samanta K, Sergejevs N, Jiang L, Dueñas ME, Heunis T, Huang TY, Kaufman RJ, Trost M, Sanyal S, Cowley SA, Carvalho P. Tapasin assembly surveillance by the RNF185/Membralin ubiquitin ligase complex regulates MHC-I surface expression. Nat Commun 2024; 15:8508. [PMID: 39353943 PMCID: PMC11445256 DOI: 10.1038/s41467-024-52772-x] [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: 02/21/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024] Open
Abstract
Immune surveillance by cytotoxic T cells eliminates tumor cells and cells infected by intracellular pathogens. This process relies on the presentation of antigenic peptides by Major Histocompatibility Complex class I (MHC-I) at the cell surface. The loading of these peptides onto MHC-I depends on the peptide loading complex (PLC) at the endoplasmic reticulum (ER). Here, we uncovered that MHC-I antigen presentation is regulated by ER-associated degradation (ERAD), a protein quality control process essential to clear misfolded and unassembled proteins. An unbiased proteomics screen identified the PLC component Tapasin, essential for peptide loading onto MHC-I, as a substrate of the RNF185/Membralin ERAD complex. Loss of RNF185/Membralin resulted in elevated Tapasin steady state levels and increased MHC-I at the surface of professional antigen presenting cells. We further show that RNF185/Membralin ERAD complex recognizes unassembled Tapasin and limits its incorporation into PLC. These findings establish a novel mechanism controlling antigen presentation and suggest RNF185/Membralin as a potential therapeutic target to modulate immune surveillance.
Collapse
Affiliation(s)
- Michael L van de Weijer
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Krishna Samanta
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Nikita Sergejevs
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - LuLin Jiang
- Degenerative Diseases Program, Genetics, and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
- Altos Labs-Bay Institute of Science, Redwood City, CA, USA
| | - Maria Emilia Dueñas
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
- Telethon Kids Institute, Perth, Nedlands, WA, 6009, Australia
| | - Tiaan Heunis
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Immunocore Ltd, 92 Park Drive, Abingdon, OX14 4RY, UK
| | - Timothy Y Huang
- Degenerative Diseases Program, Genetics, and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Genetics, and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Matthias Trost
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Sally A Cowley
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| |
Collapse
|
49
|
Gent V, Lu YJ, Lukhele S, Dhar N, Dangor Z, Hosken N, Malley R, Madhi SA, Kwatra G. Surface protein distribution in Group B Streptococcus isolates from South Africa and identifying vaccine targets through in silico analysis. Sci Rep 2024; 14:22665. [PMID: 39349584 PMCID: PMC11442663 DOI: 10.1038/s41598-024-73175-4] [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: 04/17/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Group B Streptococcus (GBS) is a major cause of pneumonia, sepsis, and meningitis in infants younger than 3 months of age. Furthermore, GBS infection in pregnant women is associated with stillbirths and pre-term delivery. It also causes disease in immunocompromised adults and the elderly, but the highest incidence of the disease occurs in neonates and young infants. At this time, there are no licensed vaccines against GBS. Complete GBS genome sequencing has helped identify genetically conserved and immunogenic proteins, which could serve as vaccine immunogens. In this study, in silico reverse vaccinology method were used to evaluate the prevalence and conservation of GBS proteins in invasive and colonizing isolates from South African infants and women, respectively. Furthermore, this study aimed to predict potential GBS vaccine targets by evaluating metrics such as antigenicity, physico-chemical properties, subcellular localization, secondary and tertiary structures, and epitope prediction and conservation. A total of 648 invasive and 603 colonizing GBS isolate sequences were screened against a panel of 89 candidate GBS proteins. Ten of the 89 proteins were highly genetically conserved in invasive and colonizing GBS isolates, nine of which were computationally inferred proteins (gbs2106, SAN_1577, SAN_0356, SAN_1808, SAN_1685, SAN_0413, SAN_0990, SAN_1040, SAN_0226) and one was the surface Immunogenic Protein (SIP). Additionally, the nine proteins were predicted to be more antigenic than the SIP protein (antigenicity score of > 0.6498), highlighting their potential as GBS vaccine antigen targets.
Collapse
Affiliation(s)
- Vicky Gent
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ying-Jie Lu
- Division of Infectious Diseases, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sindiswa Lukhele
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nisha Dhar
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Dangor
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nancy Hosken
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Richard Malley
- Division of Infectious Diseases, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shabir A Madhi
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- South African Medical Research Council: Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Department of Clinical Microbiology, Christian Medical College, Vellore, India.
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
50
|
Xanthopoulou E, Lamprou I, Mitrakas AG, Michos GD, Zois CE, Giatromanolaki A, Harris AL, Koukourakis MI. Autophagy Blockage Up-Regulates HLA-Class-I Molecule Expression in Lung Cancer and Enhances Anti-PD-L1 Immunotherapy Efficacy. Cancers (Basel) 2024; 16:3272. [PMID: 39409895 PMCID: PMC11476265 DOI: 10.3390/cancers16193272] [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: 08/30/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Immune checkpoint inhibitors have an established role in non-small cell lung cancer (NSCLC) therapy. The loss of HLA-class-I expression allows cancer cell evasion from immune surveillance, disease progression, and failure of immunotherapy. The restoration of HLA-class-I expression may prove to be a game-changer in current immunotherapy strategies. Autophagic activity has been recently postulated to repress HLA-class-I expression in cancer cells. METHODS NSCLC cell lines (A549 and H1299) underwent late-stage (chloroquine and bafilomycin) and early-stage autophagy blockage (ULK1 inhibitors and MAP1LC3A silencing). The HLA-class-I expression was assessed with flow cytometry, a Western blot, and RT-PCR. NSCLC tissues were examined for MAP1LC3A and HLA-class-I expression using double immunohistochemistry. CD8+ T-cell cytotoxicity was examined in cancer cells pre-incubated with chloroquine and anti-PD-L1 monoclonal antibodies (Moabs); Results: A striking increase in HLA-class-I expression following incubation with chloroquine, bafilomycin, and IFNγ was noted in A549 and H1299 cancer cells, respectively. This effect was further confirmed in CD133+ cancer stem cells. HLA-class-I, β2-microglobulin, and TAP1 mRNA levels remained stable. Prolonged exposure to chloroquine further enhanced HLA-class-I expression. Similar results were noted following exposure to a ULK1 and a PIKfyve inhibitor. Permanent silencing of the MAP1LC3A gene resulted in enhanced HLA-class-I expression. In immunohistochemistry experiments, double LC3A+/HLA-class-I expression was seldom. Pre-incubation of H1299 cancer cells with chloroquine and anti-PD-L1 MoAbs increased the mean % of apoptotic/necrotic cells from 2.5% to 18.4%; Conclusions: Autophagy blockers acting either at late or early stages of the autophagic process may restore HLA-class-I-mediated antigen presentation, eventually leading to enhanced immunotherapy efficacy.
Collapse
Affiliation(s)
- Erasmia Xanthopoulou
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (I.L.); (A.G.M.); (G.D.M.); (C.E.Z.)
| | - Ioannis Lamprou
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (I.L.); (A.G.M.); (G.D.M.); (C.E.Z.)
| | - Achilleas G. Mitrakas
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (I.L.); (A.G.M.); (G.D.M.); (C.E.Z.)
| | - Georgios D. Michos
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (I.L.); (A.G.M.); (G.D.M.); (C.E.Z.)
| | - Christos E. Zois
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (I.L.); (A.G.M.); (G.D.M.); (C.E.Z.)
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Alexandra Giatromanolaki
- Department of Pathology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Adrian L. Harris
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.X.); (I.L.); (A.G.M.); (G.D.M.); (C.E.Z.)
| |
Collapse
|