Generation of SARS-CoV-2 S1 Spike Glycoprotein Putative Antigenic Epitopes in Vitro by Intracellular AminopeptidasesClick to copy article linkArticle link copied!
- George StamatakisGeorge StamatakisBiomedical Sciences Research Center “Alexander Fleming”, 16672 Vari, Attica, GreeceMore by George Stamatakis
- Martina SamiotakiMartina SamiotakiBiomedical Sciences Research Center “Alexander Fleming”, 16672 Vari, Attica, GreeceMore by Martina Samiotaki
- Anastasia MpakaliAnastasia MpakaliNational Centre for Scientific Research “Demokritos”, 15310 Agia Paraskevi, Attica, GreeceMore by Anastasia Mpakali
- George PanayotouGeorge PanayotouBiomedical Sciences Research Center “Alexander Fleming”, 16672 Vari, Attica, GreeceMore by George Panayotou
- Efstratios Stratikos*Efstratios Stratikos*Email: [email protected], [email protected]National Centre for Scientific Research “Demokritos”, 15310 Agia Paraskevi, Attica, GreeceMore by Efstratios Stratikos
Abstract
Presentation of antigenic peptides by MHCI is central to cellular immune responses against viral pathogens. While adaptive immune responses versus SARS-CoV-2 can be of critical importance to both recovery and vaccine efficacy, how protein antigens from this pathogen are processed to generate antigenic peptides is largely unknown. Here, we analyzed the proteolytic processing of overlapping precursor peptides spanning the entire sequence of the S1 spike glycoprotein of SARS-CoV-2, by three key enzymes that generate antigenic peptides, aminopeptidases ERAP1, ERAP2, and IRAP. All enzymes generated shorter peptides with sequences suitable for binding onto HLA alleles, but with distinct specificity fingerprints. ERAP1 was the most efficient in generating peptides 8–11 residues long, the optimal length for HLA binding, while IRAP was the least efficient. The combination of ERAP1 with ERAP2 greatly limited the variability of peptide sequences produced. Less than 7% of computationally predicted epitopes were found to be produced experimentally, suggesting that aminopeptidase processing may constitute a significant filter to epitope presentation. These experimentally generated putative epitopes could be prioritized for SARS-CoV-2 immunogenicity studies and vaccine design. We furthermore propose that this in vitro trimming approach could constitute a general filtering method to enhance the prediction robustness for viral antigenic epitopes.
SPECIAL ISSUE
This article is part of the
Note
This article is made available via the ACS COVID-19 subset for unrestricted RESEARCH re-use and analyses in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.
Introduction
Experimental Procedures
Enzyme Expression and Purification
Peptides
Enzymatic Reactions
LC-MS/MS Analysis
Database Search
Results and Discussion
ERAP1 | ERAP2 | IRAP | ||||||
---|---|---|---|---|---|---|---|---|
peptide | score | allele | peptide | score | allele | peptide | score | allele |
KFLPFQQF | 0.01 | A2402 | EVFNATRF | 0.00 | A2601 | EVFNATRF | 0.00 | A2601 |
VYYPDKVF | 0.01 | A2402 | VLNDILSR | 0.01 | A0301 | VLNDILSR | 0.01 | A0301 |
GYLQPRTF | 0.05 | A2402 | GRLQSLQTY | 0.06 | B2705 | HADQLTPTW | 0.02 | B5801 |
VRFPNITNL | 0.05 | B2705 | YRFNGIGV | 0.08 | B2705 | GYLQPRTF | 0.05 | A2402 |
GRLQSLQTY | 0.06 | B2705 | NQKLIANQF | 0.08 | B1501 | LADAGFIKQY | 0.09 | A0101 |
YRFNGIGV | 0.08 | B2705 | DAGFIKQY | 0.10 | A2601 | DAGFIKQY | 0.10 | A2601 |
NQKLIANQF | 0.08 | B1501 | SLGAENSVAY | 0.12 | B1501 | NYNYLYRL | 0.18 | A2402 |
NLREFVFK | 0.09 | A0301 | VFKNIDGYFKI | 0.14 | A2402 | STEKSNIIRGW | 0.21 | B5801 |
DAGFIKQY | 0.10 | A2601 | STEKSNIIRGW | 0.21 | B5801 | TLADAGFIK | 0.25 | A0301 |
DSFKEELDKY | 0.10 | A2601 | ILSRLDKV | 0.24 | A0201 | GVYYPDKVF | 0.32 | B1501 |
SVASQSIIAY | 0.11 | B1501 | FKEELDKY | 0.24 | A0101 | TLADAGFIKQY | 0.39 | A2601 |
TYVPAQEKNF | 0.20 | A2402 | TRFASVYAWNR | 0.25 | B2705 | ALNTLVKQL | 0.48 | A0201 |
KRFDNPVLPFN | 0.21 | B2705 | QRNFYEPQI | 0.35 | B2705 | KVTLADAGFIK | 0.48 | A0301 |
TLADAGFIK | 0.25 | A0301 | IEDLLFNK | 0.38 | A0101 | YADSFVIR | 0.49 | A0101 |
GVYYPDKVF | 0.32 | B1501 | LPIGINITRF | 0.39 | B0702 | PFGEVFNATRF | 0.52 | A2402 |
ATRFASVY | 0.34 | A0101 | LGAENSVAY | 0.41 | B1501 | GAGAALQI | 0.57 | B5801 |
GVYYPDKV | 0.39 | A0201 | LPQGFSAL | 0.47 | B0702 | APHGVVFL | 0.61 | B0702 |
SVLNDILSR | 0.42 | A0301 | TRFQTLLA | 0.49 | B2705 | NIDGYFKI | 0.65 | A0101 |
LPQGFSAL | 0.47 | B0702 | YADSFVIR | 0.49 | A0101 | ITGRLQSLQTY | 0.68 | A0101 |
KVTLADAGFIK | 0.48 | A0301 | TPINLVRDL | 0.50 | B0702 | TRGVYYPDKVF | 0.78 | B2705 |
YADSFVIR | 0.49 | A0101 | APHGVVFL | 0.61 | B0702 | QLTPTWRV | 0.79 | A0201 |
TPINLVRDL | 0.50 | B0702 | DPLSETKCTL | 0.66 | B0702 | YPDKVFRSSV | 0.83 | B0702 |
NYKLPDDF | 0.56 | A2402 | ALGKLQDV | 0.66 | A0201 | VLYENQKLI | 0.84 | A0201 |
NFYEPQII | 0.57 | A2402 | ITGRLQSLQTY | 0.68 | A0101 | NTLVKQLSSNF | 0.89 | A2601 |
NIDGYFKI | 0.65 | A0101 | TRGVYYPDKVF | 0.78 | B2705 | TTDNTFVS | 0.96 | A0101 |
DPLSETKCTL | 0.66 | B0702 | QLTPTWRV | 0.79 | A0201 | LPPAYTNSF | 1.10 | B0702 |
ALGKLQDV | 0.66 | A0201 | GINITRFQTL | 0.79 | B0801 | QRNFYEPQII | 1.15 | B2705 |
NESLIDLQEL | 0.67 | B4001 | QPYRVVVLSF | 0.82 | B0702 | QPRTFLLKY | 1.16 | A0101 |
FVIRGDEV | 0.71 | A0201 | LYENQKLI | 0.83 | A2402 | NVYADSFVIR | 1.19 | A0301 |
QLTPTWRV | 0.79 | A0201 | YPDKVFRSSV | 0.83 | B0702 | GEVFNATRF | 1.21 | B4001 |
QPYRVVVLSF | 0.82 | B0702 | VLYENQKLI | 0.84 | A0201 | EELDKYFKNH | 1.24 | A2601 |
LYENQKLI | 0.83 | A2402 | GVLTESNKKF | 0.87 | A2601 | YYPDKVFRSSV | 1.24 | A2402 |
YPDKVFRSSV | 0.83 | B0702 | VRDLPQGFSAL | 0.93 | B2705 | AEVQIDRLITG | 1.29 | B4001 |
KNIDGYFKI | 0.84 | A2402 | DPLQPELDSF | 1.09 | B0702 | EPLVDLPI | 1.30 | B0702 |
VLYENQKLI | 0.84 | A0201 | LPPAYTNSF | 1.10 | B0702 | TGRLQSLQTY | 1.31 | B1501 |
VYADSFVIR | 0.85 | A2402 | DILSRLDKV | 1.11 | B0801 | SVLHSTQDLFL | 1.33 | A0201 |
TVYDPLQP | 0.88 | A0301 | NGIGVTQNVLY | 1.12 | A2601 | YGVSPTKL | 1.37 | A2601 |
HFPREGVF | 0.89 | A2402 | QRNFYEPQII | 1.15 | B2705 | VTLADAGFIK | 1.39 | A0301 |
QDVVNQNAQAL | 0.93 | B4001 | QPRTFLLKY | 1.16 | A0101 | LPFQQFGRDIA | 1.44 | B0801 |
FEYVSQPF | 0.96 | B4001 | EELDKYFKNH | 1.24 | A2601 | QKFNGLTVLPP | 1.45 | B2705 |
SIIAYTMSL | 1.04 | B0801 | LPFQQFGRDI | 1.29 | B0702 | DGYFKIYSKH | 1.55 | A2601 |
KKFLPFQQFGR | 1.11 | B2705 | EPLVDLPI | 1.30 | B0702 | YENQKLIANQF | 1.55 | B1501 |
DILSRLDKV | 1.11 | B0801 | TGRLQSLQTY | 1.31 | B1501 | FPREGVFVSN | 1.56 | B0702 |
QPRTFLLKY | 1.16 | A0101 | QKLIANQF | 1.31 | B1501 | GNYNYLYRL | 1.63 | B2705 |
NVYADSFVIR | 1.19 | A0301 | TTEILPVSM | 1.32 | A0101 | NATRFASVY | 1.82 | A2601 |
GEVFNATRF | 1.21 | B4001 | SPDVDLGDISG | 1.33 | B0702 | YRLFRKSNLKP | 1.83 | B2705 |
EELDKYFKNH | 1.24 | A2601 | DVVIGIVNN | 1.42 | A2601 | HADQLTPTWRV | 1.98 | A0101 |
QKLIANQF | 1.31 | B1501 | LPFQQFGRDIA | 1.44 | B0801 | YVTQQLIRA | 2.00 | A0201 |
TTEILPVSM | 1.32 | A0101 | QKFNGLTVLPP | 1.45 | B2705 | |||
SLLIVNNA | 1.37 | A0201 | DIADTTDAVRD | 1.52 | A2601 | |||
YGVSPTKL | 1.37 | A2601 | DGYFKIYSKH | 1.55 | A2601 | |||
VTLADAGFIK | 1.39 | A0301 | GNYNYLYRL | 1.63 | B2705 | |||
VGYQPYRV | 1.42 | A0201 | SRLDKVEAEV | 1.64 | B2705 | |||
DIADTTDAVRD | 1.52 | A2601 | NATRFASVY | 1.82 | A2601 | |||
DGYFKIYSKH | 1.55 | A2601 | YRLFRKSNLKP | 1.83 | B2705 | |||
RDLPQGFSAL | 1.55 | B4001 | HADQLTPTWRV | 1.98 | A0101 | |||
LEPLVDLPI | 1.64 | B4001 | YVTQQLIRA | 2.00 | A0201 | |||
SRLDKVEAEV | 1.64 | B2705 | ||||||
NATRFASVY | 1.82 | A2601 | ||||||
YVTQQLIRA | 2.00 | A0201 |
HLA-A01:01, HLA-A02:01, HLA-A03:01, HLA-A24:02, HLA-A26:01, HLA-B07:02, HLA-B08:01, HLA-B27:05, HLA-B39:01, HLA-B40:01, HLA-B58:01, and HLA-B15:01. Scores indicated are percentile ranks corresponding to the predicted affinity score for each allele (range 0–100, 0 is best, ranks below 2 are considered binders). In bold are peptides reported to be antigenic for SARS-CoV-1 found in the immune epitope database (http://www.iedb.org/).
ERAP1 | ERAP2 | IRAP | ERAP1/ERAP2 | ||||
---|---|---|---|---|---|---|---|
sequence | score | sequence | score | sequence | score | sequence | score |
HLA-B15:03 | HLA-B15:03 | HLA-B15:03 | HLA-B15:03 | ||||
ATRFASVY | 0.37 | VGYLQPRTF | 0.53 | VGYLQPRTF | 0.53 | VGYLQPRTF | 0.54 |
VGYLQPRTF | 0.53 | NQKLIANQF | 0.58 | LKYNENGTITD | 1.40 | NQKLIANQF | 0.58 |
NQKLIANQF | 0.58 | GRLQSLQTY | 0.65 | GYLQPRTF | 1.59 | LKYNENGTITD | 1.41 |
GRLQSLQTY | 0.65 | LGAENSVAY | 0.84 | GEVFNATRF | 1.63 | ||
FEYVSQPF | 0.83 | QKLIANQF | 0.91 | GVYYPDKVF | 1.71 | ||
QKLIANQF | 0.91 | LKYNENGTITD | 1.40 | AGAALQIPF | 1.91 | ||
KAHFPREGVF | 1.34 | SLGAENSVAY | 1.48 | ||||
LKYNENGTITD | 1.40 | NGIGVTQNVLY | 1.89 | ||||
AQYTSALLA | 1.42 | ||||||
KRFDNPVLPFN | 1.58 | ||||||
GYLQPRTF | 1.59 | ||||||
SVASQSIIAY | 1.59 | ||||||
GEVFNATRF | 1.63 | ||||||
GVYYPDKVF | 1.71 | ||||||
VYYPDKVF | 1.80 | ||||||
HLA-B46:01 | HLA-B46:01 | HLA-B46:01 | HLA-B46:01 | ||||
FEYVSQPF | 0.62 | LGAENSVAY | 0.037 | TLADAGFIKQY | 0.08 | – | |
SIIAYTMSL | 0.64 | LPIGINITRF | 0.71 | YVGYLQPRTF | 1.66 | ||
ATRFASVY | 0.65 | SLGAENSVAY | 0.72 | GVYYPDKVF | 1.75 | ||
SVASQSIIAY | 0.91 | GVLTESNKKF | 1.34 | TGRLQSLQTY | 1.98 | ||
IANQFNSAI | 1.25 | YVGYLQPRTF | 1.65 | ||||
GVYYPDKVF | 1.75 | NGIGVTQNVLY | 1.84 | ||||
FKEELDKY | 1.90 | ||||||
TGRLQSLQTY | 1.98 |
Scores indicated are percentile ranks corresponding to the predicted score for each allele (range 0–100, 0 is best, ranks below 2 are considered binders).
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jproteome.0c00457.
Table S1: List of generated peptides after digestion by each enzyme (XLSX)
Table S2: List of generated 8–11mers from each enzyme with best predicted binding score based on the HLAthena server (XLSX)
Table S3: Predicted binding score of peptides from the S1 spike glycoprotein of SARS-CoV-2 for HLA-B15:03 and HLA-B46:01, based on the HLAthena prediction server; in red, the peptides that are predicted to be binders (XLSX)
Table S4: List of potential epitopes from the sequence of the S1 spike glycoprotein of SARS-CoV-2 predicted using the NetMHCpan 4.1 server (XLSX)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
References
This article references 45 other publications.
- 1Coronaviridae Study Group of the International Committee on Taxonomy of Viruses The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5 (4), 536– 544, DOI: 10.1038/s41564-020-0695-zGoogle Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXktFemu7Y%253D&md5=3e43e7781469c6d748726b24b3a60361The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2Coronaviridae Study Group of the International Committee on Taxonomy of VirusesNature Microbiology (2020), 5 (4), 536-544CODEN: NMAICH; ISSN:2058-5276. (Nature Research)A review. The present outbreak of a coronavirus-assocd. acute respiratory disease called coronavirus disease 19 (COVID-19) is the third documented spillover of an animal coronavirus to humans in only two decades that has resulted in a major epidemic. The Coronaviridae Study Group (CSG) of the International Committee on Taxonomy of Viruses, which is responsible for developing the classification of viruses and taxon nomenclature of the family Coronaviridae, has assessed the placement of the human pathogen, tentatively named 2019-nCoV, within the Coronaviridae. Based on phylogeny, taxonomy and established practice, the CSG recognizes this virus as forming a sister clade to the prototype human and bat severe acute respiratory syndrome coronaviruses (SARS-CoVs) of the species Severe acute respiratory syndrome-related coronavirus, and designates it as SARS-CoV-2. In order to facilitate communication, the CSG proposes to use the following naming convention for individual isolates: SARS-CoV-2/host/location/isolate/date. While the full spectrum of clin. manifestations assocd. with SARS-CoV-2 infections in humans remains to be detd., the independent zoonotic transmission of SARS-CoV and SARS-CoV-2 highlights the need for studying viruses at the species level to complement research focused on individual pathogenic viruses of immediate significance. This will improve our understanding of virus-host interactions in an ever-changing environment and enhance our preparedness for future outbreaks.
- 2Wu, F.; Zhao, S.; Yu, B.; Chen, Y. M.; Wang, W.; Song, Z. G.; Hu, Y.; Tao, Z. W.; Tian, J. H.; Pei, Y. Y.; Yuan, M. L.; Zhang, Y. L.; Dai, F. H.; Liu, Y.; Wang, Q. M.; Zheng, J. J.; Xu, L.; Holmes, E. C.; Zhang, Y. Z. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579 (7798), 265– 269, DOI: 10.1038/s41586-020-2008-3Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksFKlsLc%253D&md5=0163a684829e880a0c3347e19f0ce52aA new coronavirus associated with human respiratory disease in ChinaWu, Fan; Zhao, Su; Yu, Bin; Chen, Yan-Mei; Wang, Wen; Song, Zhi-Gang; Hu, Yi; Tao, Zhao-Wu; Tian, Jun-Hua; Pei, Yuan-Yuan; Yuan, Ming-Li; Zhang, Yu-Ling; Dai, Fa-Hui; Liu, Yi; Wang, Qi-Min; Zheng, Jiao-Jiao; Xu, Lin; Holmes, Edward C.; Zhang, Yong-ZhenNature (London, United Kingdom) (2020), 579 (7798), 265-269CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Emerging infectious diseases, such as severe acute respiratory syndrome (SARS) and Zika virus disease, present a major threat to public health. Despite intense research efforts, how, when and where new diseases appear are still a source of considerable uncertainty. A severe respiratory disease was recently reported in Wuhan, Hubei province, China. As of 25 Jan. 2020, at least 1,975 cases had been reported since the first patient was hospitalized on 12 Dec. 2019. Epidemiol. investigations have suggested that the outbreak was assocd. with a seafood market in Wuhan. Here we study a single patient who was a worker at the market and who was admitted to the Central Hospital of Wuhan on 26 Dec. 2019 while experiencing a severe respiratory syndrome that included fever, dizziness and a cough. Metagenomic RNA sequencing of a sample of bronchoalveolar lavage fluid from the patient identified a new RNA virus strain from the family Coronaviridae, which is designated here 'WH-Human 1' coronavirus (and has also been referred to as '2019-nCoV'). Phylogenetic anal. of the complete viral genome (29,903 nucleotides) revealed that the virus was most closely related (89.1% nucleotide similarity) to a group of SARS-like coronaviruses (genus Betacoronavirus, subgenus Sarbecovirus) that had previously been found in bats in China. This outbreak highlights the ongoing ability of viral spill-over from animals to cause severe disease in humans.
- 3Zhou, P.; Yang, X. L.; Wang, X. G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H. R.; Zhu, Y.; Li, B.; Huang, C. L.; Chen, H. D.; Chen, J.; Luo, Y.; Guo, H.; Jiang, R. D.; Liu, M. Q.; Chen, Y.; Shen, X. R.; Wang, X.; Zheng, X. S.; Zhao, K.; Chen, Q. J.; Deng, F.; Liu, L. L.; Yan, B.; Zhan, F. X.; Wang, Y. Y.; Xiao, G. F.; Shi, Z. L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579 (7798), 270– 273, DOI: 10.1038/s41586-020-2012-7Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksFKlsLg%253D&md5=236f17d4d3c7978d72513e5e0258f1b3A pneumonia outbreak associated with a new coronavirus of probable bat originZhou, Peng; Yang, Xing-Lou; Wang, Xian-Guang; Hu, Ben; Zhang, Lei; Zhang, Wei; Si, Hao-Rui; Zhu, Yan; Li, Bei; Huang, Chao-Lin; Chen, Hui-Dong; Chen, Jing; Luo, Yun; Guo, Hua; Jiang, Ren-Di; Liu, Mei-Qin; Chen, Ying; Shen, Xu-Rui; Wang, Xi; Zheng, Xiao-Shuang; Zhao, Kai; Chen, Quan-Jiao; Deng, Fei; Liu, Lin-Lin; Yan, Bing; Zhan, Fa-Xian; Wang, Yan-Yi; Xiao, Geng-Fu; Shi, Zheng-LiNature (London, United Kingdom) (2020), 579 (7798), 270-273CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large no. of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats1-4. Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans5-7. Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 Dec. 2019, had caused 2,794 lab.-confirmed infections including 80 deaths by 26 Jan. 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence anal. of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addn., 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor-angiotensin converting enzyme II (ACE2)-as SARS-CoV.
- 4Tortorici, M. A.; Veesler, D. Structural insights into coronavirus entry. Adv. Virus Res. 2019, 105, 93– 116, DOI: 10.1016/bs.aivir.2019.08.002Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1aisLfE&md5=7c7d72fb74a490b922fe2da65c8615fbStructural insights into coronavirus entryTortorici, M. Alejandra; Veesler, DavidAdvances in Virus Research (2019), 105 (Complementary Strategies to Understand Virus Structure and Function), 93-116CODEN: AVREA8; ISSN:0065-3527. (Elsevier Ltd.)A review. Coronaviruses (CoVs) have caused outbreaks of deadly pneumonia in humans since the beginning of the 21st century. The severe acute respiratory syndrome coronavirus (SARS-CoV) emerged in 2002 and was responsible for an epidemic that spread to five continents with a fatality rate of 10% before being contained in 2003 (with addnl. cases reported in 2004). The Middle-East respiratory syndrome coronavirus (MERS-CoV) emerged in the Arabian Peninsula in 2012 and has caused recurrent outbreaks in humans with a fatality rate of 35%. SARS-CoV and MERS-CoV are zoonotic viruses that crossed the species barrier using bats/palm civets and dromedary camels, resp. No specific treatments or vaccines have been approved against any of the six human coronaviruses, highlighting the need to investigate the principles governing viral entry and cross-species transmission as well as to prep. for zoonotic outbreaks which are likely to occur due to the large reservoir of CoVs found in mammals and birds. Here, we review our understanding of the infection mechanism used by coronaviruses derived from recent structural and biochem. studies.
- 5Walls, A. C.; Park, Y. J.; Tortorici, M. A.; Wall, A.; McGuire, A. T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 181 (2), 281– 292, DOI: 10.1016/j.cell.2020.02.058Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVejsLk%253D&md5=ac8a8a208d9c26f88f702fb7634ab1abStructure, Function, and Antigenicity of the SARS-CoV-2 Spike GlycoproteinWalls, Alexandra C.; Park, Young-Jun; Tortorici, M. Alejandra; Wall, Abigail; McGuire, Andrew T.; Veesler, DavidCell (Cambridge, MA, United States) (2020), 181 (2), 281-292.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We detd. cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.
- 6Shang, J.; Ye, G.; Shi, K.; Wan, Y.; Luo, C.; Aihara, H.; Geng, Q.; Auerbach, A.; Li, F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020, 581 (7807), 221– 224, DOI: 10.1038/s41586-020-2179-yGoogle Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtbs%253D&md5=33bc9151641b2adcfb0dbf446621a1dcStructural basis of receptor recognition by SARS-CoV-2Shang, Jian; Ye, Gang; Shi, Ke; Wan, Yushun; Luo, Chuming; Aihara, Hideki; Geng, Qibin; Auerbach, Ashley; Li, FangNature (London, United Kingdom) (2020), 581 (7807), 221-224CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) recently emerged and is rapidly spreading in humans, causing COVID-191,2. A key to tackling this pandemic is to understand the receptor recognition mechanism of the virus, which regulates its infectivity, pathogenesis and host range. SARS-CoV-2 and SARS-CoV recognize the same receptor-angiotensin-converting enzyme 2 (ACE2)-in humans3,4. Here we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 (engineered to facilitate crystn.) in complex with ACE2. In comparison with the SARS-CoV RBD, an ACE2-binding ridge in SARS-CoV-2 RBD has a more compact conformation; moreover, several residue changes in the SARS-CoV-2 RBD stabilize two virus-binding hotspots at the RBD-ACE2 interface. These structural features of SARS-CoV-2 RBD increase its ACE2-binding affinity. Addnl., we show that RaTG13, a bat coronavirus that is closely related to SARS-CoV-2, also uses human ACE2 as its receptor. The differences among SARS-CoV-2, SARS-CoV and RaTG13 in ACE2 recognition shed light on the potential animal-to-human transmission of SARS-CoV-2. This study provides guidance for intervention strategies that target receptor recognition by SARS-CoV-2.
- 7St John, A. L.; Rathore, A. P. S. Early Insights into Immune Responses during COVID-19. J. Immunol. 2020, 205 (3), 555– 564, DOI: 10.4049/jimmunol.2000526Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1WgsbnK&md5=6d3b7343f9234da184e3ae1cf30e4b1eEarly insights into immune responses during COVID-19St. John, Ashley L.; Rathore, Abhay P. S.Journal of Immunology (2020), 205 (3), 555-564CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)A review. Coronavirus disease-2019 (COVID-19) is caused by the newly emerged virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and was recently declared as a pandemic by the World Health Organization. In its severe form, the disease is characterized by acute respiratory distress syndrome, and there are no targeted intervention strategies to treat or prevent it. The immune response is thought to both contribute to the pathogenesis of disease and provide protection during its resoln. Thus, understanding the immune response to SARS-CoV-2 is of the utmost importance for developing and testing vaccines and therapeutics. In this review, we discuss the earliest knowledge and hypotheses of the mechanisms of immune pathol. in the lung during acute infection as well at the later stages of disease resoln., recovery, and immune memory formation.
- 8Song, P.; Li, W.; Xie, J.; Hou, Y.; You, C. Cytokine storm induced by SARS-CoV-2. Clin. Chim. Acta 2020, 509, 280– 287, DOI: 10.1016/j.cca.2020.06.017Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlWns7%252FE&md5=cafd0380779103347d4867c3a893db38Cytokine storm induced by SARS-CoV-2Song, Peipei; Li, Wei; Xie, Jianqin; Hou, Yanlong; You, ChonggeClinica Chimica Acta (2020), 509 (), 280-287CODEN: CCATAR; ISSN:0009-8981. (Elsevier B.V.)A review. Coronavirus disease 2019 (COVID-19), caused by the virus designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread widely throughout the world. Despite the strict global outbreak management and quarantine measures that have been implemented, the incidence of COVID-19 continues to rise, resulting in more than 290,000 deaths and representing an extremely serious threat to human life and health. The clin. symptoms of the affected patients are heterogeneous, ranging from mild upper respiratory symptoms to severe pneumonitis and even acute respiratory distress syndrome (ARDS) or death. Systemic immune over activation due to SARS-CoV-2 infection causes the cytokine storm, which is esp. noteworthy in severely ill patients with COVID-19. Pieces of evidence from current studies have shown that the cytokine storm may be an important factor in disease progression, even leading to multiple organ failure and death. This review provides an overview of the knowledge on the COVID-19 epidemiol. profile, the mol. mechanisms of the SARS-CoV-2-induced cytokine storm and immune responses, the pathophysiol. changes that occur during infection, the main antiviral compds. used in treatment strategies and the potential drugs for targeting cytokines, this information is presented to provide valuable guidance for further studies and for a therapeutic redn. of this excessive immune response.
- 9Li, K.; Hao, Z.; Zhao, X.; Du, J.; Zhou, Y. SARS-CoV-2 infection-induced immune responses: Friends or foes?. Scand. J. Immunol. 2020, 92 (2), e12895 DOI: 10.1111/sji.12895Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVKms7rI&md5=a2e2c5f90a25f0a9ef9476f0d5517504SARS-CoV-2 infection-induced immune responses: Friends or foes?Li, Keying; Hao, Zhenhua; Zhao, Xiaohui; Du, Jiying; Zhou, YanlinScandinavian Journal of Immunology (2020), 92 (2), e12895CODEN: SJIMAX; ISSN:1365-3083. (Wiley-Blackwell)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging coronavirus that belongs to the β-genus, causing the outbreak of coronavirus disease 19 (COVID-19). SARS-CoV-2 infection can stimulate a pronounced immune response in the host, which embodies in the decrease of lymphocytes and aberrant increase of cytokines in COVID-19 patients. SARS-CoV-2 RNA and proteins interact with various pattern recognition receptors that switch on antiviral immune responses to regulate viral replication and spreading within the host in vivo. However, overactive and impaired immune responses also cause immune damage and subsequent tissue inflammation. This article focuses on the dual roles of immune system during SARS-CoV-2 infection, providing a theor. basic for identifying therapeutic targets in a situation with an unfavorable immune reaction.
- 10Peeples, L. News Feature: Avoiding pitfalls in the pursuit of a COVID-19 vaccine. Proc. Natl. Acad. Sci. U. S. A. 2020, 117 (15), 8218– 8221, DOI: 10.1073/pnas.2005456117Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXntFOisL4%253D&md5=1357011c0b1e556332c345ee33af1816News Feature: Avoiding pitfalls in the pursuit of a COVID-19 vaccinePeeples, LynneProceedings of the National Academy of Sciences of the United States of America (2020), 117 (15), 8218-8221CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)A review. As researchers scramble to develop a Covid-19 vaccine, they are debating which, if any, of the phenomena related to a virus infection-aiding immune "enhancement" such as antibody-dependent enhancement (ADE) or cell-based enhancement including allergic inflammation caused by Th2 immunopathol., could be at play in the case of Covid-19 and how these phenomena might affect the success of vaccine candidates. Vaccine experts have underscored the need to avoid past mistakes including the halting of SARS vaccine development. Ecol. disruption increases the odds that a future coronavirus will jump from other hosts to humans.
- 11Leslie, M. T cells found in coronavirus patients ‘bode well’ for long-term immunity. Science 2020, 368 (6493), 809– 810, DOI: 10.1126/science.368.6493.809Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVWnurjO&md5=697aa456e6b6a501bcb8d77fed0db726T cells found in coronavirus patients 'bode well' for long-term immunityLeslie, MitchScience (Washington, DC, United States) (2020), 368 (6493), 809-810CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)A review. Whether T cells join the immune system's fight against SARS-CoV-2, the virus that causes COVID-19, has been unclear. Two new studies have identified T cells that react to the virus in patients who had COVID-19 or were recovering from it. Both studies detected helper T cells, which orchestrate antiviral responses of other immune cells, and 1 of the studies also found killer T cells that eliminate virally infected cells. Both studies also discovered helper T cells that target the virus in blood samples from people who had never been infected. These people likely had the cells because they had been infected by similar coronaviruses that cause colds. The results suggest the virus induces a strong T cell response in patients, but they do not indicate whether people who have recovered from COVID-19 are immune to the virus.
- 12Manners, C.; Larios Bautista, E.; Sidoti, H.; Lopez, O. J. Protective Adaptive Immunity Against Severe Acute Respiratory Syndrome Coronaviruses 2 (SARS-CoV-2) and Implications for Vaccines. Cureus 2020, 12 (6), e8399 DOI: 10.7759/cureus.8399Google ScholarThere is no corresponding record for this reference.
- 13Mukherjee, S.; Tworowski, D.; Detroja, R.; Mukherjee, S. B.; Frenkel-Morgenstern, M. Immunoinformatics and Structural Analysis for Identification of Immunodominant Epitopes in SARS-CoV-2 as Potential Vaccine Targets. Vaccines (Basel, Switz.) 2020, 8 (2), 290, DOI: 10.3390/vaccines8020290Google ScholarThere is no corresponding record for this reference.
- 14Rock, K. L.; Goldberg, A. L. Degradation of cell proteins and the generation of MHC class I-presented peptides. Annu. Rev. Immunol. 1999, 17, 739– 79, DOI: 10.1146/annurev.immunol.17.1.739Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjtVWmtLY%253D&md5=ad0c421306a313b77d901d7c76c161d5Degradation of cell proteins and the generation of MHC class I-presented peptidesRock, Kenneth L.; Goldberg, Alfred L.Annual Review of Immunology (1999), 17 (), 739-779CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews Inc.)A review with 211 refs. Major histocompatibility complex (MHC) class I mols. display on the cell surface 8- to 10-residue peptides derived from the spectrum of proteins expressed in the cells. By screening for non-self MHC-bound peptides, the immune system identifies and then can eliminate cells that are producing viral or mutant proteins. These antigenic peptides are generated as side products in the continual turnover of intracellular proteins, which occurs primarily by the ubiquitin-proteasome pathway. Most of the oligopeptides generated by the proteasome are further degraded by distinct endopeptidases and aminopeptidases into amino acids, which are used for new protein synthesis or energy prodn. However, a fraction of these peptides escape complete destruction and after transport into the endoplasmic reticulum are bound by MHC class I mols. and delivered to the cell surface. Herein we review recent discoveries about the proteolytic systems that degrade cell proteins, how the ubiquitin-proteasome pathway generates the peptides presented on MHC-class I mols., and how this process is stimulated by immune modifiers to enhance antigen presentation.
- 15Weimershaus, M.; Evnouchidou, I.; Saveanu, L.; van Endert, P. Peptidases trimming MHC class I ligands. Curr. Opin. Immunol. 2013, 25 (1), 90– 6, DOI: 10.1016/j.coi.2012.10.001Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFCksLzL&md5=dbed24e460af7509ad6a8c9ac453de5bPeptidases trimming MHC class I ligandsWeimershaus, Mirjana; Evnouchidou, Irini; Saveanu, Loredana; van Endert, PeterCurrent Opinion in Immunology (2013), 25 (1), 90-96CODEN: COPIEL; ISSN:0952-7915. (Elsevier Ltd.)A review. Peptides presented by MHC class I mols. are typically produced through antigen degrdn. by the proteasome followed by trimming by exopeptidases. According to recent results, these include both aminopeptidases and carboxypeptidases in the cytosol and the endoplasmic reticulum. While cytosolic peptidases have a net neutral or destructive effect on MHC ligands, endoplasmic reticulum aminopeptidases are required for efficient class I loading and have a strong effect on the repertoire of peptide/MHC complexes. Cells lacking these enzymes can be eliminated both by NK cells and by CD8+ T cells recognizing complexes formed between an MHC class Ib mol. and a conserved peptide. Cross-presented peptides derived from internalized antigens can be processed by insulin-regulated aminopeptidase, the only endosomal trimming peptidase.
- 16Hammer, G. E.; Kanaseki, T.; Shastri, N. The final touches make perfect the peptide-MHC class I repertoire. Immunity 2007, 26 (4), 397– 406, DOI: 10.1016/j.immuni.2007.04.003Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXltVKjsrw%253D&md5=d50e9482fd0fa138cfe165615d1329d9The final touches make perfect the peptide-MHC class I repertoireHammer, Gianna Elena; Kanaseki, Takayuki; Shastri, NilabhImmunity (2007), 26 (4), 397-406CODEN: IUNIEH; ISSN:1074-7613. (Cell Press)A review. Major histocompatibility complex (MHC) class I mols. present short, perfectly cleaved peptides on the cell surface for immune surveillance by CD8+ T cells. The pathway for generating these peptides begins in the cytoplasm, and the peptide-MHC I (pMHC I) repertoire is finalized in the endoplasmic reticulum. Recent studies show that the peptides for MHC I are customized by the ER aminopeptidase assocd. with antigen processing and by dynamic interactions within the MHC peptide-loading complex. Failure to customize the pMHC I repertoire has profound immunol. consequences.
- 17Giastas, P.; Mpakali, A.; Papakyriakou, A.; Lelis, A.; Kokkala, P.; Neu, M.; Rowland, P.; Liddle, J.; Georgiadis, D.; Stratikos, E. Mechanism for antigenic peptide selection by endoplasmic reticulum aminopeptidase 1. Proc. Natl. Acad. Sci. U. S. A. 2019, 116 (52), 26709– 26716, DOI: 10.1073/pnas.1912070116Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjsFCntbw%253D&md5=3115e900c52db599cf264d7c6e0762d8Mechanism for antigenic peptide selection by endoplasmic reticulum aminopeptidase 1Giastas, Petros; Mpakali, Anastasia; Papakyriakou, Athanasios; Lelis, Aggelos; Kokkala, Paraskevi; Neu, Margarete; Rowland, Paul; Liddle, John; Georgiadis, Dimitris; Stratikos, EfstratiosProceedings of the National Academy of Sciences of the United States of America (2019), 116 (52), 26709-26716CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an intracellular enzyme that optimizes the peptide cargo of major histocompatibility class I (MHC-I) mols. and regulates adaptive immunity. It has unusual substrate selectivity for length and sequence, resulting in poorly understood effects on the cellular immunopeptidome. To understand substrate selection by ERAP1, we solved 2 crystal structures of the enzyme with bound transition-state pseudopeptide analogs at 1.68 Å and 1.72 Å. Both peptides have their N terminus bound at the active site and extend away along a large internal cavity, interacting with shallow pockets that can influence selectivity. The longer peptide is disordered through the central region of the cavity and has its C terminus bound in an allosteric pocket of domain IV that features a carboxypeptidase-like structural motif. These structures, along with enzymic and computational analyses, explain how ERAP1 can select peptides based on length while retaining the broad sequence-specificity necessary for its biol. function.
- 18Stratikos, E.; Stern, L. J. Antigenic peptide trimming by ER aminopeptidases--insights from structural studies. Mol. Immunol. 2013, 55 (3–4), 212– 9, DOI: 10.1016/j.molimm.2013.03.002Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXltVent7k%253D&md5=b6318876699d28ee425936e83c462031Antigenic peptide trimming by ER aminopeptidases - Insights from structural studiesStratikos, Efstratios; Stern, Lawrence J.Molecular Immunology (2013), 55 (3-4), 212-219CODEN: MOIMD5; ISSN:0161-5890. (Elsevier)A review. Generation and destruction of antigenic peptides by ER resident aminopeptidases ERAP1 and ERAP2 have been shown in the last few years to be important for the correct functioning and regulation of the adaptive immune response. These two highly homologous aminopeptidases appear to have evolved complex mechanisms well suited for their biol. role in antigen presentation. Furthermore, polymorphic variability in these enzymes appears to affect their function and predispose individuals to disease. This review discusses our current understanding of the mol. mechanisms behind ERAP1/2 function as suggested by several recently detd. crystallog. structures of these enzymes.
- 19Evnouchidou, I.; Kamal, R. P.; Seregin, S. S.; Goto, Y.; Tsujimoto, M.; Hattori, A.; Voulgari, P. V.; Drosos, A. A.; Amalfitano, A.; York, I. A.; Stratikos, E. Coding single nucleotide polymorphisms of endoplasmic reticulum aminopeptidase 1 can affect antigenic peptide generation in vitro by influencing basic enzymatic properties of the enzyme. J. Immunol. 2011, 186 (4), 1909– 13, DOI: 10.4049/jimmunol.1003337Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVShurg%253D&md5=10e432675dcf5b15636f8f7de5c1a6c5Cutting Edge: Coding Single Nucleotide Polymorphisms of Endoplasmic Reticulum Aminopeptidase 1 Can Affect Antigenic Peptide Generation In Vitro by Influencing Basic Enzymatic Properties of the EnzymeEvnouchidou, Irini; Kamal, Ram P.; Seregin, Sergey S.; Goto, Yoshikuni; Tsujimoto, Masafumi; Hattori, Akira; Voulgari, Paraskevi V.; Drosos, Alexandros A.; Amalfitano, Andrea; York, Ian A.; Stratikos, EfstratiosJournal of Immunology (2011), 186 (4), 1909-1913CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)ER aminopeptidase 1 (ERAP1) customizes antigenic peptide precursors for MHC class I presentation and edits the antigenic peptide repertoire. Coding single nucleotide polymorphisms (SNPs) in ERAP1 were recently linked with predisposition to autoimmune disease, suggesting a link between pathogenesis of autoimmunity and ERAP1-mediated Ag processing. To investigate this possibility, the authors analyzed the effect that disease-linked SNPs have on Ag processing by ERAP1 in vitro. Michaelis-Menten anal. revealed that the presence of SNPs affects the Michaelis const. and turnover no. of the enzyme. Strikingly, specific ERAP1 allele-substrate combinations deviate from std. Michaelis-Menten behavior, demonstrating substrate-inhibition kinetics; to the authors' knowledge, this phenomenon has not been described for this enzyme. Cell-based Ag-presentation anal. was consistent with changes in the substrate inhibition const. Ki, further supporting that ERAP1 allelic compn. may affect Ag processing in vivo. The authors propose that these phenomena should be taken into account when evaluating the possible link between Ag processing and autoimmunity.
- 20Stamogiannos, A.; Maben, Z.; Papakyriakou, A.; Mpakali, A.; Kokkala, P.; Georgiadis, D.; Stern, L. J.; Stratikos, E. Critical Role of Interdomain Interactions in the Conformational Change and Catalytic Mechanism of Endoplasmic Reticulum Aminopeptidase 1. Biochemistry 2017, 56 (10), 1546– 1558, DOI: 10.1021/acs.biochem.6b01170Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXivVOisrs%253D&md5=5a76edc1020984d4ce7348d80dbd8514Critical Role of Interdomain Interactions in the Conformational Change and Catalytic Mechanism of Endoplasmic Reticulum Aminopeptidase 1Stamogiannos, Athanasios; Maben, Zachary; Papakyriakou, Athanasios; Mpakali, Anastasia; Kokkala, Paraskevi; Georgiadis, Dimitris; Stern, Lawrence J.; Stratikos, EfstratiosBiochemistry (2017), 56 (10), 1546-1558CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) is an intracellular enzyme important for the generation of antigenic epitopes and class I MHC-restricted adaptive immune responses. ERAP1 processes a vast variety of different peptides but still shows length and sequence selectivity although the mechanism behind these properties is poorly understood. X-ray crystallog. anal. has revealed that ERAP1 can assume at least two distinct conformations in which the C-terminal domain IV is either proximal or distal to the active site domain II. To help understand the role of this conformational change in the catalytic mechanism of ERAP1 we used site-directed mutagenesis to perturb key salt-bridges between domains II and IV. Enzymic anal. revealed that these mutations, although located away from the catalytic site, greatly reduce catalytic efficiency and shift allosteric kinetic behavior. The variants were more efficiently activated by small peptides, and bound a competitive inhibitor with weaker affinity and faster dissocn. kinetics. Mol. dynamics anal. suggested that the mutations affect the conformational distribution of ERAP1, reducing the population of closed states. Small-angle x-ray scattering indicated that both wild-type and the ERAP1 variants are predominantly in an open conformational state in soln. Overall, our findings suggest that electrostatic interactions between domains II and IV in ERAP1 are crucial for driving a conformational change that regulates the structural integrity of the catalytic site. The extent of domain opening in ERAP1 probably underlies its specialization for antigenic peptide precursors and should be taken into account for inhibitor development efforts.
- 21Mpakali, A.; Giastas, P.; Mathioudakis, N.; Mavridis, I. M.; Saridakis, E.; Stratikos, E. Structural Basis for Antigenic Peptide Recognition and Processing by Endoplasmic Reticulum (ER) Aminopeptidase 2. J. Biol. Chem. 2015, 290 (43), 26021– 32, DOI: 10.1074/jbc.M115.685909Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvF2qtLbL&md5=04d68ad1491150a69457cc1eda48d498Structural basis for antigenic peptide recognition and processing by endoplasmic reticulum (ER) aminopeptidase 2Mpakali, Anastasia; Giastas, Petros; Mathioudakis, Nikolas; Mavridis, Irene M.; Saridakis, Emmanuel; Stratikos, EfstratiosJournal of Biological Chemistry (2015), 290 (43), 26021-26032CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Endoplasmic reticulum (ER) aminopeptidases process antigenic peptide precursors to generate epitopes for presentation by MHC class I mols. and help shape the antigenic peptide repertoire and cytotoxic T-cell responses. To perform this function, ER aminopeptidases have to recognize and process a vast variety of peptide sequences. To understand how these enzymes recognize substrates, we detd. crystal structures of ER aminopeptidase 2 (ERAP2) in complex with a substrate analog and a peptidic product to 2.5 and 2.7 Å, resp., and compared them to the apo-form structure detd. to 3.0 Å. The peptides were found within the internal cavity of the enzyme with no direct access to the outside solvent. The substrate analog extends away from the catalytic center toward the distal end of the internal cavity, making interactions with several shallow pockets along the path. A similar configuration was evident for the peptidic product, although decreasing electron d. toward its C terminus indicated progressive disorder. Enzymic anal. confirmed that visualized interactions can either pos. or neg. impact in vitro trimming rates. Opportunistic side-chain interactions and lack of deep specificity pockets support a limited-selectivity model for antigenic peptide processing by ERAP2. In contrast to proposed models for the homologous ERAP1, no specific recognition of the peptide C terminus by ERAP2 was evident, consistent with functional differences in length selection and self-activation between these two enzymes. Our results suggest that ERAP2 selects substrates by sequestering them in its internal cavity and allowing opportunistic interactions to det. trimming rates, thus combining substrate permissiveness with sequence bias.
- 22Mpakali, A.; Saridakis, E.; Harlos, K.; Zhao, Y.; Papakyriakou, A.; Kokkala, P.; Georgiadis, D.; Stratikos, E. Crystal Structure of Insulin-Regulated Aminopeptidase with Bound Substrate Analogue Provides Insight on Antigenic Epitope Precursor Recognition and Processing. J. Immunol. 2015, 195 (6), 2842– 2851, DOI: 10.4049/jimmunol.1501103Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVygsL7F&md5=1738ab2c66078883c93c26cc7da6c2ffCrystal Structure of Insulin-Regulated Aminopeptidase with Bound Substrate Analogue Provides Insight on Antigenic Epitope Precursor Recognition and ProcessingMpakali, Anastasia; Saridakis, Emmanuel; Harlos, Karl; Zhao, Yuguang; Papakyriakou, Athanasios; Kokkala, Paraskevi; Georgiadis, Dimitris; Stratikos, EfstratiosJournal of Immunology (2015), 195 (6), 2842-2851CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)Aminopeptidases that generate antigenic peptides influence immunodominance and adaptive cytotoxic immune responses. The mechanisms that allow these enzymes to efficiently process a vast no. of different long peptide substrates are poorly understood. In this work, we report the structure of insulin-regulated aminopeptidase, an enzyme that preps. antigenic epitopes for cross-presentation in dendritic cells, in complex with an antigenic peptide precursor analog. Insulin-regulated aminopeptidase is found in a semiclosed conformation with an extended internal cavity with limited access to the solvent. The N-terminal moiety of the peptide is located at the active site, positioned optimally for catalysis, whereas the C-terminal moiety of the peptide is stabilized along the extended internal cavity lodged between domains II and IV. Hydrophobic interactions and shape complementarity enhance peptide affinity beyond the catalytic site and support a limited selectivity model for antigenic peptide selection that may underlie the generation of complex immunopeptidomes.
- 23Saveanu, L.; Carroll, O.; Lindo, V.; Del Val, M.; Lopez, D.; Lepelletier, Y.; Greer, F.; Schomburg, L.; Fruci, D.; Niedermann, G.; van Endert, P. M. Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulum. Nat. Immunol. 2005, 6 (7), 689– 97, DOI: 10.1038/ni1208Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXltlynsro%253D&md5=bc2f4593600e4947b5e61d7c94980e93Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulumSaveanu, Loredana; Carroll, Oliver; Lindo, Vivian; Del Val, Margarita; Lopez, Daniel; Lepelletier, Yves; Greer, Fiona; Schomburg, Lutz; Fruci, Doriana; Niedermann, Gabriele; van Endert, Peter M.Nature Immunology (2005), 6 (7), 689-697CODEN: NIAMCZ; ISSN:1529-2908. (Nature Publishing Group)The generation of many HLA class I peptides entails a final trimming step in the endoplasmic reticulum that, in humans, is accomplished by two 'candidate' aminopeptidases. The authors show here that one of these, ERAP1, was unable to remove several N-terminal amino acids that were trimmed efficiently by the second enzyme, ERAP2. This trimming of a longer peptide required the concerted action of both ERAP1 and ERAP2, both for in vitro digestion and in vivo for cellular antigen presentation. ERAP1 and ERAP2 localized together in vivo and assocd. phys. in complexes that were most likely heterodimeric. Thus, the human endoplasmic reticulum is equipped with a pair of trimming aminopeptidases that have complementary functions in HLA class I peptide presentation.
- 24Evnouchidou, I.; Weimershaus, M.; Saveanu, L.; van Endert, P. ERAP1-ERAP2 dimerization increases peptide-trimming efficiency. J. Immunol. 2014, 193 (2), 901– 8, DOI: 10.4049/jimmunol.1302855Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFSmtrbO&md5=9e7e734031550d119889b8c7bfc7bc78ERAP1-ERAP2 Dimerization Increases Peptide-Trimming EfficiencyEvnouchidou, Irini; Weimershaus, Mirjana; Saveanu, Loredana; van Endert, PeterJournal of Immunology (2014), 193 (2), 901-908CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)The endoplasmic reticulum aminopeptidases (ERAP)1 and ERAP2 play a crit. role in the prodn. of final epitopes presented by MHC class I mols. Formation of heterodimers by ERAP1 and ERAP2 has been proposed to facilitate trimming of epitope precursor peptides, but the effects of dimerization on ERAP function remain unknown. In this study, we produced stabilized ERAP1-ERAP2 heterodimers and found that they produced several mature epitopes more efficiently than a mix of the two enzymes unable to dimerize. Phys. interaction with ERAP2 changes basic enzymic parameters of ERAP1 and improves its substrate-binding affinity. Thus, by bringing the two enzymes in proximity and by producing allosteric effects on ERAP1, dimerization of ERAP1/2 creates complexes with superior peptide-trimming efficacy. Such complexes are likely to enhance Ag presentation by cells displaying coordinated expression of the two enzymes.
- 25Zervoudi, E.; Papakyriakou, A.; Georgiadou, D.; Evnouchidou, I.; Gajda, A.; Poreba, M.; Salvesen, G. S.; Drag, M.; Hattori, A.; Swevers, L.; Vourloumis, D.; Stratikos, E. Probing the S1 specificity pocket of the aminopeptidases that generate antigenic peptides. Biochem. J. 2011, 435 (2), 411– 20, DOI: 10.1042/BJ20102049Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXltF2gu7s%253D&md5=8c9e81b2348656d5339b27724789066aProbing the S1 specificity pocket of the aminopeptidases that generate antigenic peptidesZervoudi, Efthalia; Papakyriakou, Athanasios; Georgiadou, Dimitra; Evnouchidou, Irini; Gajda, Anna; Poreba, Marcin; Salvesen, Guy S.; Drag, Marcin; Hattori, Akira; Swevers, Luc; Vourloumis, Dionisios; Stratikos, EfstratiosBiochemical Journal (2011), 435 (2), 411-420CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)ERAP1 (endoplasmic reticulum aminopeptidase 1), ERAP2 and IRAP (insulin-regulated aminopeptidase) are three homologous enzymes that play crit. roles in the generation of antigenic peptides. These aminopeptidases excise amino acids from N-terminally extended precursors of antigenic peptides in order to generate the correct length epitopes for binding on to MHC class I mols. The specificity of these peptidases can affect antigenic peptide selection, but has not yet been investigated in detail. In the present study we utilized a collection of 82 fluorogenic substrates to define a detailed selectivity profile for each of the three enzymes and to probe structural and functional features of the S1 (primary specificity) pocket. Mol. modeling of the three S1 pockets reveals substrate-enzyme interactions that are crit. determinants for specificity. The substrate selectivity profiles suggest that IRAP largely combines the S1 specificity of ERAP1 and ERAP2, consistent with its proposed biol. function. IRAP, however, does not achieve this dual specificity by simply combining structural features of ERAP1 and ERAP2, but rather by an unique amino acid change at position 541. The results of the present study provide insights on antigenic peptide selection and may prove valuable in designing selective inhibitors or activity markers for this class of enzymes.
- 26Chang, S. C.; Momburg, F.; Bhutani, N.; Goldberg, A. L. The ER aminopeptidase, ERAP1, trims precursors to lengths of MHC class I peptides by a “molecular ruler” mechanism. Proc. Natl. Acad. Sci. U. S. A. 2005, 102 (47), 17107– 12, DOI: 10.1073/pnas.0500721102Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1yksLbN&md5=d7057c479987e375cd70e463c802e78bThe ER aminopeptidase, ERAP1, trims precursors to lengths of MHC class I peptides by a "molecular ruler" mechanismChang, Shih-Chung; Momburg, Frank; Bhutani, Nidhi; Goldberg, Alfred L.Proceedings of the National Academy of Sciences of the United States of America (2005), 102 (47), 17107-17112CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an IFN-γ-induced aminopeptidase in the endoplasmic reticulum that trims longer precursors to the antigenic peptides presented on MHC class I mols. The authors recently reported that purified ERAP1 trimmed N-extended precursors but spared peptides of 8-9 residues, the length required for binding to MHC class I mols. Here, the authors show another remarkable property of ERAP1: that it strongly prefers substrates 9-16 residues long, the lengths of peptides transported efficiently into the ER by the transporter assocd. with antigen processing (TAP) transporter. This aminopeptidase rapidly degraded a model 13-mer to a 9-mer and then stopped, even though the substrate and the product had identical N- and C-terminal sequences. No other aminopeptidase, including the closely related ER-aminopeptidase ERAP2, showed a similar length preference. Unlike other aminopeptidases, the activity of ERAP1 depended on the C-terminal residue of the substrate. ERAP1, like most MHC class I mols., prefers peptides with hydrophobic C termini and shows low affinity for peptides with charged C termini. Thus, ERAP1 is specialized to process precursors transported by TAP to peptides that can serve as MHC class I epitopes. Its "mol. ruler" mechanism involves binding the hydrophobic C terminus of the substrate 9-16 residues away from the active site.
- 27Lorente, E.; Barriga, A.; Johnstone, C.; Mir, C.; Jimenez, M.; Lopez, D. Concerted in vitro trimming of viral HLA-B27-restricted ligands by human ERAP1 and ERAP2 aminopeptidases. PLoS One 2013, 8 (11), e79596 DOI: 10.1371/journal.pone.0079596Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslClurnJ&md5=e6bb141f3177ecbdf63412df9a9f8e87Concerted in vitro trimming of viral HLA-B27-restricted ligands by human ERAP1 and ERAP2 aminopeptidasesLorente, Elena; Barriga, Alejandro; Johnstone, Carolina; Mir, Carmen; Jimenez, Mercedes; Lopez, DanielPLoS One (2013), 8 (11), e79596CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)In the classical human leukocyte antigen (HLA) class I antigen processing and presentation pathway, the antigenic peptides are generated from viral proteins by multiple proteolytic cleavages of the proteasome (and in some cases other cytosolic proteases) and transported to the endoplasmic reticulum (ER) lumen where they are exposed to aminopeptidase activity. In human cells, two different ER-resident enzymes, ERAP1 and ERAP2, can trim the N-terminally extended residues of peptide precursors. In this study, the possible cooperative effect of generating five naturally processed HLA-B27 ligands by both proteases was analyzed. We identified differences in the products obtained with increased detection of natural HLA-B27 ligands by comparing double vs. single enzyme digestions by mass spectrometry anal. These in vitro data suggest that each enzyme can use the degrdn. products of the other as a substrate for new N-terminal trimming, indicating concerted aminoproteolytic activity of ERAP1 and ERAP2.
- 28Hulsen, T.; de Vlieg, J.; Alkema, W. BioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics 2008, 9, 488, DOI: 10.1186/1471-2164-9-488Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cjksF2ltQ%253D%253D&md5=1c79e2fa6accc5ff3c4cd77bab9aca8cBioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagramsHulsen Tim; de Vlieg Jacob; Alkema WynandBMC genomics (2008), 9 (), 488 ISSN:.BACKGROUND: In many genomics projects, numerous lists containing biological identifiers are produced. Often it is useful to see the overlap between different lists, enabling researchers to quickly observe similarities and differences between the data sets they are analyzing. One of the most popular methods to visualize the overlap and differences between data sets is the Venn diagram: a diagram consisting of two or more circles in which each circle corresponds to a data set, and the overlap between the circles corresponds to the overlap between the data sets. Venn diagrams are especially useful when they are 'area-proportional' i.e. the sizes of the circles and the overlaps correspond to the sizes of the data sets. Currently there are no programs available that can create area-proportional Venn diagrams connected to a wide range of biological databases. RESULTS: We designed a web application named BioVenn to summarize the overlap between two or three lists of identifiers, using area-proportional Venn diagrams. The user only needs to input these lists of identifiers in the textboxes and push the submit button. Parameters like colors and text size can be adjusted easily through the web interface. The position of the text can be adjusted by 'drag-and-drop' principle. The output Venn diagram can be shown as an SVG or PNG image embedded in the web application, or as a standalone SVG or PNG image. The latter option is useful for batch queries. Besides the Venn diagram, BioVenn outputs lists of identifiers for each of the resulting subsets. If an identifier is recognized as belonging to one of the supported biological databases, the output is linked to that database. Finally, BioVenn can map Affymetrix and EntrezGene identifiers to Ensembl genes. CONCLUSION: BioVenn is an easy-to-use web application to generate area-proportional Venn diagrams from lists of biological identifiers. It supports a wide range of identifiers from the most used biological databases currently available. Its implementation on the World Wide Web makes it available for use on any computer with internet connection, independent of operating system and without the need to install programs locally. BioVenn is freely accessible at http://www.cmbi.ru.nl/cdd/biovenn/.
- 29Sarkizova, S.; Klaeger, S.; Le, P. M.; Li, L. W.; Oliveira, G.; Keshishian, H.; Hartigan, C. R.; Zhang, W.; Braun, D. A.; Ligon, K. L.; Bachireddy, P.; Zervantonakis, I. K.; Rosenbluth, J. M.; Ouspenskaia, T.; Law, T.; Justesen, S.; Stevens, J.; Lane, W. J.; Eisenhaure, T.; Lan Zhang, G.; Clauser, K. R.; Hacohen, N.; Carr, S. A.; Wu, C. J.; Keskin, D. B. A large peptidome dataset improves HLA class I epitope prediction across most of the human population. Nat. Biotechnol. 2020, 38 (2), 199– 209, DOI: 10.1038/s41587-019-0322-9Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVSktbvP&md5=e24b305412a6cfed38e5bcd7decf6359A large peptidome dataset improves HLA class I epitope prediction across most of the human populationSarkizova, Siranush; Klaeger, Susan; Le, Phuong M.; Li, Letitia W.; Oliveira, Giacomo; Keshishian, Hasmik; Hartigan, Christina R.; Zhang, Wandi; Braun, David A.; Ligon, Keith L.; Bachireddy, Pavan; Zervantonakis, Ioannis K.; Rosenbluth, Jennifer M.; Ouspenskaia, Tamara; Law, Travis; Justesen, Sune; Stevens, Jonathan; Lane, William J.; Eisenhaure, Thomas; Lan Zhang, Guang; Clauser, Karl R.; Hacohen, Nir; Carr, Steven A.; Wu, Catherine J.; Keskin, Derin B.Nature Biotechnology (2020), 38 (2), 199-209CODEN: NABIF9; ISSN:1087-0156. (Nature Research)Prediction of HLA epitopes is important for the development of cancer immunotherapies and vaccines. However, current prediction algorithms have limited predictive power, in part because they were not trained on high-quality epitope datasets covering a broad range of HLA alleles. To enable prediction of endogenous HLA class I-assocd. peptides across a large fraction of the human population, we used mass spectrometry to profile >185,000 peptides eluted from 95 HLA-A, -B, -C and -G mono-allelic cell lines. We identified canonical peptide motifs per HLA allele, unique and shared binding submotifs across alleles and distinct motifs assocd. with different peptide lengths. By integrating these data with transcript abundance and peptide processing, we developed HLAthena, providing allele-and-length-specific and pan-allele-pan-length prediction models for endogenous peptide presentation. These models predicted endogenous HLA class I-assocd. ligands with 1.5-fold improvement in pos. predictive value compared with existing tools and correctly identified >75% of HLA-bound peptides that were obsd. exptl. in 11 patient-derived tumor cell lines.
- 30Vita, R.; Zarebski, L.; Greenbaum, J. A.; Emami, H.; Hoof, I.; Salimi, N.; Damle, R.; Sette, A.; Peters, B. The immune epitope database 2.0. Nucleic Acids Res. 2010, 38 (Database issue), D854– D862, DOI: 10.1093/nar/gkp1004Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXktl2isg%253D%253D&md5=a8502a555f1a46955251c2819097f099The Immune Epitope Database 2.0Vita, Randi; Zarebski, Laura; Greenbaum, Jason A.; Emami, Hussein; Hoof, Ilka; Salimi, Nima; Damle, Rohini; Sette, Alessandro; Peters, BjoernNucleic Acids Research (2010), 38 (Database Iss), D854-D862CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Immune Epitope Database (IEDB, www.iedb.org) provides a catalog of exptl. characterized B and T cell epitopes, as well as data on Major Histocompatibility Complex (MHC) binding and MHC ligand elution expts. The database represents the mol. structures recognized by adaptive immune receptors and the exptl. contexts in which these mols. were detd. to be immune epitopes. Epitopes recognized in humans, nonhuman primates, rodents, pigs, cats and all other tested species are included. Both pos. and neg. exptl. results are captured. Over the course of 4 years, the data from 180 978 expts. were curated manually from the literature, which covers ∼99% of all publicly available information on peptide epitopes mapped in infectious agents (excluding HIV) and 93% of those mapped in allergens. In addn., data that would otherwise be unavailable to the public from 129 186 expts. were submitted directly by investigators. The curation of epitopes related to autoimmunity is expected to be completed by the end of 2010. The database can be queried by epitope structure, source organism, MHC restriction, assay type or host organism, among other criteria. The database structure, as well as its querying, browsing and reporting interfaces, was completely redesigned for the IEDB 2.0 release, which became publicly available in early 2009.
- 31Nguyen, A.; David, J. K.; Maden, S. K.; Wood, M. A.; Weeder, B. R.; Nellore, A.; Thompson, R. F. Human leukocyte antigen susceptibility map for SARS-CoV-2. J. Virol. 2020, DOI: 10.1128/JVI.00510-20Google ScholarThere is no corresponding record for this reference.
- 32Reynisson, B.; Alvarez, B.; Paul, S.; Peters, B.; Nielsen, M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 2020, 48 (W1), W449– W454, DOI: 10.1093/nar/gkaa379Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXis1entr7E&md5=c777c9c3cb0858e4bef24fe6b9358244NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand dataReynisson, Birkir; Alvarez, Bruno; Paul, Sinu; Peters, Bjoern; Nielsen, MortenNucleic Acids Research (2020), 48 (W1), W449-W454CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Major histocompatibility complex (MHC) mols. are expressed on the cell surface, where they present peptides to T cells, which gives them a key role in the development of T-cell immune responses. MHC mols. come in two main variants: MHC Class I (MHC-I) and MHC Class II (MHC-II). MHC-I predominantly present peptides derived from intracellular proteins, whereas MHC-II predominantly presents peptides from extracellular proteins. In both cases, the binding between MHC and antigenic peptides is the most selective step in the antigen presentation pathway. Therefore, the prediction of peptide binding to MHC is a powerful utility to predict the possible specificity of a T-cell immune response. Commonly MHC binding prediction tools are trained on binding affinity or mass spectrometry-eluted ligands. Recent studies have however demonstrated how the integration of both data types can boost predictive performances. Inspired by this, we here present NetMHCpan-4.1 and NetMHCIIpan-4.0, two web servers created to predict binding between peptides and MHC-I and MHC-II, resp. Both methods exploit tailored machine learning strategies to integrate different training data types, resulting in state-of-the-art performance and outperforming their competitors.
- 33Komov, L.; Kadosh, D. M.; Barnea, E.; Milner, E.; Hendler, A.; Admon, A. Cell Surface MHC Class I Expression Is Limited by the Availability of Peptide-Receptive “Empty” Molecules Rather than by the Supply of Peptide Ligands. Proteomics 2018, 18 (12), e1700248 DOI: 10.1002/pmic.201700248Google ScholarThere is no corresponding record for this reference.
- 34Hearn, A.; York, I. A.; Rock, K. L. The specificity of trimming of MHC class I-presented peptides in the endoplasmic reticulum. J. Immunol. 2009, 183 (9), 5526– 36, DOI: 10.4049/jimmunol.0803663Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1ymtb%252FK&md5=6eddb77faa4a92adbb899fd26d842406The Specificity of Trimming of MHC Class I-Presented Peptides in the Endoplasmic ReticulumHearn, Arron; York, Ian A.; Rock, Kenneth L.Journal of Immunology (2009), 183 (9), 5526-5536CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)Aminopeptidases in the endoplasmic reticulum (ER) can cleave antigenic peptides and in so doing either create or destroy MHC class I-presented epitopes. However, the specificity of this trimming process overall and of the major ER aminopeptidase ERAP1 in particular is not well understood. This issue is important because peptide trimming influences the magnitude and specificity of CD8 T cell responses. By systematically varying the N-terminal flanking sequences of peptides in a cell-free biochem. system and in intact cells, the authors elucidated the specificity of ERAP1 and of ER trimming overall. ERAP1 can cleave after many amino acids on the N terminus of epitope precursors but does so at markedly different rates. The specificity seen with purified ERAP1 is similar to that obsd. for trimming and presentation of epitopes in the ER of intact cells. The authors define N-terminal sequences that are favorable or unfavorable for Ag presentation in ways that are independent from the epitopes core sequence. When databases of known presented peptides were analyzed, the residues that were preferred for the trimming of model peptide precursors were overrepresented in N-terminal flanking sequences of epitopes generally. These data define key determinants in the specificity of Ag processing.
- 35Georgiadou, D.; Hearn, A.; Evnouchidou, I.; Chroni, A.; Leondiadis, L.; York, I. A.; Rock, K. L.; Stratikos, E. Placental leucine aminopeptidase efficiently generates mature antigenic peptides in vitro but in patterns distinct from endoplasmic reticulum aminopeptidase 1. J. Immunol. 2010, 185 (3), 1584– 92, DOI: 10.4049/jimmunol.0902502Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptV2jtr8%253D&md5=0ec50b33d7b7752368d4ed9da7f13c81Placental Leucine Aminopeptidase Efficiently Generates Mature Antigenic Peptides In Vitro but in Patterns Distinct from Endoplasmic Reticulum Aminopeptidase 1Georgiadou, Dimitra; Hearn, Arron; Evnouchidou, Irini; Chroni, Angeliki; Leondiadis, Leondios; York, Ian A.; Rock, Kenneth L.; Stratikos, EfstratiosJournal of Immunology (2010), 185 (3), 1584-1592CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)All three members of the oxytocinase subfamily of M1 aminopeptidases, endoplasmic reticulum aminopeptidase 1 (ERAP1), ERAP2, and placental leucine aminopeptidase (PLAP), also known as insulin-regulated aminopeptidase, have been implicated in the generation of MHC class I-presented peptides. ERAP1 and 2 trim peptides in the endoplasmic reticulum for direct presentation, whereas PLAP has been recently implicated in cross-presentation. The best characterized member of the family, ERAP1, has unique enzymic properties that fit well with its role in Ag processing. ERAP1 can trim a large variety of long peptide sequences and efficiently accumulate mature antigenic epitopes of 8-9 aa long. In this study, the authors evaluate the ability of PLAP to process antigenic peptide precursors in vitro and compare it with ERAP1. The authors find that, similar to ERAP1, PLAP can trim a variety of long peptide sequences efficiently and, in most cases, accumulates appreciable amts. of correct length mature antigenic epitope. Again, similar to ERAP1, PLAP continued trimming some of the epitopes tested and accumulated smaller products effectively destroying the epitope. However, the intermediate accumulation properties of ERAP1 and PLAP are distinct and epitope dependent, suggesting that these two enzymes may impose different selective pressures on epitope generation. Overall, although PLAP has the necessary enzymic properties to participate in generating or destroying MHC class I-presented peptides, its trimming behavior is distinct from that of ERAP1, something that supports a sep. role for these two enzymes in Ag processing.
- 36Kuiper, J. J.; Van Setten, J.; Ripke, S.; Van, T. S. R.; Mulder, F.; Missotten, T.; Baarsma, G. S.; Francioli, L. C.; Pulit, S. L.; De Kovel, C. G.; Ten Dam-Van Loon, N.; Den Hollander, A. I.; Huis in het Veld, P.; Hoyng, C. B.; Cordero-Coma, M.; Martin, J.; Llorenc, V.; Arya, B.; Thomas, D.; Bakker, S. C.; Ophoff, R. A.; Rothova, A.; De Bakker, P. I.; Mutis, T.; Koeleman, B. P. A genome-wide association study identifies a functional ERAP2 haplotype associated with birdshot chorioretinopathy. Hum. Mol. Genet. 2014, 23 (22), 6081– 6087, DOI: 10.1093/hmg/ddu307Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFOrtLbI&md5=896d45e2605159f4036e0cb4e3481484A genome-wide association study identifies a functional ERAP2 haplotype associated with birdshot chorioretinopathyKuiper, Jonas J. W.; Van Setten, Jessica; Ripke, Stephan; Slot, Ruben Van 'T.; Mulder, Flip; Missotten, Tom; Baarsma, G. Seerp; Francioli, Laurent C.; Pulit, Sara L.; Kovel, Carolien G. F. De; Loon, Ninette Ten Dam-Van; Hollander, Anneke I. Den; Veld, Paulien Huis in het; Hoyng, Carel B.; Cordero-Coma, Miguel; Martin, Javier; Llorenc, Victor; Arya, Bharti; Thomas, Dhanes; Bakker, Steven C.; Ophoff, Roel A.; Rothova, Aniki; Bakker, Paul I. W. De; Mutis, Tuna; Koeleman, Bobby P. C.Human Molecular Genetics (2014), 23 (22), 6081-6087CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)Birdshot chorioretinopathy (BSCR) is a rare form of autoimmune uveitis that can lead to severe visual impairment. Intriguingly, >95% of cases carry the HLA-A29 allele, which defines the strongest documented HLA assocn. for a human disease. We have conducted a genome-wide assocn. study in 96 Dutch and 27 Spanish cases, and 398 unrelated Dutch and 380 Spanish controls. Fine-mapping the primary MHC assocn. through high-resoln. imputation at classical HLA loci, identified HLA-A*29:02 as the principal MHC assocn. (odds ratio (OR) = 157.5, 95% CI 91.6-272.6, P = 6.6 × 10-74). We also identified two novel susceptibility loci at 5q15 near ERAP2 (rs7705093;OR = 2.3,95%CI 1.7-3.1, for the T allele, P = 8.6 × 10-8) and at 14q32.31 in the TECPR2 gene (rs150571175;OR = 6.1,95%CI 3.2-11.7, for the A allele, P = 3.2 × 10-8). The assocn. near ERAP2 was confirmed in an independent British case-control samples (combined meta-anal. P = 1.7 × 10-9). Functional analyses revealed that the risk allele of the polymorphism near ERAP2 is strongly assocd. with high mRNA and protein expression of ERAP2 in B cells. This study further defined an extremely strong MHC risk component in BSCR, and detected evidence for a novel disease mechanism that affects peptide processing in the endoplasmic reticulum.
- 37Grifoni, A.; Weiskopf, D.; Ramirez, S. I.; Mateus, J.; Dan, J. M.; Moderbacher, C. R.; Rawlings, S. A.; Sutherland, A.; Premkumar, L.; Jadi, R. S.; Marrama, D.; de Silva, A. M.; Frazier, A.; Carlin, A. F.; Greenbaum, J. A.; Peters, B.; Krammer, F.; Smith, D. M.; Crotty, S.; Sette, A. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181 (7), 1489– 1501, DOI: 10.1016/j.cell.2020.05.015Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVOmu73N&md5=6f89bf52ae2d734af758163a4940b356Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed IndividualsGrifoni, Alba; Weiskopf, Daniela; Ramirez, Sydney I.; Mateus, Jose; Dan, Jennifer M.; Moderbacher, Carolyn Rydyznski; Rawlings, Stephen A.; Sutherland, Aaron; Premkumar, Lakshmanane; Jadi, Ramesh S.; Marrama, Daniel; de Silva, Aravinda M.; Frazier, April; Carlin, Aaron F.; Greenbaum, Jason A.; Peters, Bjoern; Krammer, Florian; Smith, Davey M.; Crotty, Shane; Sette, AlessandroCell (Cambridge, MA, United States) (2020), 181 (7), 1489-1501.e15CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide "megapools," circulating SARS-CoV-2-specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, resp. CD4+ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike, and N proteins each accounted for 11%-27% of the total CD4+ response, with addnl. responses commonly targeting nsp3, nsp4, ORF3a, and ORF8, among others. For CD8+ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2-reactive CD4+ T cells in ∼40%-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating "common cold" coronaviruses and SARS-CoV-2.
- 38Wilk, A. J.; Rustagi, A.; Zhao, N. Q.; Roque, J.; Martinez-Colon, G. J.; McKechnie, J. L.; Ivison, G. T.; Ranganath, T.; Vergara, R.; Hollis, T.; Simpson, L. J.; Grant, P.; Subramanian, A.; Rogers, A. J.; Blish, C. A. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020, 26 (7), 1070– 1076, DOI: 10.1038/s41591-020-0944-yGoogle Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFWltb%252FO&md5=93c273cbcdba588f4a980a2b0fa00460A single-cell atlas of the peripheral immune response in patients with severe COVID-19Wilk, Aaron J.; Rustagi, Arjun; Zhao, Nancy Q.; Roque, Jonasel; Martinez-Colon, Giovanny J.; McKechnie, Julia L.; Ivison, Geoffrey T.; Ranganath, Thanmayi; Vergara, Rosemary; Hollis, Taylor; Simpson, Laura J.; Grant, Philip; Subramanian, Aruna; Rogers, Angela J.; Blish, Catherine A.Nature Medicine (New York, NY, United States) (2020), 26 (7), 1070-1076CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)Abstr.: There is an urgent need to better understand the pathophysiol. of Coronavirus disease 2019 (COVID-19), the global pandemic caused by SARS-CoV-2, which has infected more than three million people worldwide1. Approx. 20% of patients with COVID-19 develop severe disease and 5% of patients require intensive care2. Severe disease has been assocd. with changes in peripheral immune activity, including increased levels of pro-inflammatory cytokines that may be produced by a subset of inflammatory monocytes , lymphopenia and T cell exhaustion . To elucidate pathways in peripheral immune cells that might lead to immunopathol. or protective immunity in severe COVID-19, we applied single-cell RNA sequencing (scRNA-seq) to profile peripheral blood mononuclear cells (PBMCs) from seven patients hospitalized for COVID-19, four of whom had acute respiratory distress syndrome, and six healthy controls. We identify reconfiguration of peripheral immune cell phenotype in COVID-19, including a heterogeneous interferon-stimulated gene signature, HLA class II downregulation and a developing neutrophil population that appears closely related to plasmablasts appearing in patients with acute respiratory failure requiring mech. ventilation. Importantly, we found that peripheral monocytes and lymphocytes do not express substantial amts. of pro-inflammatory cytokines. Collectively, we provide a cell atlas of the peripheral immune response to severe COVID-19.
- 39Wang, W.; Zhang, W.; Zhang, J.; He, J.; Zhu, F. Distribution of HLA allele frequencies in 82 Chinese individuals with coronavirus disease-2019 (COVID-19). HLA 2020, 96 (2), 194– 196, DOI: 10.1111/tan.13941Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWnur7O&md5=d15100330a68e382ff06d9790340e796Distribution of HLA allele frequencies in 82 Chinese individuals with coronavirus disease-2019 (COVID-19)Wang, Wei; Zhang, Wei; Zhang, Jingjing; He, Ji; Zhu, FamingHLA (2020), 96 (2), 194-196CODEN: HLAAAK; ISSN:2059-2310. (John Wiley & Sons Ltd.)COVID-19 is a respiratory disease caused by a novel coronavirus and is currently a global pandemic. HLA variation is assocd. with COVID-19 because HLA plays a pivotal role in the immune response to pathogens. Here, 82 individuals with COVID-19 were genotyped for HLA-A, -B, -C, -DRB1, -DRB3/4/5, -DQA1, -DQB1, -DPA1, and -DPB1 loci using next-generation sequencing (NGS). Frequencies of the HLA-C*07:29, C*08:01G, B*15:27, B*40:06, DRB1*04:06, and DPB1*36:01 alleles were higher, while the frequencies of the DRB1*12:02 and DPB1*04:01 alleles were lower in COVID-19 patients than in the control population, with uncorrected statistical significance. Only HLA-C*07:29 and B*15:27 were significant when the cor. P-value was considered. These data suggested that some HLA alleles may be assocd. with the occurrence of COVID-19.
- 40de Castro, J. A. L. How ERAP1 and ERAP2 Shape the Peptidomes of Disease-Associated MHC-I Proteins. Front. Immunol. 2018, 9, 2463, DOI: 10.3389/fimmu.2018.02463Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXosVGmtbk%253D&md5=b76e03061a0bac1bdfa3983c25b32b6cHow ERAP1 and ERAP2 shape the peptidomes of disease-associated MHC-I proteinsde Castro, Jose A. LopezFrontiers in Immunology (2018), 9 (), 2463/1-2463/17CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. Four inflammatory diseases are strongly assocd. with Major Histocompatibility Complex class I (MHC-I) mols birdshot chorioretinopathy (HLA-A*29:02), ankylosing spondylitis (HLA-B*27), Behcet's disease (HLA-B*51), and psoriasis (HLA-C*06:02). The endoplasmic reticulum aminopeptidases (ERAP) 1 and 2 are also risk factors for these diseases. Since both enzymes are involved in the final processing steps of MHC-I ligands it is reasonable to assume that MHC-I-bound peptides play a significant pathogenetic role. This review will mainly focus on recent studies concerning the effects of ERAP1 and ERAP2 polymorphism and expression on shaping the peptidome of disease-assocd. MHC-I mols. in live cells. These studies will be discussed in the context of the distinct mechanisms and substrate preferences of both enzymes, their different patterns of genetic assocn. with various diseases, the role of polymorphisms detg. changes in enzymic activity or expression levels, and the distinct peptidomes of disease-assocd. MHC-I allotypes. ERAP1 and ERAP2 polymorphism and expression induce significant changes in multiple MHC-I-bound peptidomes. These changes are MHC allotype-specific and, without excluding a degree of functional inter-dependence between both enzymes, reflect largely sep. roles in their processing of MHC-I ligands. The studies reviewed here provide a mol. basis for the distinct patterns of genetic assocn. of ERAP1 and ERAP2 with disease and for the pathogenetic role of peptides. The allotype-dependent alterations induced on distinct peptidomes may explain that the joint assocn. of both enzymes and unrelated MHC-I alleles influence different pathol. outcomes.
- 41Infantes, S.; Samino, Y.; Lorente, E.; Jimenez, M.; Garcia, R.; Del Val, M.; Lopez, D. Cutting Edge: H-2L(d) Class I Molecule Protects an HIV N-Extended Epitope from In Vitro Trimming by Endoplasmic Reticulum Aminopeptidase Associated with Antigen Processing. J. Immunol. 2010, 184 (7), 3351– 3355, DOI: 10.4049/jimmunol.0901560Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1Wrtr8%253D&md5=b8b6326efbcd612b287b7677c35cb4d4Cutting Edge: H-2Ld Class I Molecule Protects an HIV N-Extended Epitope from In Vitro Trimming by Endoplasmic Reticulum Aminopeptidase Associated with Antigen ProcessingInfantes, Susana; Samino, Yolanda; Lorente, Elena; Jimenez, Mercedes; Garcia, Ruth; Del Val, Margarita; Lopez, DanielJournal of Immunology (2010), 184 (7), 3351-3355CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)In the classical MHC class I Ag presentation pathway, antigenic peptides derived from viral proteins by multiple proteolytic cleavages are transported to the endoplasmic reticulum lumen and are then exposed to aminopeptidase activity. In the current study, a long MHC class I natural ligand recognized by cytotoxic T lymphocytes was used to study the kinetics of degrdn. by aminopeptidase. The in vitro data indicate that this N-extended peptide is efficiently trimmed to a 9-mer, unless its binding to the MHC mols. protects the full-length peptide.
- 42Thomas, C.; Tampe, R. MHC I chaperone complexes shaping immunity. Curr. Opin. Immunol. 2019, 58, 9– 15, DOI: 10.1016/j.coi.2019.01.001Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1CltLo%253D&md5=a9963ca05f5b64cd0118525ec7b9a8c0MHC I chaperone complexes shaping immunityThomas, Christoph; Tampe, RobertCurrent Opinion in Immunology (2019), 58 (), 9-15CODEN: COPIEL; ISSN:0952-7915. (Elsevier Ltd.)Major histocompatibility complex class I (MHC I) mols. present peptides on the surface of most nucleated cells and allow the immune system to detect and eliminate infected or malignantly transformed cells. The peptides are derived from endogenous proteins by proteasomal degrdn. or aberrant translation, and are translocated from the cytosol into the endoplasmic reticulum (ER) by the transporter assocd. with antigen processing (TAP), a central component of the peptide-loading complex (PLC). The peptides are subsequently processed by ER-resident aminopeptidases (ERAP1/2) and loaded onto MHC I. This loading, however, does not happen indiscriminately: in a process called peptide editing or peptide proofreading, the MHC I-specific chaperones tapasin and TAPBPR (TAP-binding protein-related) catalyze the selection of high-affinity peptides and stable peptide-MHC I (pMHC I) complexes. Once correctly loaded with a high-affinity peptide, pMHC I complexes travel to the cell surface where they are recognized by T lymphocytes to control their differentiation in the thymus, their priming in the lymph node, and their final long-term surveillance of target cells in the periphery. Recent structural studies of the PLC and of TAPBPR-MHC I complexes by single-particle cryo-electron microscopy, X-ray crystallog., and NMR spectroscopy have provided fundamental insights into the mechanisms of MHC I peptide loading and proofreading, highlighting the dynamic nature of the involved complexes and the conformational plasticity of the individual proteins.
- 43Chen, H.; Li, L.; Weimershaus, M.; Evnouchidou, I.; van Endert, P.; Bouvier, M. ERAP1-ERAP2 dimers trim MHC I-bound precursor peptides; implications for understanding peptide editing. Sci. Rep. 2016, 6, 28902, DOI: 10.1038/srep28902Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlCit7fP&md5=9f3e7a8e07dd2a54a0af3f78772bcd04ERAP1-ERAP2 dimers trim MHC I-bound precursor peptides; implications for understanding peptide editingChen, Hanna; Li, Lenong; Weimershaus, Mirjana; Evnouchidou, Irini; van Endert, Peter; Bouvier, MarleneScientific Reports (2016), 6 (), 28902CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)The processing of MHC class I antigenic precursor peptides by the endoplasmic reticulum aminopeptidase 1 (ERAP1) and ERAP2 is an important event in the cell biol. of antigen presentation. To date, the mol. context by which the ERAP enzymes trim precursor peptides, and how ERAPs shape peptide repertoires, remain open questions. Using ERAP1 and ERAP2 heterodimers (ERAP1/2), and N-terminally extended model and natural peptides in their free and HLA-B*0801-bound forms, we characterized the mode of action of ERAPs. We provide evidence that ERAP1/2 can trim MHC I-bound precursor peptides to their correct and final lengths, albeit more slowly than the corresponding free precursors. Trimming of MHC I-bound precursors by ERAP1/2 increases the conformational stability of MHC I/peptide complexes. From the data, we propose a mol. mechanistic model of ERAP1/2 as peptide editors. Overall, our study provides new findings on a significant issue of the ERAP-mediated processing pathway of MHC class I antigens.
- 44Mavridis, G.; Arya, R.; Domnick, A.; Zoidakis, J.; Makridakis, M.; Vlahou, A.; Mpakali, A.; Lelis, A.; Georgiadis, D.; Tampe, R.; Papakyriakou, A.; Stern, L. J.; Stratikos, E. A systematic re-examination of processing of MHCI-bound antigenic peptide precursors by endoplasmic reticulum aminopeptidase 1. J. Biol. Chem. 2020, 295 (21), 7193– 7210, DOI: 10.1074/jbc.RA120.012976Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1Wqt7vP&md5=5385c68385264ad3cc20973aff0fbf69A systematic re-examination of processing of MHCI-bound antigenic peptide precursors by endoplasmic reticulum aminopeptidase 1Mavridis, George; Arya, Richa; Domnick, Alexander; Zoidakis, Jerome; Makridakis, Manousos; Vlahou, Antonia; Mpakali, Anastasia; Lelis, Angelos; Georgiadis, Dimitris; Tampe, Robert; Papakyriakou, Athanasios; Stern, Lawrence J.; Stratikos, EfstratiosJournal of Biological Chemistry (2020), 295 (21), 7193-7211CODEN: JBCHA3; ISSN:1083-351X. (American Society for Biochemistry and Molecular Biology)Endoplasmic reticulum aminopeptidase 1 (ERAP1) trims antigenic peptide precursors to generate mature antigenic peptides for presentation by major histocompatibility complex class I (MHCI) mols. and regulates adaptive immune responses. ERAP1 has been proposed to trim peptide precursors both in soln. and in preformed MHCI-peptide complexes, but which mode is more relevant to its biol. function remains controversial. Here, we compared ERAP1-mediated trimming of antigenic peptide precursors in soln. or when bound to three MHCI alleles, HLA-B*58, HLA-B*08, and HLA-A*02. For all MHCI-peptide combinations, peptide binding onto MHCI protected against ERAP1-mediated trimming. In only a single MHCI-peptide combination, trimming of an HLA-B*08-bound 12-mer progressed at a considerable rate, albeit still slower than in soln. Results from thermodn., kinetic, and computational analyses suggested that this 12-mer is highly labile and that apparent on-MHC trimming rates are always slower than that of MHCI-peptide dissocn. Both ERAP2 and leucine aminopeptidase, an enzyme unrelated to antigen processing, could trim this labile peptide from preformed MHCI complexes as efficiently as ERAP1. A pseudopeptide analog with high affinity for both HLA-B*08 and the ERAP1 active site could not promote the formation of a ternary ERAP1/MHCI/peptide complex. Similarly, no interactions between ERAP1 and purified peptide-loading complex were detected in the absence or presence of a pseudopeptide trap. We conclude that MHCI binding protects peptides from ERAP1 degrdn. and that trimming in soln. along with the dynamic nature of peptide binding to MHCI are sufficient to explain ERAP1 processing of antigenic peptide precursors.
- 45Perez-Riverol, Y.; Csordas, A.; Bai, J.; Bernal-Llinares, M.; Hewapathirana, S.; Kundu, D. J.; Inuganti, A.; Griss, J.; Mayer, G.; Eisenacher, M.; Perez, E.; Uszkoreit, J.; Pfeuffer, J.; Sachsenberg, T.; Yilmaz, S.; Tiwary, S.; Cox, J.; Audain, E.; Walzer, M.; Jarnuczak, A. F.; Ternent, T.; Brazma, A.; Vizcaino, J. A. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 2019, 47 (D1), D442– D450, DOI: 10.1093/nar/gky1106Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1GqtrzK&md5=4cd0f929c2df840a5cc0631d0d708d6cThe PRIDE database and related tools and resources in 2019: improving support for quantification dataPerez-Riverol, Yasset; Csordas, Attila; Bai, Jingwen; Bernal-Llinare, Manuel; Hewapathirana, Suresh; Kundu, Deepti J.; Iuganti, Avinash; Griss, Johannes; Mayer, Gerhard; Eisenacher, Martin; Perez, Enrique; Uszkoreit, Julian; Pfeuffer, Julianus; Sachsenberg, Timo; Yilmaz, Sule; Tiwary, Shivani; Cox, Jurgen; Audain, Enrique; Walzer, Mathias; Jarnuczak, Andrew F.; Ternent, Tobias; Brazma, Alvis; Vizcaino, Juan AntonioNucleic Acids Research (2019), 47 (D1), D442-D450CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The PRoteomics IDEntifications (PRIDE) database is the world's largest data repository of mass spectrometry-based proteomics data, and is one of the founding members of the global ProteomeXchange (PX) consortium. In this manuscript, we summarize the developments in PRIDE resources and related tools since the previous update manuscript was published in Nucleic Acids Research in 2016. In the last 3 years, public data sharing through PRIDE (as part of PX) has definitely become the norm in the field. In parallel, data re-use of public proteomics data has increased enormously, with multiple applications. We first describe the new architecture of PRIDE Archive, the archival component of PRIDE. PRIDE Archive and the related data submission framework have been further developed to support the increase in submitted data vols. and addnl. data types. A new scalable and fault tolerant storage backed, Application Programming Interface and web interface have been implemented, as a part of an ongoing process. Addnl., we emphasize the improved support for quant. proteomics data through the mzTab format. At last, we outline key statistics on the current data contents and vol. of downloads, and how PRIDE data are starting to be disseminated to added-value resources including Ensembl, UniProt and Expression Atlas.
Cited By
This article is cited by 12 publications.
- Amany A. Ghazy, Abdulrahman H. Almaeen, Ibrahim A. Taher, Abdullah N. Alrasheedi, Amel Elsheredy. Impact of ACE and Endoplasmic Reticulum Aminopeptidases Polymorphisms on COVID-19 Outcome. Diagnostics 2023, 13
(2)
, 305. https://doi.org/10.3390/diagnostics13020305
- Diana Laura Pacheco-Olvera, Stephanie Saint Remy-Hernández, María Guadalupe García-Valeriano, Tania Rivera-Hernández, Constantino López-Macías. Bioinformatic Analysis of B- and T-cell Epitopes from SARS-CoV-2 Structural Proteins and their Potential Cross-reactivity with Emerging Variants and other Human Coronaviruses. Archives of Medical Research 2022, 53
(7)
, 694-710. https://doi.org/10.1016/j.arcmed.2022.10.007
- Dennis Jiménez, Marbel Torres Arias. Immunouniverse of SARS-CoV-2. Immunological Medicine 2022, 45
(4)
, 186-224. https://doi.org/10.1080/25785826.2022.2066251
- Virgyl Camberlein, Charlotte Fléau, Pierre Sierocki, Lenong Li, Ronan Gealageas, Damien Bosc, Valentin Guillaume, Sandrine Warenghem, Florence Leroux, Melissa Rosell, Keguang Cheng, Laura Medve, Mathilde Prigent, Myriam Decanter, Catherine Piveteau, Alexandre Biela, Maxime Eveque, Julie Dumont, Anastasia Mpakali, Petros Giastas, Adrien Herledan, Cyril Couturier, Jörg Haupenthal, Laetitia Lesire, Anna K. H. Hirsch, Benoit Deprez, Efstratios Stratikos, Marlene Bouvier, Rebecca Deprez‐Poulain. Discovery of the First Selective Nanomolar Inhibitors of ERAP2 by Kinetic Target‐Guided Synthesis. Angewandte Chemie International Edition 2022, 61
(39)
https://doi.org/10.1002/anie.202203560
- Virgyl Camberlein, Charlotte Fléau, Pierre Sierocki, Lenong Li, Ronan Gealageas, Damien Bosc, Valentin Guillaume, Sandrine Warenghem, Florence Leroux, Melissa Rosell, Keguang Cheng, Laura Medve, Mathilde Prigent, Myriam Decanter, Catherine Piveteau, Alexandre Biela, Maxime Eveque, Julie Dumont, Anastasia Mpakali, Petros Giastas, Adrien Herledan, Cyril Couturier, Jörg Haupenthal, Laetitia Lesire, Anna K. H. Hirsch, Benoit Deprez, Efstratios Stratikos, Marlene Bouvier, Rebecca Deprez‐Poulain. Discovery of the First Selective Nanomolar Inhibitors of ERAP2 by Kinetic Target‐Guided Synthesis. Angewandte Chemie 2022, 134
(39)
https://doi.org/10.1002/ange.202203560
- Piotr Kuśnierczyk. To Be or Not to Be: The Case of Endoplasmic Reticulum Aminopeptidase 2. Frontiers in Immunology 2022, 13 https://doi.org/10.3389/fimmu.2022.902567
- Ahmed Samir Abdelhafiz, Asmaa Ali, Merhan A. Fouda, Douaa M. Sayed, Mahmoud M Kamel, Lamyaa Mohamed Kamal, Mahmoud Ali Khalil, Rania M. Bakry. HLA-B*15 predicts survival in Egyptian patients with COVID-19. Human Immunology 2022, 83
(1)
, 10-16. https://doi.org/10.1016/j.humimm.2021.09.007
- Zhenlin Yang, He Tian, Fenglong Bie, Jiachen Xu, Zheng Zhou, Junhui Yang, Renda Li, Yue Peng, Guangyu Bai, Yanhua Tian, Ying Chen, Lei Liu, Tao Fan, Chu Xiao, Yujia Zheng, Bo Zheng, Jie Wang, Chunxiang Li, Shugeng Gao, Jie He. ERAP2 Is Associated With Immune Infiltration and Predicts Favorable Prognosis in SqCLC. Frontiers in Immunology 2021, 12 https://doi.org/10.3389/fimmu.2021.788985
- George Stamatakis, Martina Samiotaki, Ioannis Temponeras, George Panayotou, Efstratios Stratikos. Allotypic variation in antigen processing controls antigenic peptide generation from SARS-CoV-2 S1 spike glycoprotein. Journal of Biological Chemistry 2021, 297
(5)
, 101329. https://doi.org/10.1016/j.jbc.2021.101329
- Dannielle Wellington, Zixi Yin, Benedikt M Kessler, Tao Dong. Immunodominance complexity: lessons yet to be learned from dominant T cell responses to SARS-COV-2. Current Opinion in Virology 2021, 50 , 183-191. https://doi.org/10.1016/j.coviro.2021.08.009
- Irma Saulle, Chiara Vicentini, Mario Clerici, Mara Biasin. Antigen presentation in SARS-CoV-2 infection: the role of class I HLA and ERAP polymorphisms. Human Immunology 2021, 82
(8)
, 551-560. https://doi.org/10.1016/j.humimm.2021.05.003
- Silvia D’Amico, Patrizia Tempora, Valeria Lucarini, Ombretta Melaiu, Stefania Gaspari, Mattia Algeri, Doriana Fruci. ERAP1 and ERAP2 Enzymes: A Protective Shield for RAS against COVID-19?. International Journal of Molecular Sciences 2021, 22
(4)
, 1705. https://doi.org/10.3390/ijms22041705
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 45 other publications.
- 1Coronaviridae Study Group of the International Committee on Taxonomy of Viruses The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5 (4), 536– 544, DOI: 10.1038/s41564-020-0695-z1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXktFemu7Y%253D&md5=3e43e7781469c6d748726b24b3a60361The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2Coronaviridae Study Group of the International Committee on Taxonomy of VirusesNature Microbiology (2020), 5 (4), 536-544CODEN: NMAICH; ISSN:2058-5276. (Nature Research)A review. The present outbreak of a coronavirus-assocd. acute respiratory disease called coronavirus disease 19 (COVID-19) is the third documented spillover of an animal coronavirus to humans in only two decades that has resulted in a major epidemic. The Coronaviridae Study Group (CSG) of the International Committee on Taxonomy of Viruses, which is responsible for developing the classification of viruses and taxon nomenclature of the family Coronaviridae, has assessed the placement of the human pathogen, tentatively named 2019-nCoV, within the Coronaviridae. Based on phylogeny, taxonomy and established practice, the CSG recognizes this virus as forming a sister clade to the prototype human and bat severe acute respiratory syndrome coronaviruses (SARS-CoVs) of the species Severe acute respiratory syndrome-related coronavirus, and designates it as SARS-CoV-2. In order to facilitate communication, the CSG proposes to use the following naming convention for individual isolates: SARS-CoV-2/host/location/isolate/date. While the full spectrum of clin. manifestations assocd. with SARS-CoV-2 infections in humans remains to be detd., the independent zoonotic transmission of SARS-CoV and SARS-CoV-2 highlights the need for studying viruses at the species level to complement research focused on individual pathogenic viruses of immediate significance. This will improve our understanding of virus-host interactions in an ever-changing environment and enhance our preparedness for future outbreaks.
- 2Wu, F.; Zhao, S.; Yu, B.; Chen, Y. M.; Wang, W.; Song, Z. G.; Hu, Y.; Tao, Z. W.; Tian, J. H.; Pei, Y. Y.; Yuan, M. L.; Zhang, Y. L.; Dai, F. H.; Liu, Y.; Wang, Q. M.; Zheng, J. J.; Xu, L.; Holmes, E. C.; Zhang, Y. Z. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579 (7798), 265– 269, DOI: 10.1038/s41586-020-2008-32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksFKlsLc%253D&md5=0163a684829e880a0c3347e19f0ce52aA new coronavirus associated with human respiratory disease in ChinaWu, Fan; Zhao, Su; Yu, Bin; Chen, Yan-Mei; Wang, Wen; Song, Zhi-Gang; Hu, Yi; Tao, Zhao-Wu; Tian, Jun-Hua; Pei, Yuan-Yuan; Yuan, Ming-Li; Zhang, Yu-Ling; Dai, Fa-Hui; Liu, Yi; Wang, Qi-Min; Zheng, Jiao-Jiao; Xu, Lin; Holmes, Edward C.; Zhang, Yong-ZhenNature (London, United Kingdom) (2020), 579 (7798), 265-269CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Emerging infectious diseases, such as severe acute respiratory syndrome (SARS) and Zika virus disease, present a major threat to public health. Despite intense research efforts, how, when and where new diseases appear are still a source of considerable uncertainty. A severe respiratory disease was recently reported in Wuhan, Hubei province, China. As of 25 Jan. 2020, at least 1,975 cases had been reported since the first patient was hospitalized on 12 Dec. 2019. Epidemiol. investigations have suggested that the outbreak was assocd. with a seafood market in Wuhan. Here we study a single patient who was a worker at the market and who was admitted to the Central Hospital of Wuhan on 26 Dec. 2019 while experiencing a severe respiratory syndrome that included fever, dizziness and a cough. Metagenomic RNA sequencing of a sample of bronchoalveolar lavage fluid from the patient identified a new RNA virus strain from the family Coronaviridae, which is designated here 'WH-Human 1' coronavirus (and has also been referred to as '2019-nCoV'). Phylogenetic anal. of the complete viral genome (29,903 nucleotides) revealed that the virus was most closely related (89.1% nucleotide similarity) to a group of SARS-like coronaviruses (genus Betacoronavirus, subgenus Sarbecovirus) that had previously been found in bats in China. This outbreak highlights the ongoing ability of viral spill-over from animals to cause severe disease in humans.
- 3Zhou, P.; Yang, X. L.; Wang, X. G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H. R.; Zhu, Y.; Li, B.; Huang, C. L.; Chen, H. D.; Chen, J.; Luo, Y.; Guo, H.; Jiang, R. D.; Liu, M. Q.; Chen, Y.; Shen, X. R.; Wang, X.; Zheng, X. S.; Zhao, K.; Chen, Q. J.; Deng, F.; Liu, L. L.; Yan, B.; Zhan, F. X.; Wang, Y. Y.; Xiao, G. F.; Shi, Z. L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579 (7798), 270– 273, DOI: 10.1038/s41586-020-2012-73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXksFKlsLg%253D&md5=236f17d4d3c7978d72513e5e0258f1b3A pneumonia outbreak associated with a new coronavirus of probable bat originZhou, Peng; Yang, Xing-Lou; Wang, Xian-Guang; Hu, Ben; Zhang, Lei; Zhang, Wei; Si, Hao-Rui; Zhu, Yan; Li, Bei; Huang, Chao-Lin; Chen, Hui-Dong; Chen, Jing; Luo, Yun; Guo, Hua; Jiang, Ren-Di; Liu, Mei-Qin; Chen, Ying; Shen, Xu-Rui; Wang, Xi; Zheng, Xiao-Shuang; Zhao, Kai; Chen, Quan-Jiao; Deng, Fei; Liu, Lin-Lin; Yan, Bing; Zhan, Fa-Xian; Wang, Yan-Yi; Xiao, Geng-Fu; Shi, Zheng-LiNature (London, United Kingdom) (2020), 579 (7798), 270-273CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large no. of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats1-4. Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans5-7. Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 Dec. 2019, had caused 2,794 lab.-confirmed infections including 80 deaths by 26 Jan. 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence anal. of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addn., 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor-angiotensin converting enzyme II (ACE2)-as SARS-CoV.
- 4Tortorici, M. A.; Veesler, D. Structural insights into coronavirus entry. Adv. Virus Res. 2019, 105, 93– 116, DOI: 10.1016/bs.aivir.2019.08.0024https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1aisLfE&md5=7c7d72fb74a490b922fe2da65c8615fbStructural insights into coronavirus entryTortorici, M. Alejandra; Veesler, DavidAdvances in Virus Research (2019), 105 (Complementary Strategies to Understand Virus Structure and Function), 93-116CODEN: AVREA8; ISSN:0065-3527. (Elsevier Ltd.)A review. Coronaviruses (CoVs) have caused outbreaks of deadly pneumonia in humans since the beginning of the 21st century. The severe acute respiratory syndrome coronavirus (SARS-CoV) emerged in 2002 and was responsible for an epidemic that spread to five continents with a fatality rate of 10% before being contained in 2003 (with addnl. cases reported in 2004). The Middle-East respiratory syndrome coronavirus (MERS-CoV) emerged in the Arabian Peninsula in 2012 and has caused recurrent outbreaks in humans with a fatality rate of 35%. SARS-CoV and MERS-CoV are zoonotic viruses that crossed the species barrier using bats/palm civets and dromedary camels, resp. No specific treatments or vaccines have been approved against any of the six human coronaviruses, highlighting the need to investigate the principles governing viral entry and cross-species transmission as well as to prep. for zoonotic outbreaks which are likely to occur due to the large reservoir of CoVs found in mammals and birds. Here, we review our understanding of the infection mechanism used by coronaviruses derived from recent structural and biochem. studies.
- 5Walls, A. C.; Park, Y. J.; Tortorici, M. A.; Wall, A.; McGuire, A. T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 181 (2), 281– 292, DOI: 10.1016/j.cell.2020.02.0585https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVejsLk%253D&md5=ac8a8a208d9c26f88f702fb7634ab1abStructure, Function, and Antigenicity of the SARS-CoV-2 Spike GlycoproteinWalls, Alexandra C.; Park, Young-Jun; Tortorici, M. Alejandra; Wall, Abigail; McGuire, Andrew T.; Veesler, DavidCell (Cambridge, MA, United States) (2020), 181 (2), 281-292.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We detd. cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.
- 6Shang, J.; Ye, G.; Shi, K.; Wan, Y.; Luo, C.; Aihara, H.; Geng, Q.; Auerbach, A.; Li, F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020, 581 (7807), 221– 224, DOI: 10.1038/s41586-020-2179-y6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtbs%253D&md5=33bc9151641b2adcfb0dbf446621a1dcStructural basis of receptor recognition by SARS-CoV-2Shang, Jian; Ye, Gang; Shi, Ke; Wan, Yushun; Luo, Chuming; Aihara, Hideki; Geng, Qibin; Auerbach, Ashley; Li, FangNature (London, United Kingdom) (2020), 581 (7807), 221-224CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) recently emerged and is rapidly spreading in humans, causing COVID-191,2. A key to tackling this pandemic is to understand the receptor recognition mechanism of the virus, which regulates its infectivity, pathogenesis and host range. SARS-CoV-2 and SARS-CoV recognize the same receptor-angiotensin-converting enzyme 2 (ACE2)-in humans3,4. Here we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 (engineered to facilitate crystn.) in complex with ACE2. In comparison with the SARS-CoV RBD, an ACE2-binding ridge in SARS-CoV-2 RBD has a more compact conformation; moreover, several residue changes in the SARS-CoV-2 RBD stabilize two virus-binding hotspots at the RBD-ACE2 interface. These structural features of SARS-CoV-2 RBD increase its ACE2-binding affinity. Addnl., we show that RaTG13, a bat coronavirus that is closely related to SARS-CoV-2, also uses human ACE2 as its receptor. The differences among SARS-CoV-2, SARS-CoV and RaTG13 in ACE2 recognition shed light on the potential animal-to-human transmission of SARS-CoV-2. This study provides guidance for intervention strategies that target receptor recognition by SARS-CoV-2.
- 7St John, A. L.; Rathore, A. P. S. Early Insights into Immune Responses during COVID-19. J. Immunol. 2020, 205 (3), 555– 564, DOI: 10.4049/jimmunol.20005267https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1WgsbnK&md5=6d3b7343f9234da184e3ae1cf30e4b1eEarly insights into immune responses during COVID-19St. John, Ashley L.; Rathore, Abhay P. S.Journal of Immunology (2020), 205 (3), 555-564CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)A review. Coronavirus disease-2019 (COVID-19) is caused by the newly emerged virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and was recently declared as a pandemic by the World Health Organization. In its severe form, the disease is characterized by acute respiratory distress syndrome, and there are no targeted intervention strategies to treat or prevent it. The immune response is thought to both contribute to the pathogenesis of disease and provide protection during its resoln. Thus, understanding the immune response to SARS-CoV-2 is of the utmost importance for developing and testing vaccines and therapeutics. In this review, we discuss the earliest knowledge and hypotheses of the mechanisms of immune pathol. in the lung during acute infection as well at the later stages of disease resoln., recovery, and immune memory formation.
- 8Song, P.; Li, W.; Xie, J.; Hou, Y.; You, C. Cytokine storm induced by SARS-CoV-2. Clin. Chim. Acta 2020, 509, 280– 287, DOI: 10.1016/j.cca.2020.06.0178https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlWns7%252FE&md5=cafd0380779103347d4867c3a893db38Cytokine storm induced by SARS-CoV-2Song, Peipei; Li, Wei; Xie, Jianqin; Hou, Yanlong; You, ChonggeClinica Chimica Acta (2020), 509 (), 280-287CODEN: CCATAR; ISSN:0009-8981. (Elsevier B.V.)A review. Coronavirus disease 2019 (COVID-19), caused by the virus designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread widely throughout the world. Despite the strict global outbreak management and quarantine measures that have been implemented, the incidence of COVID-19 continues to rise, resulting in more than 290,000 deaths and representing an extremely serious threat to human life and health. The clin. symptoms of the affected patients are heterogeneous, ranging from mild upper respiratory symptoms to severe pneumonitis and even acute respiratory distress syndrome (ARDS) or death. Systemic immune over activation due to SARS-CoV-2 infection causes the cytokine storm, which is esp. noteworthy in severely ill patients with COVID-19. Pieces of evidence from current studies have shown that the cytokine storm may be an important factor in disease progression, even leading to multiple organ failure and death. This review provides an overview of the knowledge on the COVID-19 epidemiol. profile, the mol. mechanisms of the SARS-CoV-2-induced cytokine storm and immune responses, the pathophysiol. changes that occur during infection, the main antiviral compds. used in treatment strategies and the potential drugs for targeting cytokines, this information is presented to provide valuable guidance for further studies and for a therapeutic redn. of this excessive immune response.
- 9Li, K.; Hao, Z.; Zhao, X.; Du, J.; Zhou, Y. SARS-CoV-2 infection-induced immune responses: Friends or foes?. Scand. J. Immunol. 2020, 92 (2), e12895 DOI: 10.1111/sji.128959https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVKms7rI&md5=a2e2c5f90a25f0a9ef9476f0d5517504SARS-CoV-2 infection-induced immune responses: Friends or foes?Li, Keying; Hao, Zhenhua; Zhao, Xiaohui; Du, Jiying; Zhou, YanlinScandinavian Journal of Immunology (2020), 92 (2), e12895CODEN: SJIMAX; ISSN:1365-3083. (Wiley-Blackwell)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging coronavirus that belongs to the β-genus, causing the outbreak of coronavirus disease 19 (COVID-19). SARS-CoV-2 infection can stimulate a pronounced immune response in the host, which embodies in the decrease of lymphocytes and aberrant increase of cytokines in COVID-19 patients. SARS-CoV-2 RNA and proteins interact with various pattern recognition receptors that switch on antiviral immune responses to regulate viral replication and spreading within the host in vivo. However, overactive and impaired immune responses also cause immune damage and subsequent tissue inflammation. This article focuses on the dual roles of immune system during SARS-CoV-2 infection, providing a theor. basic for identifying therapeutic targets in a situation with an unfavorable immune reaction.
- 10Peeples, L. News Feature: Avoiding pitfalls in the pursuit of a COVID-19 vaccine. Proc. Natl. Acad. Sci. U. S. A. 2020, 117 (15), 8218– 8221, DOI: 10.1073/pnas.200545611710https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXntFOisL4%253D&md5=1357011c0b1e556332c345ee33af1816News Feature: Avoiding pitfalls in the pursuit of a COVID-19 vaccinePeeples, LynneProceedings of the National Academy of Sciences of the United States of America (2020), 117 (15), 8218-8221CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)A review. As researchers scramble to develop a Covid-19 vaccine, they are debating which, if any, of the phenomena related to a virus infection-aiding immune "enhancement" such as antibody-dependent enhancement (ADE) or cell-based enhancement including allergic inflammation caused by Th2 immunopathol., could be at play in the case of Covid-19 and how these phenomena might affect the success of vaccine candidates. Vaccine experts have underscored the need to avoid past mistakes including the halting of SARS vaccine development. Ecol. disruption increases the odds that a future coronavirus will jump from other hosts to humans.
- 11Leslie, M. T cells found in coronavirus patients ‘bode well’ for long-term immunity. Science 2020, 368 (6493), 809– 810, DOI: 10.1126/science.368.6493.80911https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVWnurjO&md5=697aa456e6b6a501bcb8d77fed0db726T cells found in coronavirus patients 'bode well' for long-term immunityLeslie, MitchScience (Washington, DC, United States) (2020), 368 (6493), 809-810CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)A review. Whether T cells join the immune system's fight against SARS-CoV-2, the virus that causes COVID-19, has been unclear. Two new studies have identified T cells that react to the virus in patients who had COVID-19 or were recovering from it. Both studies detected helper T cells, which orchestrate antiviral responses of other immune cells, and 1 of the studies also found killer T cells that eliminate virally infected cells. Both studies also discovered helper T cells that target the virus in blood samples from people who had never been infected. These people likely had the cells because they had been infected by similar coronaviruses that cause colds. The results suggest the virus induces a strong T cell response in patients, but they do not indicate whether people who have recovered from COVID-19 are immune to the virus.
- 12Manners, C.; Larios Bautista, E.; Sidoti, H.; Lopez, O. J. Protective Adaptive Immunity Against Severe Acute Respiratory Syndrome Coronaviruses 2 (SARS-CoV-2) and Implications for Vaccines. Cureus 2020, 12 (6), e8399 DOI: 10.7759/cureus.8399There is no corresponding record for this reference.
- 13Mukherjee, S.; Tworowski, D.; Detroja, R.; Mukherjee, S. B.; Frenkel-Morgenstern, M. Immunoinformatics and Structural Analysis for Identification of Immunodominant Epitopes in SARS-CoV-2 as Potential Vaccine Targets. Vaccines (Basel, Switz.) 2020, 8 (2), 290, DOI: 10.3390/vaccines8020290There is no corresponding record for this reference.
- 14Rock, K. L.; Goldberg, A. L. Degradation of cell proteins and the generation of MHC class I-presented peptides. Annu. Rev. Immunol. 1999, 17, 739– 79, DOI: 10.1146/annurev.immunol.17.1.73914https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXjtVWmtLY%253D&md5=ad0c421306a313b77d901d7c76c161d5Degradation of cell proteins and the generation of MHC class I-presented peptidesRock, Kenneth L.; Goldberg, Alfred L.Annual Review of Immunology (1999), 17 (), 739-779CODEN: ARIMDU; ISSN:0732-0582. (Annual Reviews Inc.)A review with 211 refs. Major histocompatibility complex (MHC) class I mols. display on the cell surface 8- to 10-residue peptides derived from the spectrum of proteins expressed in the cells. By screening for non-self MHC-bound peptides, the immune system identifies and then can eliminate cells that are producing viral or mutant proteins. These antigenic peptides are generated as side products in the continual turnover of intracellular proteins, which occurs primarily by the ubiquitin-proteasome pathway. Most of the oligopeptides generated by the proteasome are further degraded by distinct endopeptidases and aminopeptidases into amino acids, which are used for new protein synthesis or energy prodn. However, a fraction of these peptides escape complete destruction and after transport into the endoplasmic reticulum are bound by MHC class I mols. and delivered to the cell surface. Herein we review recent discoveries about the proteolytic systems that degrade cell proteins, how the ubiquitin-proteasome pathway generates the peptides presented on MHC-class I mols., and how this process is stimulated by immune modifiers to enhance antigen presentation.
- 15Weimershaus, M.; Evnouchidou, I.; Saveanu, L.; van Endert, P. Peptidases trimming MHC class I ligands. Curr. Opin. Immunol. 2013, 25 (1), 90– 6, DOI: 10.1016/j.coi.2012.10.00115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsFCksLzL&md5=dbed24e460af7509ad6a8c9ac453de5bPeptidases trimming MHC class I ligandsWeimershaus, Mirjana; Evnouchidou, Irini; Saveanu, Loredana; van Endert, PeterCurrent Opinion in Immunology (2013), 25 (1), 90-96CODEN: COPIEL; ISSN:0952-7915. (Elsevier Ltd.)A review. Peptides presented by MHC class I mols. are typically produced through antigen degrdn. by the proteasome followed by trimming by exopeptidases. According to recent results, these include both aminopeptidases and carboxypeptidases in the cytosol and the endoplasmic reticulum. While cytosolic peptidases have a net neutral or destructive effect on MHC ligands, endoplasmic reticulum aminopeptidases are required for efficient class I loading and have a strong effect on the repertoire of peptide/MHC complexes. Cells lacking these enzymes can be eliminated both by NK cells and by CD8+ T cells recognizing complexes formed between an MHC class Ib mol. and a conserved peptide. Cross-presented peptides derived from internalized antigens can be processed by insulin-regulated aminopeptidase, the only endosomal trimming peptidase.
- 16Hammer, G. E.; Kanaseki, T.; Shastri, N. The final touches make perfect the peptide-MHC class I repertoire. Immunity 2007, 26 (4), 397– 406, DOI: 10.1016/j.immuni.2007.04.00316https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXltVKjsrw%253D&md5=d50e9482fd0fa138cfe165615d1329d9The final touches make perfect the peptide-MHC class I repertoireHammer, Gianna Elena; Kanaseki, Takayuki; Shastri, NilabhImmunity (2007), 26 (4), 397-406CODEN: IUNIEH; ISSN:1074-7613. (Cell Press)A review. Major histocompatibility complex (MHC) class I mols. present short, perfectly cleaved peptides on the cell surface for immune surveillance by CD8+ T cells. The pathway for generating these peptides begins in the cytoplasm, and the peptide-MHC I (pMHC I) repertoire is finalized in the endoplasmic reticulum. Recent studies show that the peptides for MHC I are customized by the ER aminopeptidase assocd. with antigen processing and by dynamic interactions within the MHC peptide-loading complex. Failure to customize the pMHC I repertoire has profound immunol. consequences.
- 17Giastas, P.; Mpakali, A.; Papakyriakou, A.; Lelis, A.; Kokkala, P.; Neu, M.; Rowland, P.; Liddle, J.; Georgiadis, D.; Stratikos, E. Mechanism for antigenic peptide selection by endoplasmic reticulum aminopeptidase 1. Proc. Natl. Acad. Sci. U. S. A. 2019, 116 (52), 26709– 26716, DOI: 10.1073/pnas.191207011617https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXjsFCntbw%253D&md5=3115e900c52db599cf264d7c6e0762d8Mechanism for antigenic peptide selection by endoplasmic reticulum aminopeptidase 1Giastas, Petros; Mpakali, Anastasia; Papakyriakou, Athanasios; Lelis, Aggelos; Kokkala, Paraskevi; Neu, Margarete; Rowland, Paul; Liddle, John; Georgiadis, Dimitris; Stratikos, EfstratiosProceedings of the National Academy of Sciences of the United States of America (2019), 116 (52), 26709-26716CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an intracellular enzyme that optimizes the peptide cargo of major histocompatibility class I (MHC-I) mols. and regulates adaptive immunity. It has unusual substrate selectivity for length and sequence, resulting in poorly understood effects on the cellular immunopeptidome. To understand substrate selection by ERAP1, we solved 2 crystal structures of the enzyme with bound transition-state pseudopeptide analogs at 1.68 Å and 1.72 Å. Both peptides have their N terminus bound at the active site and extend away along a large internal cavity, interacting with shallow pockets that can influence selectivity. The longer peptide is disordered through the central region of the cavity and has its C terminus bound in an allosteric pocket of domain IV that features a carboxypeptidase-like structural motif. These structures, along with enzymic and computational analyses, explain how ERAP1 can select peptides based on length while retaining the broad sequence-specificity necessary for its biol. function.
- 18Stratikos, E.; Stern, L. J. Antigenic peptide trimming by ER aminopeptidases--insights from structural studies. Mol. Immunol. 2013, 55 (3–4), 212– 9, DOI: 10.1016/j.molimm.2013.03.00218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXltVent7k%253D&md5=b6318876699d28ee425936e83c462031Antigenic peptide trimming by ER aminopeptidases - Insights from structural studiesStratikos, Efstratios; Stern, Lawrence J.Molecular Immunology (2013), 55 (3-4), 212-219CODEN: MOIMD5; ISSN:0161-5890. (Elsevier)A review. Generation and destruction of antigenic peptides by ER resident aminopeptidases ERAP1 and ERAP2 have been shown in the last few years to be important for the correct functioning and regulation of the adaptive immune response. These two highly homologous aminopeptidases appear to have evolved complex mechanisms well suited for their biol. role in antigen presentation. Furthermore, polymorphic variability in these enzymes appears to affect their function and predispose individuals to disease. This review discusses our current understanding of the mol. mechanisms behind ERAP1/2 function as suggested by several recently detd. crystallog. structures of these enzymes.
- 19Evnouchidou, I.; Kamal, R. P.; Seregin, S. S.; Goto, Y.; Tsujimoto, M.; Hattori, A.; Voulgari, P. V.; Drosos, A. A.; Amalfitano, A.; York, I. A.; Stratikos, E. Coding single nucleotide polymorphisms of endoplasmic reticulum aminopeptidase 1 can affect antigenic peptide generation in vitro by influencing basic enzymatic properties of the enzyme. J. Immunol. 2011, 186 (4), 1909– 13, DOI: 10.4049/jimmunol.100333719https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsVShurg%253D&md5=10e432675dcf5b15636f8f7de5c1a6c5Cutting Edge: Coding Single Nucleotide Polymorphisms of Endoplasmic Reticulum Aminopeptidase 1 Can Affect Antigenic Peptide Generation In Vitro by Influencing Basic Enzymatic Properties of the EnzymeEvnouchidou, Irini; Kamal, Ram P.; Seregin, Sergey S.; Goto, Yoshikuni; Tsujimoto, Masafumi; Hattori, Akira; Voulgari, Paraskevi V.; Drosos, Alexandros A.; Amalfitano, Andrea; York, Ian A.; Stratikos, EfstratiosJournal of Immunology (2011), 186 (4), 1909-1913CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)ER aminopeptidase 1 (ERAP1) customizes antigenic peptide precursors for MHC class I presentation and edits the antigenic peptide repertoire. Coding single nucleotide polymorphisms (SNPs) in ERAP1 were recently linked with predisposition to autoimmune disease, suggesting a link between pathogenesis of autoimmunity and ERAP1-mediated Ag processing. To investigate this possibility, the authors analyzed the effect that disease-linked SNPs have on Ag processing by ERAP1 in vitro. Michaelis-Menten anal. revealed that the presence of SNPs affects the Michaelis const. and turnover no. of the enzyme. Strikingly, specific ERAP1 allele-substrate combinations deviate from std. Michaelis-Menten behavior, demonstrating substrate-inhibition kinetics; to the authors' knowledge, this phenomenon has not been described for this enzyme. Cell-based Ag-presentation anal. was consistent with changes in the substrate inhibition const. Ki, further supporting that ERAP1 allelic compn. may affect Ag processing in vivo. The authors propose that these phenomena should be taken into account when evaluating the possible link between Ag processing and autoimmunity.
- 20Stamogiannos, A.; Maben, Z.; Papakyriakou, A.; Mpakali, A.; Kokkala, P.; Georgiadis, D.; Stern, L. J.; Stratikos, E. Critical Role of Interdomain Interactions in the Conformational Change and Catalytic Mechanism of Endoplasmic Reticulum Aminopeptidase 1. Biochemistry 2017, 56 (10), 1546– 1558, DOI: 10.1021/acs.biochem.6b0117020https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXivVOisrs%253D&md5=5a76edc1020984d4ce7348d80dbd8514Critical Role of Interdomain Interactions in the Conformational Change and Catalytic Mechanism of Endoplasmic Reticulum Aminopeptidase 1Stamogiannos, Athanasios; Maben, Zachary; Papakyriakou, Athanasios; Mpakali, Anastasia; Kokkala, Paraskevi; Georgiadis, Dimitris; Stern, Lawrence J.; Stratikos, EfstratiosBiochemistry (2017), 56 (10), 1546-1558CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Endoplasmic Reticulum Aminopeptidase 1 (ERAP1) is an intracellular enzyme important for the generation of antigenic epitopes and class I MHC-restricted adaptive immune responses. ERAP1 processes a vast variety of different peptides but still shows length and sequence selectivity although the mechanism behind these properties is poorly understood. X-ray crystallog. anal. has revealed that ERAP1 can assume at least two distinct conformations in which the C-terminal domain IV is either proximal or distal to the active site domain II. To help understand the role of this conformational change in the catalytic mechanism of ERAP1 we used site-directed mutagenesis to perturb key salt-bridges between domains II and IV. Enzymic anal. revealed that these mutations, although located away from the catalytic site, greatly reduce catalytic efficiency and shift allosteric kinetic behavior. The variants were more efficiently activated by small peptides, and bound a competitive inhibitor with weaker affinity and faster dissocn. kinetics. Mol. dynamics anal. suggested that the mutations affect the conformational distribution of ERAP1, reducing the population of closed states. Small-angle x-ray scattering indicated that both wild-type and the ERAP1 variants are predominantly in an open conformational state in soln. Overall, our findings suggest that electrostatic interactions between domains II and IV in ERAP1 are crucial for driving a conformational change that regulates the structural integrity of the catalytic site. The extent of domain opening in ERAP1 probably underlies its specialization for antigenic peptide precursors and should be taken into account for inhibitor development efforts.
- 21Mpakali, A.; Giastas, P.; Mathioudakis, N.; Mavridis, I. M.; Saridakis, E.; Stratikos, E. Structural Basis for Antigenic Peptide Recognition and Processing by Endoplasmic Reticulum (ER) Aminopeptidase 2. J. Biol. Chem. 2015, 290 (43), 26021– 32, DOI: 10.1074/jbc.M115.68590921https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvF2qtLbL&md5=04d68ad1491150a69457cc1eda48d498Structural basis for antigenic peptide recognition and processing by endoplasmic reticulum (ER) aminopeptidase 2Mpakali, Anastasia; Giastas, Petros; Mathioudakis, Nikolas; Mavridis, Irene M.; Saridakis, Emmanuel; Stratikos, EfstratiosJournal of Biological Chemistry (2015), 290 (43), 26021-26032CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Endoplasmic reticulum (ER) aminopeptidases process antigenic peptide precursors to generate epitopes for presentation by MHC class I mols. and help shape the antigenic peptide repertoire and cytotoxic T-cell responses. To perform this function, ER aminopeptidases have to recognize and process a vast variety of peptide sequences. To understand how these enzymes recognize substrates, we detd. crystal structures of ER aminopeptidase 2 (ERAP2) in complex with a substrate analog and a peptidic product to 2.5 and 2.7 Å, resp., and compared them to the apo-form structure detd. to 3.0 Å. The peptides were found within the internal cavity of the enzyme with no direct access to the outside solvent. The substrate analog extends away from the catalytic center toward the distal end of the internal cavity, making interactions with several shallow pockets along the path. A similar configuration was evident for the peptidic product, although decreasing electron d. toward its C terminus indicated progressive disorder. Enzymic anal. confirmed that visualized interactions can either pos. or neg. impact in vitro trimming rates. Opportunistic side-chain interactions and lack of deep specificity pockets support a limited-selectivity model for antigenic peptide processing by ERAP2. In contrast to proposed models for the homologous ERAP1, no specific recognition of the peptide C terminus by ERAP2 was evident, consistent with functional differences in length selection and self-activation between these two enzymes. Our results suggest that ERAP2 selects substrates by sequestering them in its internal cavity and allowing opportunistic interactions to det. trimming rates, thus combining substrate permissiveness with sequence bias.
- 22Mpakali, A.; Saridakis, E.; Harlos, K.; Zhao, Y.; Papakyriakou, A.; Kokkala, P.; Georgiadis, D.; Stratikos, E. Crystal Structure of Insulin-Regulated Aminopeptidase with Bound Substrate Analogue Provides Insight on Antigenic Epitope Precursor Recognition and Processing. J. Immunol. 2015, 195 (6), 2842– 2851, DOI: 10.4049/jimmunol.150110322https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVygsL7F&md5=1738ab2c66078883c93c26cc7da6c2ffCrystal Structure of Insulin-Regulated Aminopeptidase with Bound Substrate Analogue Provides Insight on Antigenic Epitope Precursor Recognition and ProcessingMpakali, Anastasia; Saridakis, Emmanuel; Harlos, Karl; Zhao, Yuguang; Papakyriakou, Athanasios; Kokkala, Paraskevi; Georgiadis, Dimitris; Stratikos, EfstratiosJournal of Immunology (2015), 195 (6), 2842-2851CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)Aminopeptidases that generate antigenic peptides influence immunodominance and adaptive cytotoxic immune responses. The mechanisms that allow these enzymes to efficiently process a vast no. of different long peptide substrates are poorly understood. In this work, we report the structure of insulin-regulated aminopeptidase, an enzyme that preps. antigenic epitopes for cross-presentation in dendritic cells, in complex with an antigenic peptide precursor analog. Insulin-regulated aminopeptidase is found in a semiclosed conformation with an extended internal cavity with limited access to the solvent. The N-terminal moiety of the peptide is located at the active site, positioned optimally for catalysis, whereas the C-terminal moiety of the peptide is stabilized along the extended internal cavity lodged between domains II and IV. Hydrophobic interactions and shape complementarity enhance peptide affinity beyond the catalytic site and support a limited selectivity model for antigenic peptide selection that may underlie the generation of complex immunopeptidomes.
- 23Saveanu, L.; Carroll, O.; Lindo, V.; Del Val, M.; Lopez, D.; Lepelletier, Y.; Greer, F.; Schomburg, L.; Fruci, D.; Niedermann, G.; van Endert, P. M. Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulum. Nat. Immunol. 2005, 6 (7), 689– 97, DOI: 10.1038/ni120823https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXltlynsro%253D&md5=bc2f4593600e4947b5e61d7c94980e93Concerted peptide trimming by human ERAP1 and ERAP2 aminopeptidase complexes in the endoplasmic reticulumSaveanu, Loredana; Carroll, Oliver; Lindo, Vivian; Del Val, Margarita; Lopez, Daniel; Lepelletier, Yves; Greer, Fiona; Schomburg, Lutz; Fruci, Doriana; Niedermann, Gabriele; van Endert, Peter M.Nature Immunology (2005), 6 (7), 689-697CODEN: NIAMCZ; ISSN:1529-2908. (Nature Publishing Group)The generation of many HLA class I peptides entails a final trimming step in the endoplasmic reticulum that, in humans, is accomplished by two 'candidate' aminopeptidases. The authors show here that one of these, ERAP1, was unable to remove several N-terminal amino acids that were trimmed efficiently by the second enzyme, ERAP2. This trimming of a longer peptide required the concerted action of both ERAP1 and ERAP2, both for in vitro digestion and in vivo for cellular antigen presentation. ERAP1 and ERAP2 localized together in vivo and assocd. phys. in complexes that were most likely heterodimeric. Thus, the human endoplasmic reticulum is equipped with a pair of trimming aminopeptidases that have complementary functions in HLA class I peptide presentation.
- 24Evnouchidou, I.; Weimershaus, M.; Saveanu, L.; van Endert, P. ERAP1-ERAP2 dimerization increases peptide-trimming efficiency. J. Immunol. 2014, 193 (2), 901– 8, DOI: 10.4049/jimmunol.130285524https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFSmtrbO&md5=9e7e734031550d119889b8c7bfc7bc78ERAP1-ERAP2 Dimerization Increases Peptide-Trimming EfficiencyEvnouchidou, Irini; Weimershaus, Mirjana; Saveanu, Loredana; van Endert, PeterJournal of Immunology (2014), 193 (2), 901-908CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)The endoplasmic reticulum aminopeptidases (ERAP)1 and ERAP2 play a crit. role in the prodn. of final epitopes presented by MHC class I mols. Formation of heterodimers by ERAP1 and ERAP2 has been proposed to facilitate trimming of epitope precursor peptides, but the effects of dimerization on ERAP function remain unknown. In this study, we produced stabilized ERAP1-ERAP2 heterodimers and found that they produced several mature epitopes more efficiently than a mix of the two enzymes unable to dimerize. Phys. interaction with ERAP2 changes basic enzymic parameters of ERAP1 and improves its substrate-binding affinity. Thus, by bringing the two enzymes in proximity and by producing allosteric effects on ERAP1, dimerization of ERAP1/2 creates complexes with superior peptide-trimming efficacy. Such complexes are likely to enhance Ag presentation by cells displaying coordinated expression of the two enzymes.
- 25Zervoudi, E.; Papakyriakou, A.; Georgiadou, D.; Evnouchidou, I.; Gajda, A.; Poreba, M.; Salvesen, G. S.; Drag, M.; Hattori, A.; Swevers, L.; Vourloumis, D.; Stratikos, E. Probing the S1 specificity pocket of the aminopeptidases that generate antigenic peptides. Biochem. J. 2011, 435 (2), 411– 20, DOI: 10.1042/BJ2010204925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXltF2gu7s%253D&md5=8c9e81b2348656d5339b27724789066aProbing the S1 specificity pocket of the aminopeptidases that generate antigenic peptidesZervoudi, Efthalia; Papakyriakou, Athanasios; Georgiadou, Dimitra; Evnouchidou, Irini; Gajda, Anna; Poreba, Marcin; Salvesen, Guy S.; Drag, Marcin; Hattori, Akira; Swevers, Luc; Vourloumis, Dionisios; Stratikos, EfstratiosBiochemical Journal (2011), 435 (2), 411-420CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)ERAP1 (endoplasmic reticulum aminopeptidase 1), ERAP2 and IRAP (insulin-regulated aminopeptidase) are three homologous enzymes that play crit. roles in the generation of antigenic peptides. These aminopeptidases excise amino acids from N-terminally extended precursors of antigenic peptides in order to generate the correct length epitopes for binding on to MHC class I mols. The specificity of these peptidases can affect antigenic peptide selection, but has not yet been investigated in detail. In the present study we utilized a collection of 82 fluorogenic substrates to define a detailed selectivity profile for each of the three enzymes and to probe structural and functional features of the S1 (primary specificity) pocket. Mol. modeling of the three S1 pockets reveals substrate-enzyme interactions that are crit. determinants for specificity. The substrate selectivity profiles suggest that IRAP largely combines the S1 specificity of ERAP1 and ERAP2, consistent with its proposed biol. function. IRAP, however, does not achieve this dual specificity by simply combining structural features of ERAP1 and ERAP2, but rather by an unique amino acid change at position 541. The results of the present study provide insights on antigenic peptide selection and may prove valuable in designing selective inhibitors or activity markers for this class of enzymes.
- 26Chang, S. C.; Momburg, F.; Bhutani, N.; Goldberg, A. L. The ER aminopeptidase, ERAP1, trims precursors to lengths of MHC class I peptides by a “molecular ruler” mechanism. Proc. Natl. Acad. Sci. U. S. A. 2005, 102 (47), 17107– 12, DOI: 10.1073/pnas.050072110226https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXht1yksLbN&md5=d7057c479987e375cd70e463c802e78bThe ER aminopeptidase, ERAP1, trims precursors to lengths of MHC class I peptides by a "molecular ruler" mechanismChang, Shih-Chung; Momburg, Frank; Bhutani, Nidhi; Goldberg, Alfred L.Proceedings of the National Academy of Sciences of the United States of America (2005), 102 (47), 17107-17112CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an IFN-γ-induced aminopeptidase in the endoplasmic reticulum that trims longer precursors to the antigenic peptides presented on MHC class I mols. The authors recently reported that purified ERAP1 trimmed N-extended precursors but spared peptides of 8-9 residues, the length required for binding to MHC class I mols. Here, the authors show another remarkable property of ERAP1: that it strongly prefers substrates 9-16 residues long, the lengths of peptides transported efficiently into the ER by the transporter assocd. with antigen processing (TAP) transporter. This aminopeptidase rapidly degraded a model 13-mer to a 9-mer and then stopped, even though the substrate and the product had identical N- and C-terminal sequences. No other aminopeptidase, including the closely related ER-aminopeptidase ERAP2, showed a similar length preference. Unlike other aminopeptidases, the activity of ERAP1 depended on the C-terminal residue of the substrate. ERAP1, like most MHC class I mols., prefers peptides with hydrophobic C termini and shows low affinity for peptides with charged C termini. Thus, ERAP1 is specialized to process precursors transported by TAP to peptides that can serve as MHC class I epitopes. Its "mol. ruler" mechanism involves binding the hydrophobic C terminus of the substrate 9-16 residues away from the active site.
- 27Lorente, E.; Barriga, A.; Johnstone, C.; Mir, C.; Jimenez, M.; Lopez, D. Concerted in vitro trimming of viral HLA-B27-restricted ligands by human ERAP1 and ERAP2 aminopeptidases. PLoS One 2013, 8 (11), e79596 DOI: 10.1371/journal.pone.007959627https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslClurnJ&md5=e6bb141f3177ecbdf63412df9a9f8e87Concerted in vitro trimming of viral HLA-B27-restricted ligands by human ERAP1 and ERAP2 aminopeptidasesLorente, Elena; Barriga, Alejandro; Johnstone, Carolina; Mir, Carmen; Jimenez, Mercedes; Lopez, DanielPLoS One (2013), 8 (11), e79596CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)In the classical human leukocyte antigen (HLA) class I antigen processing and presentation pathway, the antigenic peptides are generated from viral proteins by multiple proteolytic cleavages of the proteasome (and in some cases other cytosolic proteases) and transported to the endoplasmic reticulum (ER) lumen where they are exposed to aminopeptidase activity. In human cells, two different ER-resident enzymes, ERAP1 and ERAP2, can trim the N-terminally extended residues of peptide precursors. In this study, the possible cooperative effect of generating five naturally processed HLA-B27 ligands by both proteases was analyzed. We identified differences in the products obtained with increased detection of natural HLA-B27 ligands by comparing double vs. single enzyme digestions by mass spectrometry anal. These in vitro data suggest that each enzyme can use the degrdn. products of the other as a substrate for new N-terminal trimming, indicating concerted aminoproteolytic activity of ERAP1 and ERAP2.
- 28Hulsen, T.; de Vlieg, J.; Alkema, W. BioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics 2008, 9, 488, DOI: 10.1186/1471-2164-9-48828https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cjksF2ltQ%253D%253D&md5=1c79e2fa6accc5ff3c4cd77bab9aca8cBioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagramsHulsen Tim; de Vlieg Jacob; Alkema WynandBMC genomics (2008), 9 (), 488 ISSN:.BACKGROUND: In many genomics projects, numerous lists containing biological identifiers are produced. Often it is useful to see the overlap between different lists, enabling researchers to quickly observe similarities and differences between the data sets they are analyzing. One of the most popular methods to visualize the overlap and differences between data sets is the Venn diagram: a diagram consisting of two or more circles in which each circle corresponds to a data set, and the overlap between the circles corresponds to the overlap between the data sets. Venn diagrams are especially useful when they are 'area-proportional' i.e. the sizes of the circles and the overlaps correspond to the sizes of the data sets. Currently there are no programs available that can create area-proportional Venn diagrams connected to a wide range of biological databases. RESULTS: We designed a web application named BioVenn to summarize the overlap between two or three lists of identifiers, using area-proportional Venn diagrams. The user only needs to input these lists of identifiers in the textboxes and push the submit button. Parameters like colors and text size can be adjusted easily through the web interface. The position of the text can be adjusted by 'drag-and-drop' principle. The output Venn diagram can be shown as an SVG or PNG image embedded in the web application, or as a standalone SVG or PNG image. The latter option is useful for batch queries. Besides the Venn diagram, BioVenn outputs lists of identifiers for each of the resulting subsets. If an identifier is recognized as belonging to one of the supported biological databases, the output is linked to that database. Finally, BioVenn can map Affymetrix and EntrezGene identifiers to Ensembl genes. CONCLUSION: BioVenn is an easy-to-use web application to generate area-proportional Venn diagrams from lists of biological identifiers. It supports a wide range of identifiers from the most used biological databases currently available. Its implementation on the World Wide Web makes it available for use on any computer with internet connection, independent of operating system and without the need to install programs locally. BioVenn is freely accessible at http://www.cmbi.ru.nl/cdd/biovenn/.
- 29Sarkizova, S.; Klaeger, S.; Le, P. M.; Li, L. W.; Oliveira, G.; Keshishian, H.; Hartigan, C. R.; Zhang, W.; Braun, D. A.; Ligon, K. L.; Bachireddy, P.; Zervantonakis, I. K.; Rosenbluth, J. M.; Ouspenskaia, T.; Law, T.; Justesen, S.; Stevens, J.; Lane, W. J.; Eisenhaure, T.; Lan Zhang, G.; Clauser, K. R.; Hacohen, N.; Carr, S. A.; Wu, C. J.; Keskin, D. B. A large peptidome dataset improves HLA class I epitope prediction across most of the human population. Nat. Biotechnol. 2020, 38 (2), 199– 209, DOI: 10.1038/s41587-019-0322-929https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVSktbvP&md5=e24b305412a6cfed38e5bcd7decf6359A large peptidome dataset improves HLA class I epitope prediction across most of the human populationSarkizova, Siranush; Klaeger, Susan; Le, Phuong M.; Li, Letitia W.; Oliveira, Giacomo; Keshishian, Hasmik; Hartigan, Christina R.; Zhang, Wandi; Braun, David A.; Ligon, Keith L.; Bachireddy, Pavan; Zervantonakis, Ioannis K.; Rosenbluth, Jennifer M.; Ouspenskaia, Tamara; Law, Travis; Justesen, Sune; Stevens, Jonathan; Lane, William J.; Eisenhaure, Thomas; Lan Zhang, Guang; Clauser, Karl R.; Hacohen, Nir; Carr, Steven A.; Wu, Catherine J.; Keskin, Derin B.Nature Biotechnology (2020), 38 (2), 199-209CODEN: NABIF9; ISSN:1087-0156. (Nature Research)Prediction of HLA epitopes is important for the development of cancer immunotherapies and vaccines. However, current prediction algorithms have limited predictive power, in part because they were not trained on high-quality epitope datasets covering a broad range of HLA alleles. To enable prediction of endogenous HLA class I-assocd. peptides across a large fraction of the human population, we used mass spectrometry to profile >185,000 peptides eluted from 95 HLA-A, -B, -C and -G mono-allelic cell lines. We identified canonical peptide motifs per HLA allele, unique and shared binding submotifs across alleles and distinct motifs assocd. with different peptide lengths. By integrating these data with transcript abundance and peptide processing, we developed HLAthena, providing allele-and-length-specific and pan-allele-pan-length prediction models for endogenous peptide presentation. These models predicted endogenous HLA class I-assocd. ligands with 1.5-fold improvement in pos. predictive value compared with existing tools and correctly identified >75% of HLA-bound peptides that were obsd. exptl. in 11 patient-derived tumor cell lines.
- 30Vita, R.; Zarebski, L.; Greenbaum, J. A.; Emami, H.; Hoof, I.; Salimi, N.; Damle, R.; Sette, A.; Peters, B. The immune epitope database 2.0. Nucleic Acids Res. 2010, 38 (Database issue), D854– D862, DOI: 10.1093/nar/gkp100430https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXktl2isg%253D%253D&md5=a8502a555f1a46955251c2819097f099The Immune Epitope Database 2.0Vita, Randi; Zarebski, Laura; Greenbaum, Jason A.; Emami, Hussein; Hoof, Ilka; Salimi, Nima; Damle, Rohini; Sette, Alessandro; Peters, BjoernNucleic Acids Research (2010), 38 (Database Iss), D854-D862CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Immune Epitope Database (IEDB, www.iedb.org) provides a catalog of exptl. characterized B and T cell epitopes, as well as data on Major Histocompatibility Complex (MHC) binding and MHC ligand elution expts. The database represents the mol. structures recognized by adaptive immune receptors and the exptl. contexts in which these mols. were detd. to be immune epitopes. Epitopes recognized in humans, nonhuman primates, rodents, pigs, cats and all other tested species are included. Both pos. and neg. exptl. results are captured. Over the course of 4 years, the data from 180 978 expts. were curated manually from the literature, which covers ∼99% of all publicly available information on peptide epitopes mapped in infectious agents (excluding HIV) and 93% of those mapped in allergens. In addn., data that would otherwise be unavailable to the public from 129 186 expts. were submitted directly by investigators. The curation of epitopes related to autoimmunity is expected to be completed by the end of 2010. The database can be queried by epitope structure, source organism, MHC restriction, assay type or host organism, among other criteria. The database structure, as well as its querying, browsing and reporting interfaces, was completely redesigned for the IEDB 2.0 release, which became publicly available in early 2009.
- 31Nguyen, A.; David, J. K.; Maden, S. K.; Wood, M. A.; Weeder, B. R.; Nellore, A.; Thompson, R. F. Human leukocyte antigen susceptibility map for SARS-CoV-2. J. Virol. 2020, DOI: 10.1128/JVI.00510-20There is no corresponding record for this reference.
- 32Reynisson, B.; Alvarez, B.; Paul, S.; Peters, B.; Nielsen, M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 2020, 48 (W1), W449– W454, DOI: 10.1093/nar/gkaa37932https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXis1entr7E&md5=c777c9c3cb0858e4bef24fe6b9358244NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand dataReynisson, Birkir; Alvarez, Bruno; Paul, Sinu; Peters, Bjoern; Nielsen, MortenNucleic Acids Research (2020), 48 (W1), W449-W454CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Major histocompatibility complex (MHC) mols. are expressed on the cell surface, where they present peptides to T cells, which gives them a key role in the development of T-cell immune responses. MHC mols. come in two main variants: MHC Class I (MHC-I) and MHC Class II (MHC-II). MHC-I predominantly present peptides derived from intracellular proteins, whereas MHC-II predominantly presents peptides from extracellular proteins. In both cases, the binding between MHC and antigenic peptides is the most selective step in the antigen presentation pathway. Therefore, the prediction of peptide binding to MHC is a powerful utility to predict the possible specificity of a T-cell immune response. Commonly MHC binding prediction tools are trained on binding affinity or mass spectrometry-eluted ligands. Recent studies have however demonstrated how the integration of both data types can boost predictive performances. Inspired by this, we here present NetMHCpan-4.1 and NetMHCIIpan-4.0, two web servers created to predict binding between peptides and MHC-I and MHC-II, resp. Both methods exploit tailored machine learning strategies to integrate different training data types, resulting in state-of-the-art performance and outperforming their competitors.
- 33Komov, L.; Kadosh, D. M.; Barnea, E.; Milner, E.; Hendler, A.; Admon, A. Cell Surface MHC Class I Expression Is Limited by the Availability of Peptide-Receptive “Empty” Molecules Rather than by the Supply of Peptide Ligands. Proteomics 2018, 18 (12), e1700248 DOI: 10.1002/pmic.201700248There is no corresponding record for this reference.
- 34Hearn, A.; York, I. A.; Rock, K. L. The specificity of trimming of MHC class I-presented peptides in the endoplasmic reticulum. J. Immunol. 2009, 183 (9), 5526– 36, DOI: 10.4049/jimmunol.080366334https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1ymtb%252FK&md5=6eddb77faa4a92adbb899fd26d842406The Specificity of Trimming of MHC Class I-Presented Peptides in the Endoplasmic ReticulumHearn, Arron; York, Ian A.; Rock, Kenneth L.Journal of Immunology (2009), 183 (9), 5526-5536CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)Aminopeptidases in the endoplasmic reticulum (ER) can cleave antigenic peptides and in so doing either create or destroy MHC class I-presented epitopes. However, the specificity of this trimming process overall and of the major ER aminopeptidase ERAP1 in particular is not well understood. This issue is important because peptide trimming influences the magnitude and specificity of CD8 T cell responses. By systematically varying the N-terminal flanking sequences of peptides in a cell-free biochem. system and in intact cells, the authors elucidated the specificity of ERAP1 and of ER trimming overall. ERAP1 can cleave after many amino acids on the N terminus of epitope precursors but does so at markedly different rates. The specificity seen with purified ERAP1 is similar to that obsd. for trimming and presentation of epitopes in the ER of intact cells. The authors define N-terminal sequences that are favorable or unfavorable for Ag presentation in ways that are independent from the epitopes core sequence. When databases of known presented peptides were analyzed, the residues that were preferred for the trimming of model peptide precursors were overrepresented in N-terminal flanking sequences of epitopes generally. These data define key determinants in the specificity of Ag processing.
- 35Georgiadou, D.; Hearn, A.; Evnouchidou, I.; Chroni, A.; Leondiadis, L.; York, I. A.; Rock, K. L.; Stratikos, E. Placental leucine aminopeptidase efficiently generates mature antigenic peptides in vitro but in patterns distinct from endoplasmic reticulum aminopeptidase 1. J. Immunol. 2010, 185 (3), 1584– 92, DOI: 10.4049/jimmunol.090250235https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptV2jtr8%253D&md5=0ec50b33d7b7752368d4ed9da7f13c81Placental Leucine Aminopeptidase Efficiently Generates Mature Antigenic Peptides In Vitro but in Patterns Distinct from Endoplasmic Reticulum Aminopeptidase 1Georgiadou, Dimitra; Hearn, Arron; Evnouchidou, Irini; Chroni, Angeliki; Leondiadis, Leondios; York, Ian A.; Rock, Kenneth L.; Stratikos, EfstratiosJournal of Immunology (2010), 185 (3), 1584-1592CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)All three members of the oxytocinase subfamily of M1 aminopeptidases, endoplasmic reticulum aminopeptidase 1 (ERAP1), ERAP2, and placental leucine aminopeptidase (PLAP), also known as insulin-regulated aminopeptidase, have been implicated in the generation of MHC class I-presented peptides. ERAP1 and 2 trim peptides in the endoplasmic reticulum for direct presentation, whereas PLAP has been recently implicated in cross-presentation. The best characterized member of the family, ERAP1, has unique enzymic properties that fit well with its role in Ag processing. ERAP1 can trim a large variety of long peptide sequences and efficiently accumulate mature antigenic epitopes of 8-9 aa long. In this study, the authors evaluate the ability of PLAP to process antigenic peptide precursors in vitro and compare it with ERAP1. The authors find that, similar to ERAP1, PLAP can trim a variety of long peptide sequences efficiently and, in most cases, accumulates appreciable amts. of correct length mature antigenic epitope. Again, similar to ERAP1, PLAP continued trimming some of the epitopes tested and accumulated smaller products effectively destroying the epitope. However, the intermediate accumulation properties of ERAP1 and PLAP are distinct and epitope dependent, suggesting that these two enzymes may impose different selective pressures on epitope generation. Overall, although PLAP has the necessary enzymic properties to participate in generating or destroying MHC class I-presented peptides, its trimming behavior is distinct from that of ERAP1, something that supports a sep. role for these two enzymes in Ag processing.
- 36Kuiper, J. J.; Van Setten, J.; Ripke, S.; Van, T. S. R.; Mulder, F.; Missotten, T.; Baarsma, G. S.; Francioli, L. C.; Pulit, S. L.; De Kovel, C. G.; Ten Dam-Van Loon, N.; Den Hollander, A. I.; Huis in het Veld, P.; Hoyng, C. B.; Cordero-Coma, M.; Martin, J.; Llorenc, V.; Arya, B.; Thomas, D.; Bakker, S. C.; Ophoff, R. A.; Rothova, A.; De Bakker, P. I.; Mutis, T.; Koeleman, B. P. A genome-wide association study identifies a functional ERAP2 haplotype associated with birdshot chorioretinopathy. Hum. Mol. Genet. 2014, 23 (22), 6081– 6087, DOI: 10.1093/hmg/ddu30736https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFOrtLbI&md5=896d45e2605159f4036e0cb4e3481484A genome-wide association study identifies a functional ERAP2 haplotype associated with birdshot chorioretinopathyKuiper, Jonas J. W.; Van Setten, Jessica; Ripke, Stephan; Slot, Ruben Van 'T.; Mulder, Flip; Missotten, Tom; Baarsma, G. Seerp; Francioli, Laurent C.; Pulit, Sara L.; Kovel, Carolien G. F. De; Loon, Ninette Ten Dam-Van; Hollander, Anneke I. Den; Veld, Paulien Huis in het; Hoyng, Carel B.; Cordero-Coma, Miguel; Martin, Javier; Llorenc, Victor; Arya, Bharti; Thomas, Dhanes; Bakker, Steven C.; Ophoff, Roel A.; Rothova, Aniki; Bakker, Paul I. W. De; Mutis, Tuna; Koeleman, Bobby P. C.Human Molecular Genetics (2014), 23 (22), 6081-6087CODEN: HMGEE5; ISSN:0964-6906. (Oxford University Press)Birdshot chorioretinopathy (BSCR) is a rare form of autoimmune uveitis that can lead to severe visual impairment. Intriguingly, >95% of cases carry the HLA-A29 allele, which defines the strongest documented HLA assocn. for a human disease. We have conducted a genome-wide assocn. study in 96 Dutch and 27 Spanish cases, and 398 unrelated Dutch and 380 Spanish controls. Fine-mapping the primary MHC assocn. through high-resoln. imputation at classical HLA loci, identified HLA-A*29:02 as the principal MHC assocn. (odds ratio (OR) = 157.5, 95% CI 91.6-272.6, P = 6.6 × 10-74). We also identified two novel susceptibility loci at 5q15 near ERAP2 (rs7705093;OR = 2.3,95%CI 1.7-3.1, for the T allele, P = 8.6 × 10-8) and at 14q32.31 in the TECPR2 gene (rs150571175;OR = 6.1,95%CI 3.2-11.7, for the A allele, P = 3.2 × 10-8). The assocn. near ERAP2 was confirmed in an independent British case-control samples (combined meta-anal. P = 1.7 × 10-9). Functional analyses revealed that the risk allele of the polymorphism near ERAP2 is strongly assocd. with high mRNA and protein expression of ERAP2 in B cells. This study further defined an extremely strong MHC risk component in BSCR, and detected evidence for a novel disease mechanism that affects peptide processing in the endoplasmic reticulum.
- 37Grifoni, A.; Weiskopf, D.; Ramirez, S. I.; Mateus, J.; Dan, J. M.; Moderbacher, C. R.; Rawlings, S. A.; Sutherland, A.; Premkumar, L.; Jadi, R. S.; Marrama, D.; de Silva, A. M.; Frazier, A.; Carlin, A. F.; Greenbaum, J. A.; Peters, B.; Krammer, F.; Smith, D. M.; Crotty, S.; Sette, A. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181 (7), 1489– 1501, DOI: 10.1016/j.cell.2020.05.01537https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVOmu73N&md5=6f89bf52ae2d734af758163a4940b356Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed IndividualsGrifoni, Alba; Weiskopf, Daniela; Ramirez, Sydney I.; Mateus, Jose; Dan, Jennifer M.; Moderbacher, Carolyn Rydyznski; Rawlings, Stephen A.; Sutherland, Aaron; Premkumar, Lakshmanane; Jadi, Ramesh S.; Marrama, Daniel; de Silva, Aravinda M.; Frazier, April; Carlin, Aaron F.; Greenbaum, Jason A.; Peters, Bjoern; Krammer, Florian; Smith, Davey M.; Crotty, Shane; Sette, AlessandroCell (Cambridge, MA, United States) (2020), 181 (7), 1489-1501.e15CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide "megapools," circulating SARS-CoV-2-specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, resp. CD4+ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike, and N proteins each accounted for 11%-27% of the total CD4+ response, with addnl. responses commonly targeting nsp3, nsp4, ORF3a, and ORF8, among others. For CD8+ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2-reactive CD4+ T cells in ∼40%-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating "common cold" coronaviruses and SARS-CoV-2.
- 38Wilk, A. J.; Rustagi, A.; Zhao, N. Q.; Roque, J.; Martinez-Colon, G. J.; McKechnie, J. L.; Ivison, G. T.; Ranganath, T.; Vergara, R.; Hollis, T.; Simpson, L. J.; Grant, P.; Subramanian, A.; Rogers, A. J.; Blish, C. A. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020, 26 (7), 1070– 1076, DOI: 10.1038/s41591-020-0944-y38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFWltb%252FO&md5=93c273cbcdba588f4a980a2b0fa00460A single-cell atlas of the peripheral immune response in patients with severe COVID-19Wilk, Aaron J.; Rustagi, Arjun; Zhao, Nancy Q.; Roque, Jonasel; Martinez-Colon, Giovanny J.; McKechnie, Julia L.; Ivison, Geoffrey T.; Ranganath, Thanmayi; Vergara, Rosemary; Hollis, Taylor; Simpson, Laura J.; Grant, Philip; Subramanian, Aruna; Rogers, Angela J.; Blish, Catherine A.Nature Medicine (New York, NY, United States) (2020), 26 (7), 1070-1076CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)Abstr.: There is an urgent need to better understand the pathophysiol. of Coronavirus disease 2019 (COVID-19), the global pandemic caused by SARS-CoV-2, which has infected more than three million people worldwide1. Approx. 20% of patients with COVID-19 develop severe disease and 5% of patients require intensive care2. Severe disease has been assocd. with changes in peripheral immune activity, including increased levels of pro-inflammatory cytokines that may be produced by a subset of inflammatory monocytes , lymphopenia and T cell exhaustion . To elucidate pathways in peripheral immune cells that might lead to immunopathol. or protective immunity in severe COVID-19, we applied single-cell RNA sequencing (scRNA-seq) to profile peripheral blood mononuclear cells (PBMCs) from seven patients hospitalized for COVID-19, four of whom had acute respiratory distress syndrome, and six healthy controls. We identify reconfiguration of peripheral immune cell phenotype in COVID-19, including a heterogeneous interferon-stimulated gene signature, HLA class II downregulation and a developing neutrophil population that appears closely related to plasmablasts appearing in patients with acute respiratory failure requiring mech. ventilation. Importantly, we found that peripheral monocytes and lymphocytes do not express substantial amts. of pro-inflammatory cytokines. Collectively, we provide a cell atlas of the peripheral immune response to severe COVID-19.
- 39Wang, W.; Zhang, W.; Zhang, J.; He, J.; Zhu, F. Distribution of HLA allele frequencies in 82 Chinese individuals with coronavirus disease-2019 (COVID-19). HLA 2020, 96 (2), 194– 196, DOI: 10.1111/tan.1394139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWnur7O&md5=d15100330a68e382ff06d9790340e796Distribution of HLA allele frequencies in 82 Chinese individuals with coronavirus disease-2019 (COVID-19)Wang, Wei; Zhang, Wei; Zhang, Jingjing; He, Ji; Zhu, FamingHLA (2020), 96 (2), 194-196CODEN: HLAAAK; ISSN:2059-2310. (John Wiley & Sons Ltd.)COVID-19 is a respiratory disease caused by a novel coronavirus and is currently a global pandemic. HLA variation is assocd. with COVID-19 because HLA plays a pivotal role in the immune response to pathogens. Here, 82 individuals with COVID-19 were genotyped for HLA-A, -B, -C, -DRB1, -DRB3/4/5, -DQA1, -DQB1, -DPA1, and -DPB1 loci using next-generation sequencing (NGS). Frequencies of the HLA-C*07:29, C*08:01G, B*15:27, B*40:06, DRB1*04:06, and DPB1*36:01 alleles were higher, while the frequencies of the DRB1*12:02 and DPB1*04:01 alleles were lower in COVID-19 patients than in the control population, with uncorrected statistical significance. Only HLA-C*07:29 and B*15:27 were significant when the cor. P-value was considered. These data suggested that some HLA alleles may be assocd. with the occurrence of COVID-19.
- 40de Castro, J. A. L. How ERAP1 and ERAP2 Shape the Peptidomes of Disease-Associated MHC-I Proteins. Front. Immunol. 2018, 9, 2463, DOI: 10.3389/fimmu.2018.0246340https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXosVGmtbk%253D&md5=b76e03061a0bac1bdfa3983c25b32b6cHow ERAP1 and ERAP2 shape the peptidomes of disease-associated MHC-I proteinsde Castro, Jose A. LopezFrontiers in Immunology (2018), 9 (), 2463/1-2463/17CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)A review. Four inflammatory diseases are strongly assocd. with Major Histocompatibility Complex class I (MHC-I) mols birdshot chorioretinopathy (HLA-A*29:02), ankylosing spondylitis (HLA-B*27), Behcet's disease (HLA-B*51), and psoriasis (HLA-C*06:02). The endoplasmic reticulum aminopeptidases (ERAP) 1 and 2 are also risk factors for these diseases. Since both enzymes are involved in the final processing steps of MHC-I ligands it is reasonable to assume that MHC-I-bound peptides play a significant pathogenetic role. This review will mainly focus on recent studies concerning the effects of ERAP1 and ERAP2 polymorphism and expression on shaping the peptidome of disease-assocd. MHC-I mols. in live cells. These studies will be discussed in the context of the distinct mechanisms and substrate preferences of both enzymes, their different patterns of genetic assocn. with various diseases, the role of polymorphisms detg. changes in enzymic activity or expression levels, and the distinct peptidomes of disease-assocd. MHC-I allotypes. ERAP1 and ERAP2 polymorphism and expression induce significant changes in multiple MHC-I-bound peptidomes. These changes are MHC allotype-specific and, without excluding a degree of functional inter-dependence between both enzymes, reflect largely sep. roles in their processing of MHC-I ligands. The studies reviewed here provide a mol. basis for the distinct patterns of genetic assocn. of ERAP1 and ERAP2 with disease and for the pathogenetic role of peptides. The allotype-dependent alterations induced on distinct peptidomes may explain that the joint assocn. of both enzymes and unrelated MHC-I alleles influence different pathol. outcomes.
- 41Infantes, S.; Samino, Y.; Lorente, E.; Jimenez, M.; Garcia, R.; Del Val, M.; Lopez, D. Cutting Edge: H-2L(d) Class I Molecule Protects an HIV N-Extended Epitope from In Vitro Trimming by Endoplasmic Reticulum Aminopeptidase Associated with Antigen Processing. J. Immunol. 2010, 184 (7), 3351– 3355, DOI: 10.4049/jimmunol.090156041https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjs1Wrtr8%253D&md5=b8b6326efbcd612b287b7677c35cb4d4Cutting Edge: H-2Ld Class I Molecule Protects an HIV N-Extended Epitope from In Vitro Trimming by Endoplasmic Reticulum Aminopeptidase Associated with Antigen ProcessingInfantes, Susana; Samino, Yolanda; Lorente, Elena; Jimenez, Mercedes; Garcia, Ruth; Del Val, Margarita; Lopez, DanielJournal of Immunology (2010), 184 (7), 3351-3355CODEN: JOIMA3; ISSN:0022-1767. (American Association of Immunologists)In the classical MHC class I Ag presentation pathway, antigenic peptides derived from viral proteins by multiple proteolytic cleavages are transported to the endoplasmic reticulum lumen and are then exposed to aminopeptidase activity. In the current study, a long MHC class I natural ligand recognized by cytotoxic T lymphocytes was used to study the kinetics of degrdn. by aminopeptidase. The in vitro data indicate that this N-extended peptide is efficiently trimmed to a 9-mer, unless its binding to the MHC mols. protects the full-length peptide.
- 42Thomas, C.; Tampe, R. MHC I chaperone complexes shaping immunity. Curr. Opin. Immunol. 2019, 58, 9– 15, DOI: 10.1016/j.coi.2019.01.00142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1CltLo%253D&md5=a9963ca05f5b64cd0118525ec7b9a8c0MHC I chaperone complexes shaping immunityThomas, Christoph; Tampe, RobertCurrent Opinion in Immunology (2019), 58 (), 9-15CODEN: COPIEL; ISSN:0952-7915. (Elsevier Ltd.)Major histocompatibility complex class I (MHC I) mols. present peptides on the surface of most nucleated cells and allow the immune system to detect and eliminate infected or malignantly transformed cells. The peptides are derived from endogenous proteins by proteasomal degrdn. or aberrant translation, and are translocated from the cytosol into the endoplasmic reticulum (ER) by the transporter assocd. with antigen processing (TAP), a central component of the peptide-loading complex (PLC). The peptides are subsequently processed by ER-resident aminopeptidases (ERAP1/2) and loaded onto MHC I. This loading, however, does not happen indiscriminately: in a process called peptide editing or peptide proofreading, the MHC I-specific chaperones tapasin and TAPBPR (TAP-binding protein-related) catalyze the selection of high-affinity peptides and stable peptide-MHC I (pMHC I) complexes. Once correctly loaded with a high-affinity peptide, pMHC I complexes travel to the cell surface where they are recognized by T lymphocytes to control their differentiation in the thymus, their priming in the lymph node, and their final long-term surveillance of target cells in the periphery. Recent structural studies of the PLC and of TAPBPR-MHC I complexes by single-particle cryo-electron microscopy, X-ray crystallog., and NMR spectroscopy have provided fundamental insights into the mechanisms of MHC I peptide loading and proofreading, highlighting the dynamic nature of the involved complexes and the conformational plasticity of the individual proteins.
- 43Chen, H.; Li, L.; Weimershaus, M.; Evnouchidou, I.; van Endert, P.; Bouvier, M. ERAP1-ERAP2 dimers trim MHC I-bound precursor peptides; implications for understanding peptide editing. Sci. Rep. 2016, 6, 28902, DOI: 10.1038/srep2890243https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlCit7fP&md5=9f3e7a8e07dd2a54a0af3f78772bcd04ERAP1-ERAP2 dimers trim MHC I-bound precursor peptides; implications for understanding peptide editingChen, Hanna; Li, Lenong; Weimershaus, Mirjana; Evnouchidou, Irini; van Endert, Peter; Bouvier, MarleneScientific Reports (2016), 6 (), 28902CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)The processing of MHC class I antigenic precursor peptides by the endoplasmic reticulum aminopeptidase 1 (ERAP1) and ERAP2 is an important event in the cell biol. of antigen presentation. To date, the mol. context by which the ERAP enzymes trim precursor peptides, and how ERAPs shape peptide repertoires, remain open questions. Using ERAP1 and ERAP2 heterodimers (ERAP1/2), and N-terminally extended model and natural peptides in their free and HLA-B*0801-bound forms, we characterized the mode of action of ERAPs. We provide evidence that ERAP1/2 can trim MHC I-bound precursor peptides to their correct and final lengths, albeit more slowly than the corresponding free precursors. Trimming of MHC I-bound precursors by ERAP1/2 increases the conformational stability of MHC I/peptide complexes. From the data, we propose a mol. mechanistic model of ERAP1/2 as peptide editors. Overall, our study provides new findings on a significant issue of the ERAP-mediated processing pathway of MHC class I antigens.
- 44Mavridis, G.; Arya, R.; Domnick, A.; Zoidakis, J.; Makridakis, M.; Vlahou, A.; Mpakali, A.; Lelis, A.; Georgiadis, D.; Tampe, R.; Papakyriakou, A.; Stern, L. J.; Stratikos, E. A systematic re-examination of processing of MHCI-bound antigenic peptide precursors by endoplasmic reticulum aminopeptidase 1. J. Biol. Chem. 2020, 295 (21), 7193– 7210, DOI: 10.1074/jbc.RA120.01297644https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1Wqt7vP&md5=5385c68385264ad3cc20973aff0fbf69A systematic re-examination of processing of MHCI-bound antigenic peptide precursors by endoplasmic reticulum aminopeptidase 1Mavridis, George; Arya, Richa; Domnick, Alexander; Zoidakis, Jerome; Makridakis, Manousos; Vlahou, Antonia; Mpakali, Anastasia; Lelis, Angelos; Georgiadis, Dimitris; Tampe, Robert; Papakyriakou, Athanasios; Stern, Lawrence J.; Stratikos, EfstratiosJournal of Biological Chemistry (2020), 295 (21), 7193-7211CODEN: JBCHA3; ISSN:1083-351X. (American Society for Biochemistry and Molecular Biology)Endoplasmic reticulum aminopeptidase 1 (ERAP1) trims antigenic peptide precursors to generate mature antigenic peptides for presentation by major histocompatibility complex class I (MHCI) mols. and regulates adaptive immune responses. ERAP1 has been proposed to trim peptide precursors both in soln. and in preformed MHCI-peptide complexes, but which mode is more relevant to its biol. function remains controversial. Here, we compared ERAP1-mediated trimming of antigenic peptide precursors in soln. or when bound to three MHCI alleles, HLA-B*58, HLA-B*08, and HLA-A*02. For all MHCI-peptide combinations, peptide binding onto MHCI protected against ERAP1-mediated trimming. In only a single MHCI-peptide combination, trimming of an HLA-B*08-bound 12-mer progressed at a considerable rate, albeit still slower than in soln. Results from thermodn., kinetic, and computational analyses suggested that this 12-mer is highly labile and that apparent on-MHC trimming rates are always slower than that of MHCI-peptide dissocn. Both ERAP2 and leucine aminopeptidase, an enzyme unrelated to antigen processing, could trim this labile peptide from preformed MHCI complexes as efficiently as ERAP1. A pseudopeptide analog with high affinity for both HLA-B*08 and the ERAP1 active site could not promote the formation of a ternary ERAP1/MHCI/peptide complex. Similarly, no interactions between ERAP1 and purified peptide-loading complex were detected in the absence or presence of a pseudopeptide trap. We conclude that MHCI binding protects peptides from ERAP1 degrdn. and that trimming in soln. along with the dynamic nature of peptide binding to MHCI are sufficient to explain ERAP1 processing of antigenic peptide precursors.
- 45Perez-Riverol, Y.; Csordas, A.; Bai, J.; Bernal-Llinares, M.; Hewapathirana, S.; Kundu, D. J.; Inuganti, A.; Griss, J.; Mayer, G.; Eisenacher, M.; Perez, E.; Uszkoreit, J.; Pfeuffer, J.; Sachsenberg, T.; Yilmaz, S.; Tiwary, S.; Cox, J.; Audain, E.; Walzer, M.; Jarnuczak, A. F.; Ternent, T.; Brazma, A.; Vizcaino, J. A. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 2019, 47 (D1), D442– D450, DOI: 10.1093/nar/gky110645https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1GqtrzK&md5=4cd0f929c2df840a5cc0631d0d708d6cThe PRIDE database and related tools and resources in 2019: improving support for quantification dataPerez-Riverol, Yasset; Csordas, Attila; Bai, Jingwen; Bernal-Llinare, Manuel; Hewapathirana, Suresh; Kundu, Deepti J.; Iuganti, Avinash; Griss, Johannes; Mayer, Gerhard; Eisenacher, Martin; Perez, Enrique; Uszkoreit, Julian; Pfeuffer, Julianus; Sachsenberg, Timo; Yilmaz, Sule; Tiwary, Shivani; Cox, Jurgen; Audain, Enrique; Walzer, Mathias; Jarnuczak, Andrew F.; Ternent, Tobias; Brazma, Alvis; Vizcaino, Juan AntonioNucleic Acids Research (2019), 47 (D1), D442-D450CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The PRoteomics IDEntifications (PRIDE) database is the world's largest data repository of mass spectrometry-based proteomics data, and is one of the founding members of the global ProteomeXchange (PX) consortium. In this manuscript, we summarize the developments in PRIDE resources and related tools since the previous update manuscript was published in Nucleic Acids Research in 2016. In the last 3 years, public data sharing through PRIDE (as part of PX) has definitely become the norm in the field. In parallel, data re-use of public proteomics data has increased enormously, with multiple applications. We first describe the new architecture of PRIDE Archive, the archival component of PRIDE. PRIDE Archive and the related data submission framework have been further developed to support the increase in submitted data vols. and addnl. data types. A new scalable and fault tolerant storage backed, Application Programming Interface and web interface have been implemented, as a part of an ongoing process. Addnl., we emphasize the improved support for quant. proteomics data through the mzTab format. At last, we outline key statistics on the current data contents and vol. of downloads, and how PRIDE data are starting to be disseminated to added-value resources including Ensembl, UniProt and Expression Atlas.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jproteome.0c00457.
Table S1: List of generated peptides after digestion by each enzyme (XLSX)
Table S2: List of generated 8–11mers from each enzyme with best predicted binding score based on the HLAthena server (XLSX)
Table S3: Predicted binding score of peptides from the S1 spike glycoprotein of SARS-CoV-2 for HLA-B15:03 and HLA-B46:01, based on the HLAthena prediction server; in red, the peptides that are predicted to be binders (XLSX)
Table S4: List of potential epitopes from the sequence of the S1 spike glycoprotein of SARS-CoV-2 predicted using the NetMHCpan 4.1 server (XLSX)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.