1
|
Masuyer G, Taverner A, MacKay J, Lima Marques AR, Wang Y, Hunter T, Liu K, Mrsny RJ. Discovery of mono-ADP ribosylating toxins with high structural homology to Pseudomonas exotoxin A. Commun Biol 2025; 8:413. [PMID: 40069285 PMCID: PMC11897225 DOI: 10.1038/s42003-025-07845-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
Mono-ADP-ribosyl transferase (mART) proteins are secreted virulence factors produced by several human pathogens, the founding member being diphtheria toxin (DT). Pseudomonas aeruginosa can also secrete a mART toxin, known as exotoxin A (PE), but with an organization of its three functional domains (receptor, translocation, and enzymatic elements) that is opposite to DT. Two additional PE-like toxins (PLTs) have been identified from Vibrio cholerae and Aeromonas hydrophila, suggesting more PLT family members may exist. Database mining discovered six additional putative homologues, considerably extending this group of PLTs across a wide range of bacterial species. Here, we examine sequence and structural information for these new family members with respect to previously identified PLTs. The X-ray crystal structures of four new homologues show the conservation of critical features responsible for structure and function. This study shows the potential of these newly described toxins for the development of novel drug delivery platforms. Additionally, genomic analysis suggests horizontal gene transfer to account for the wide distribution of PLTs across a range of eubacteria species, highlighting the need to monitor emerging pathogens and their virulence factors.
Collapse
Affiliation(s)
- Geoffrey Masuyer
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
- Department of Life Sciences, University of Bath, Bath, UK.
- Centre for Therapeutic Innovation, University of Bath, Bath, UK.
| | | | - Julia MacKay
- Department of Life Sciences, University of Bath, Bath, UK
| | | | - Yuye Wang
- Department of Life Sciences, University of Bath, Bath, UK
| | - Tom Hunter
- Applied Molecular Transport Inc., South San Francisco, CA, USA
| | - Keyi Liu
- Applied Molecular Transport Inc., South San Francisco, CA, USA
| | - Randall J Mrsny
- Department of Life Sciences, University of Bath, Bath, UK.
- Centre for Therapeutic Innovation, University of Bath, Bath, UK.
| |
Collapse
|
2
|
Cruz FM, Orellano LAA, Chan A, Rock KL. Alternate MHC I Antigen Presentation Pathways Allow CD8+ T-cell Recognition and Killing of Cancer Cells in the Absence of β2M or TAP. Cancer Immunol Res 2025; 13:98-108. [PMID: 39485834 PMCID: PMC11717633 DOI: 10.1158/2326-6066.cir-24-0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 11/03/2024]
Abstract
MHC I antigen presentation allows CD8+ T cells to detect and eliminate cancerous or virally infected cells. The MHC I pathway is not essential for cell growth and viability, so cancers and viruses can evade control by CD8+ T cells by inactivating antigen presentation. In cancers, two common ways for this evasion are the loss of either the MHC I light chain [β2 microglobulin (β2M)] or the transporter-associated with antigen processing (TAP). β2M-null cells are generally thought to lack the MHC I pathway because the MHC I heavy chain by itself lacks the proper conformation for peptide display. TAP-null cells are thought to have severely defective MHC I antigen presentation because they are incapable of supplying peptides from the cytosol to MHC I molecules in the endoplasmic reticulum (ER). However, we have found that highly reactive memory CD8+ T cells could still recognize cells that completely lacked β2M or TAP. This was at least in part because in TAP-null cells, the Sec62 component of the Sec61 translocon supported the transfer of cytosolic peptides into the ER. In β2M-negative cells, free MHC I heavy chains were able to bind peptides and assume a conformation that was sufficiently recognized by CD8+ T cells. This process required ER chaperones and the peptide-loading complex. We found that these mechanisms supported antigen presentation at a level that was sufficient for memory CD8+ T cells to kill melanoma cells both in vitro and in tumor-bearing mice. The implications for tumor immunotherapy are discussed.
Collapse
Affiliation(s)
- Freidrich M Cruz
- Department of Pathology, UMass Chan Medical School, Worcester, Massachusetts
| | - Laura A A Orellano
- Department of Pathology, UMass Chan Medical School, Worcester, Massachusetts
| | - Amanda Chan
- Department of Pathology, UMass Chan Medical School, Worcester, Massachusetts
| | - Kenneth L Rock
- Department of Pathology, UMass Chan Medical School, Worcester, Massachusetts
| |
Collapse
|
3
|
Benitez Fuentes JD, Bartolome Arcilla J, Mohamed Mohamed K, Lopez de Sa A, de Luna Aguilar A, Guevara-Hoyer K, Ballestin Martinez P, Lazaro Sanchez AD, Carosella ED, Ocaña A, Sánchez-Ramon S. Targeting of Non-Classical Human Leukocyte Antigens as Novel Therapeutic Strategies in Cancer. Cancers (Basel) 2024; 16:4266. [PMID: 39766165 PMCID: PMC11675049 DOI: 10.3390/cancers16244266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Human leukocyte antigens (HLAs) are essential regulators of immune responses against cancer, with classical HLAs well-documented for their role in tumor recognition and immune surveillance. In recent years, non-classical HLAs-including HLA-E, HLA-F, HLA-G, and HLA-H-have emerged as critical players in the immune landscape of cancer due to their diverse and less conventional functions in immune modulation. These molecules exhibit unique mechanisms that enable tumors to escape immune detection, promote tumor progression, and contribute to therapeutic resistance. This review provides a comprehensive examination of the current understanding of non-classical HLAs in solid cancers, focusing on their specific roles in shaping the tumor microenvironment and influencing immune responses. By analyzing how HLA-E, HLA-F, HLA-G, and HLA-H modulate interactions with immune cells, such as T cells, natural killer cells, and antigen-presenting cells, we highlight key pathways through which these molecules contribute to immune evasion and metastasis. Additionally, we review promising therapeutic strategies aimed at targeting non-classical HLAs, including emerging immunotherapies that could potentially enhance cancer treatment outcomes by reversing immune suppression within tumors. Understanding the influence of these non-classical HLAs in solid cancers may offer new insights into cancer immunology and may lead to the development of innovative and more effective immunotherapeutic approaches. This review underscores the importance of non-classical HLAs as potential therapeutic targets, providing a necessary foundation for future studies in the evolving field of cancer immunotherapy.
Collapse
Affiliation(s)
| | - Jorge Bartolome Arcilla
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.B.A.); (A.L.d.S.); (P.B.M.)
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Kauzar Mohamed Mohamed
- Department of Immunology, IML and IdISSC, Hospital Clinico San Carlos, 28040 Madrid, Spain; (K.M.M.); (K.G.-H.); (S.S.-R.)
| | - Alfonso Lopez de Sa
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.B.A.); (A.L.d.S.); (P.B.M.)
| | - Alicia de Luna Aguilar
- Department of Medical Oncology, Hospital General Universitario Morales Meseguer, 30008 Murcia, Spain;
| | - Kissy Guevara-Hoyer
- Department of Immunology, IML and IdISSC, Hospital Clinico San Carlos, 28040 Madrid, Spain; (K.M.M.); (K.G.-H.); (S.S.-R.)
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Pablo Ballestin Martinez
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.B.A.); (A.L.d.S.); (P.B.M.)
- Department of Medical Oncology, Hospital 12 de Octubre, 28041 Madrid, Spain
| | | | - Edgardo D. Carosella
- CEA, DRF-Institut de Biologie François Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, 75010 Paris, France;
- U976 HIPI Unit, IRSL, Université Paris, 75006 Paris, France
| | - Alberto Ocaña
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.B.A.); (A.L.d.S.); (P.B.M.)
- Experimental Therapeutics in Cancer Unit, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, 28040 Madrid, Spain
| | - Silvia Sánchez-Ramon
- Department of Immunology, IML and IdISSC, Hospital Clinico San Carlos, 28040 Madrid, Spain; (K.M.M.); (K.G.-H.); (S.S.-R.)
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
4
|
Gonçalves MP, Farah R, Bikorimana JP, Abusarah J, EL-Hachem N, Saad W, Talbot S, Stanga D, Beaudoin S, Plouffe S, Rafei M. A1-reprogrammed mesenchymal stromal cells prime potent antitumoral responses. iScience 2024; 27:109248. [PMID: 38433914 PMCID: PMC10907831 DOI: 10.1016/j.isci.2024.109248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) have been modified via genetic or pharmacological engineering into potent antigen-presenting cells-like capable of priming responding CD8 T cells. In this study, our screening of a variant library of Accum molecule revealed a molecule (A1) capable of eliciting antigen cross-presentation properties in MSCs. A1-reprogrammed MSCs (ARM) exhibited improved soluble antigen uptake and processing. Our comprehensive analysis, encompassing cross-presentation assays and molecular profiling, among other cellular investigations, elucidated A1's impact on endosomal escape, reactive oxygen species production, and cytokine secretion. By evaluating ARM-based cellular vaccine in mouse models of lymphoma and melanoma, we observe significant therapeutic potency, particularly in allogeneic setting and in combination with anti-PD-1 immune checkpoint inhibitor. Overall, this study introduces a strong target for developing an antigen-adaptable vaccination platform, capable of synergizing with immune checkpoint blockers to trigger tumor regression, supporting further investigation of ARMs as an effective and versatile anti-cancer vaccine.
Collapse
Affiliation(s)
| | - Roudy Farah
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Jean-Pierre Bikorimana
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Jamilah Abusarah
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Nehme EL-Hachem
- Pediatric Hematology-Oncology Division, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, QC, Canada
| | - Wael Saad
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Sebastien Talbot
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Daniela Stanga
- Defence Therapeutics Inc., Research and Development branch, Montreal, QC, Canada
| | - Simon Beaudoin
- Defence Therapeutics Inc., Research and Development branch, Montreal, QC, Canada
| | - Sebastien Plouffe
- Defence Therapeutics Inc., Research and Development branch, Montreal, QC, Canada
| | - Moutih Rafei
- Molecular Biology Program, Université de Montréal, Montreal, QC, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
5
|
Ho NI, Huis In 't Veld LGM, van Eck van der Sluijs J, Heuts BMH, Looman MWG, Kers-Rebel ED, van den Dries K, Dolstra H, Martens JHA, Hobo W, Adema GJ. Saponin-based adjuvants enhance antigen cross-presentation in human CD11c + CD1c + CD5 - CD163 + conventional type 2 dendritic cells. J Immunother Cancer 2023; 11:e007082. [PMID: 37612044 PMCID: PMC10450066 DOI: 10.1136/jitc-2023-007082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Adjuvants are key for effective vaccination against cancer and chronic infectious diseases. Saponin-based adjuvants (SBAs) are unique among adjuvants in their ability to induce robust cell-mediated immune responses in addition to antibody responses. Recent preclinical studies revealed that SBAs induced cross-presentation and lipid bodies in otherwise poorly cross-presenting CD11b+ murine dendritic cells (DCs). METHOD Here, we investigated the response of human DC subsets to SBAs with RNA sequencing and pathway analyses, lipid body induction visualized by laser scanning microscopy, antigen translocation to the cytosol, and antigen cross-presentation to CD8+ T cells. RESULTS RNA sequencing of SBA-treated conventional type 1 DC (cDC1) and type 2 DC (cDC2) subsets uncovered that SBAs upregulated lipid-related pathways in CD11c+ CD1c+ cDC2s, especially in the CD5- CD163+ CD14+ cDC2 subset. Moreover, SBAs induced lipid bodies and enhanced endosomal antigen translocation into the cytosol in this particular cDC2 subset. Finally, SBAs enhanced cross-presentation only in cDC2s, which requires the CD163+ CD14+ cDC2 subset. CONCLUSIONS These data thus identify the CD163+ CD14+ cDC2 subset as the main SBA-responsive DC subset in humans and imply new strategies to optimize the application of saponin-based adjuvants in a potent cancer vaccine.
Collapse
Affiliation(s)
- Nataschja I Ho
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Lisa G M Huis In 't Veld
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jesper van Eck van der Sluijs
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Branco M H Heuts
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Maaike W G Looman
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Esther D Kers-Rebel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Koen van den Dries
- Radboud Technology Center Microscopy, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Nijmegen Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Apcher S, Vojtesek B, Fahraeus R. In search of the cell biology for self- versus non-self- recognition. Curr Opin Immunol 2023; 83:102334. [PMID: 37210933 DOI: 10.1016/j.coi.2023.102334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/23/2023]
Abstract
Several of today's cancer treatments are based on the immune system's capacity to detect and destroy cells expressing neoantigens on major histocompatibility class-I molecules (MHC-I). Despite this, we still do not know the cell biology behind how antigenic peptide substrates (APSs) for the MHC-I pathway are produced. Indeed, there are few research fields with so many divergent views as the one concerning the source of APSs. This is quite remarkable considering their fundamental role in the immune systems' capacity to detect and destroy virus-infected or transformed cells. A better understanding of the processes generating APSs and how these are regulated will shed light on the evolution of self-recognition and provide new targets for therapeutic intervention. We discuss the search for the elusive source of MHC-I peptides and highlight the cell biology that is still missing to explain how they are synthesised and where they come from.
Collapse
Affiliation(s)
- Sebastien Apcher
- Institut Gustave Roussy, Université Paris Sud, UMR 1015, Villejuif, France
| | - Borek Vojtesek
- RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 65653 Brno, Czech Republic
| | - Robin Fahraeus
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St. Louis, France; Department of Medical Biosciences, Building 6M, Umeå University, 901 85 Umeå, Sweden; RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 65653 Brno, Czech Republic.
| |
Collapse
|
7
|
White C, Bader C, Teter K. The manipulation of cell signaling and host cell biology by cholera toxin. Cell Signal 2022; 100:110489. [PMID: 36216164 PMCID: PMC10082135 DOI: 10.1016/j.cellsig.2022.110489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/01/2022] [Indexed: 11/03/2022]
Abstract
Vibrio cholerae colonizes the small intestine and releases cholera toxin into the extracellular space. The toxin binds to the apical surface of the epithelium, is internalized into the host endomembrane system, and escapes into the cytosol where it activates the stimulatory alpha subunit of the heterotrimeric G protein by ADP-ribosylation. This initiates a cAMP-dependent signaling pathway that stimulates chloride efflux into the gut, with diarrhea resulting from the accompanying osmotic movement of water into the intestinal lumen. G protein signaling is not the only host system manipulated by cholera toxin, however. Other cellular mechanisms and signaling pathways active in the intoxication process include endocytosis through lipid rafts, retrograde transport to the endoplasmic reticulum, the endoplasmic reticulum-associated degradation system for protein delivery to the cytosol, the unfolded protein response, and G protein de-activation through degradation or the function of ADP-ribosyl hydrolases. Although toxin-induced chloride efflux is thought to be an irreversible event, alterations to these processes could facilitate cellular recovery from intoxication. This review will highlight how cholera toxin exploits signaling pathways and other cell biology events to elicit a diarrheal response from the host.
Collapse
Affiliation(s)
- Christopher White
- Burnett School of Biomedical Sciences, 12722 Research Parkway, University of Central Florida, Orlando, FL 32826, USA.
| | - Carly Bader
- Burnett School of Biomedical Sciences, 12722 Research Parkway, University of Central Florida, Orlando, FL 32826, USA.
| | - Ken Teter
- Burnett School of Biomedical Sciences, 12722 Research Parkway, University of Central Florida, Orlando, FL 32826, USA.
| |
Collapse
|
8
|
Gros M, Segura E, Rookhuizen DC, Baudon B, Heurtebise-Chrétien S, Burgdorf N, Maurin M, Kapp EA, Simpson RJ, Kozik P, Villadangos JA, Bertrand MJM, Burbage M, Amigorena S. Endocytic membrane repair by ESCRT-III controls antigen export to the cytosol during antigen cross-presentation. Cell Rep 2022; 40:111205. [PMID: 35977488 PMCID: PMC9396532 DOI: 10.1016/j.celrep.2022.111205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/10/2022] [Accepted: 07/22/2022] [Indexed: 11/03/2022] Open
Abstract
Despite its crucial role in initiation of cytotoxic immune responses, the molecular pathways underlying antigen cross-presentation remain incompletely understood. The mechanism of antigen exit from endocytic compartments into the cytosol is a long-standing matter of controversy, confronting two main models: transfer through specific channels/transporters or rupture of endocytic membranes and leakage of luminal content. By monitoring the occurrence of intracellular damage in conventional dendritic cells (cDCs), we show that cross-presenting cDC1s display more frequent endomembrane injuries and increased recruitment of endosomal sorting complex required for transport (ESCRT)-III, the main repair system for intracellular membranes, relative to cDC2s. Silencing of CHMP2a or CHMP4b, two effector subunits of ESCRT-III, enhances cytosolic antigen export and cross-presentation. This phenotype is partially reversed by chemical inhibition of RIPK3, suggesting that endocytic damage is related to basal activation of the necroptosis pathway. Membrane repair therefore proves crucial in containing antigen export to the cytosol and cross-presentation in cDCs.
Collapse
Affiliation(s)
- Marine Gros
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France.
| | - Elodie Segura
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France; Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Biochemistry and Molecular Biology at the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Derek C Rookhuizen
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| | - Blandine Baudon
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| | | | - Nina Burgdorf
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| | - Mathieu Maurin
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| | - Eugene A Kapp
- Walter & Eliza Hall Institute of Medical Research, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC 3086, Australia
| | - Patrycja Kozik
- Protein & Nucleic Acid Chemistry Division, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Jose A Villadangos
- Department of Microbiology and Immunology at the Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Biochemistry and Molecular Biology at the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mathieu J M Bertrand
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwinjaarde 71, 9052 Zwinaarde-Ghent, Belgium; VIB Center for Inflammation Research, Technologiepark-Zwinjaarde 71, 9052 Zwinaarde-Ghent, Belgium
| | - Marianne Burbage
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France.
| | - Sebastian Amigorena
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| |
Collapse
|
9
|
Gonzales GA, Canton J. The Delivery of Extracellular “Danger” Signals to Cytosolic Sensors in Phagocytes. Front Immunol 2022; 13:944142. [PMID: 35911757 PMCID: PMC9329928 DOI: 10.3389/fimmu.2022.944142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Phagocytes, such as macrophages and dendritic cells, possess the ability to ingest large quantities of exogenous material into membrane-bound endocytic organelles such as macropinosomes and phagosomes. Typically, the ingested material, which consists of diverse macromolecules such as proteins and nucleic acids, is delivered to lysosomes where it is digested into smaller molecules like amino acids and nucleosides. These smaller molecules can then be exported out of the lysosomes by transmembrane transporters for incorporation into the cell’s metabolic pathways or for export from the cell. There are, however, exceptional instances when undigested macromolecules escape degradation and are instead delivered across the membrane of endocytic organelles into the cytosol of the phagocyte. For example, double stranded DNA, a damage associated molecular pattern shed by necrotic tumor cells, is endocytosed by phagocytes in the tumor microenvironment and delivered to the cytosol for detection by the cytosolic “danger” sensor cGAS. Other macromolecular “danger” signals including lipopolysaccharide, intact proteins, and peptidoglycans can also be actively transferred from within endocytic organelles to the cytosol. Despite the obvious biological importance of these processes, we know relatively little of how macromolecular “danger” signals are transferred across endocytic organelle membranes for detection by cytosolic sensors. Here we review the emerging evidence for the active cytosolic transfer of diverse macromolecular “danger” signals across endocytic organelle membranes. We will highlight developing trends and discuss the potential molecular mechanisms driving this emerging phenomenon.
Collapse
Affiliation(s)
- Gerone A. Gonzales
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Johnathan Canton
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- *Correspondence: Johnathan Canton,
| |
Collapse
|
10
|
Saponin-based adjuvant-induced dendritic cell cross-presentation is dependent on PERK activation. Cell Mol Life Sci 2022; 79:231. [PMID: 35396971 PMCID: PMC8994093 DOI: 10.1007/s00018-022-04253-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/22/2022]
Abstract
Saponin-based adjuvants (SBAs) are promising new adjuvants that stand out as they not only enforce CD4 + T cell-mediated immunity and antibody responses, but also induce an unprecedented level of antigen cross-presentation by dendritic cells (DC) and subsequent CD8 + T cell activation. We discovered that SBA’s ability to boost cross-presentation depends on the induction of lipid bodies (LBs). Moreover, the MHCIIloCD11bhi DC subset was identified to be most responsive to SBA-induced cross-presentation. The aim is to further unravel the mechanisms behind the induction of DC cross-presentation by SBAs. Here we show that SBAs specifically induce the PKR-like Endoplasmic Reticulum kinase (PERK) pathway and that SBA-induced DC cross-presentation is dependent on activation of the PERK pathway. PERK activation and LB formation are both crucial for SBA-induced cross-presentation and PERK inhibition has little or no effect on SBA-induced LB formation. SBA’s responsiveness, LB formation and PERK activation are specific for the MHCIIloCD11bhi DCs. These findings contribute to understanding the pathways involved in SBA-induced cross-presentation and immune activation which will ultimately lead to the development of vaccines with improved efficiency and safety.
Collapse
|
11
|
Ammon A, Mellenthin L, Emmerich C, Naschberger E, Stürzl M, Mackensen A, Müller F. Reduced cytotoxicity by mutation of Lysine 590 of Pseudomonas exotoxin can be restored in an optimized, Lysine-free immunotoxin. IMMUNOTHERAPY ADVANCES 2022; 2:ltac007. [PMID: 35919491 PMCID: PMC9327129 DOI: 10.1093/immadv/ltac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/17/2022] [Indexed: 12/02/2022] Open
Abstract
Immunotoxins, which are fusion proteins of an antibody fragment and a fragment of a bacterial or a plant toxin, induce apoptosis in target cells by inhibition of protein synthesis. ADP-ribosylating toxins often have few lysine residues in their catalytic domain. As they are the target for ubiquitination, the low number of lysines possibly prevents ubiquitin-dependent degradation of the toxin in the cytosol. To reduce this potential degradation, we aimed to generate a lysine-free (noK), Pseudomonas exotoxin (PE)-based immunotoxin. The new generation 24 kDa PE, which lacks all but the furin-cleavage site of domain II, was mutated at lysine 590 (K590) and at K606 in a CD22-targeting immunotoxin and activity was determined against various B cell malignancies in vitro and in vivo. On average, K590 mutated to arginine (R) reduced cytotoxicity by 1.3-fold and K606R enhanced cytotoxicity by 1.3-fold compared to wild type (wt). Mutating K590 to histidine or deleting K590 did not prevent this loss in cytotoxicity. Neither stability nor internalization rate of K590R could explain reduced cytotoxicity. These results highlight the relevance of lysine 590 for PE intoxication. In line with in vitro results, the K606R mutant was more than 1.8-fold more active than the other variants in vivo suggesting that this single mutation may be beneficial when targeting CD22-positive malignancies. Finally, reduced cytotoxicity by K590R was compensated for by K606R and the resulting lysine-free variant achieved wt-like activity in vitro and in vivo. Thus, PE24-noK may represent a promising candidate for down-stream applications that would interfere with lysines.
Collapse
Affiliation(s)
- Anna Ammon
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Lisa Mellenthin
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Charlotte Emmerich
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Fabian Müller
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
12
|
Mathematical modeling and stochastic simulations suggest that low-affinity peptides can bisect MHC1-mediated export of high-affinity peptides into "early"- and "late"-phases. Heliyon 2021; 7:e07466. [PMID: 34286133 PMCID: PMC8278427 DOI: 10.1016/j.heliyon.2021.e07466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/23/2021] [Accepted: 06/29/2021] [Indexed: 02/01/2023] Open
Abstract
The peptide loading complex (PLC) is a multi-protein complex of the endoplasmic reticulum (ER) which optimizes major histocompatibility I (MHC1)-mediated export of intracellular high-affinity peptides. Whilst, the molecular biology of MHC1-mediated export is well supported by empirical data, the stoichiometry, kinetics and spatio-temporal profile of the participating molecular entities are a matter of considerable debate. Here, a low-affinity peptide-driven (LAPD)-model of MHC1-mediated high-affinity peptide export is formulated, implemented, analyzed and simulated. The model is parameterized in terms of the contribution of the shunt reaction to the concentration of exportable MHC1. Theoretical analyses and simulation studies of the model suggest that low-affinity peptides can bisect MHC1-mediated export of high-affinity peptides into time-dependent distinct “early”- and “late”-phases. The net exportable MHC1 (eM1β(t)) is a function of the retrograde (rM1β(t))- and anterograde (aM1β(t))-derived fractions. The “early”-phase is dominated by the contribution of the retrograde/recyclable (rM1β≈61%,aM1β≈39%) pathway to exportable MHC1, is characterized by Tapasin-mediated peptide-editing and is ATP-independent. The “late”-phase on the other hand, is characterized by de novo PLC-assembly, rapid disassembly and a significant contribution of the anterograde pathway to exportable MHC1 (rM1β≈21%,aM1β≈79%). The shunt reaction is rate limiting and may integrate peptide translocation with PLC-assembly/disassembly thereby, regulating peptide export under physiological and pathological (viral infections, dysplastic alterations) conditions.
Collapse
|
13
|
Sicking M, Lang S, Bochen F, Roos A, Drenth JPH, Zakaria M, Zimmermann R, Linxweiler M. Complexity and Specificity of Sec61-Channelopathies: Human Diseases Affecting Gating of the Sec61 Complex. Cells 2021; 10:1036. [PMID: 33925740 PMCID: PMC8147068 DOI: 10.3390/cells10051036] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
The rough endoplasmic reticulum (ER) of nucleated human cells has crucial functions in protein biogenesis, calcium (Ca2+) homeostasis, and signal transduction. Among the roughly one hundred components, which are involved in protein import and protein folding or assembly, two components stand out: The Sec61 complex and BiP. The Sec61 complex in the ER membrane represents the major entry point for precursor polypeptides into the membrane or lumen of the ER and provides a conduit for Ca2+ ions from the ER lumen to the cytosol. The second component, the Hsp70-type molecular chaperone immunoglobulin heavy chain binding protein, short BiP, plays central roles in protein folding and assembly (hence its name), protein import, cellular Ca2+ homeostasis, and various intracellular signal transduction pathways. For the purpose of this review, we focus on these two components, their relevant allosteric effectors and on the question of how their respective functional cycles are linked in order to reconcile the apparently contradictory features of the ER membrane, selective permeability for precursor polypeptides, and impermeability for Ca2+. The key issues are that the Sec61 complex exists in two conformations: An open and a closed state that are in a dynamic equilibrium with each other, and that BiP contributes to its gating in both directions in cooperation with different co-chaperones. While the open Sec61 complex forms an aqueous polypeptide-conducting- and transiently Ca2+-permeable channel, the closed complex is impermeable even to Ca2+. Therefore, we discuss the human hereditary and tumor diseases that are linked to Sec61 channel gating, termed Sec61-channelopathies, as disturbances of selective polypeptide-impermeability and/or aberrant Ca2+-permeability.
Collapse
Affiliation(s)
- Mark Sicking
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Sven Lang
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Florian Bochen
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, D-66421 Homburg, Germany; (F.B.); (M.L.)
| | - Andreas Roos
- Department of Neuropediatrics, Essen University Hospital, D-45147 Essen, Germany;
| | - Joost P. H. Drenth
- Department of Molecular Gastroenterology and Hepatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Muhammad Zakaria
- Department of Genetics, Hazara University, Mansehra 21300, Pakistan;
| | - Richard Zimmermann
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Maximilian Linxweiler
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, D-66421 Homburg, Germany; (F.B.); (M.L.)
| |
Collapse
|
14
|
Lugo MR, Merrill AR. Development of Anti-Virulence Therapeutics against Mono-ADP-Ribosyltransferase Toxins. Toxins (Basel) 2020; 13:toxins13010016. [PMID: 33375750 PMCID: PMC7824265 DOI: 10.3390/toxins13010016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/27/2022] Open
Abstract
Mono-ADP-ribosyltransferase toxins are often key virulence factors produced by pathogenic bacteria as tools to compromise the target host cell. These toxins are enzymes that use host cellular NAD+ as the substrate to modify a critical macromolecule target in the host cell machinery. This post-translational modification of the target macromolecule (usually protein or DNA) acts like a switch to turn the target activity on or off resulting in impairment of a critical process or pathway in the host. One approach to stymie bacterial pathogens is to curtail the toxic action of these factors by designing small molecules that bind tightly to the enzyme active site and prevent catalytic function. The inactivation of these toxins/enzymes is targeted for the site of action within the host cell and small molecule therapeutics can function as anti-virulence agents by disarming the pathogen. This represents an alternative strategy to antibiotic therapy with the potential as a paradigm shift that may circumvent multi-drug resistance in the offending microbe. In this review, work that has been accomplished during the past two decades on this approach to develop anti-virulence compounds against mono-ADP-ribosyltransferase toxins will be discussed.
Collapse
|
15
|
Qin X, Denton WD, Huiting LN, Smith KS, Feng H. Unraveling the regulatory role of endoplasmic-reticulum-associated degradation in tumor immunity. Crit Rev Biochem Mol Biol 2020; 55:322-353. [PMID: 32633575 DOI: 10.1080/10409238.2020.1784085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During malignant transformation and cancer progression, tumor cells face both intrinsic and extrinsic stress, endoplasmic reticulum (ER) stress in particular. To survive and proliferate, tumor cells use multiple stress response pathways to mitigate ER stress, promoting disease aggression and treatment resistance. Among the stress response pathways is ER-associated degradation (ERAD), which consists of multiple components and steps working together to ensure protein quality and quantity. In addition to its established role in stress responses and tumor cell survival, ERAD has recently been shown to regulate tumor immunity. Here we summarize current knowledge on how ERAD promotes protein degradation, regulates immune cell development and function, participates in antigen presentation, exerts paradoxical roles on tumorigenesis and immunity, and thus impacts current cancer therapy. Collectively, ERAD is a critical protein homeostasis pathway intertwined with cancer development and tumor immunity. Of particular importance is the need to further unveil ERAD's enigmatic roles in tumor immunity to develop effective targeted and combination therapy for successful treatment of cancer.
Collapse
Affiliation(s)
- Xiaodan Qin
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - William D Denton
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - Leah N Huiting
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - Kaylee S Smith
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| | - Hui Feng
- Departments of Pharmacology and Medicine, Section of Hematology and Medical Oncology, Center for Cancer Research, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
16
|
Masuyer G. Crystal Structure of Exotoxin A from Aeromonas Pathogenic Species. Toxins (Basel) 2020; 12:toxins12060397. [PMID: 32549399 PMCID: PMC7354439 DOI: 10.3390/toxins12060397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
Aeromonas exotoxin A (AE) is a bacterial virulence factor recently discovered in a clinical case of necrotising fasciitis caused by the flesh-eating Aeromonas hydrophila. Here, database mining shows that AE is present in the genome of several emerging Aeromonas pathogenic species. The X-ray crystal structure of AE was solved at 2.3 Å and presents all the hallmarks common to diphthamide-specific mono-ADP-ribosylating toxins, suggesting AE is a fourth member of this family alongside the diphtheria toxin, Pseudomonas exotoxin A and cholix. Structural homology indicates AE may use a similar mechanism of cytotoxicity that targets eukaryotic elongation factor 2 and thus inhibition of protein synthesis. The structure of AE also highlights unique features including a metal binding site, and a negatively charged cleft that could play a role in interdomain interactions and may affect toxicity. This study raises new opportunities to engineer alternative toxin-based molecules with pharmaceutical potential.
Collapse
Affiliation(s)
- Geoffrey Masuyer
- Department of Pharmacy and Pharmacology, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
17
|
Imai J, Ohashi S, Sakai T. Endoplasmic Reticulum-Associated Degradation-Dependent Processing in Cross-Presentation and Its Potential for Dendritic Cell Vaccinations: A Review. Pharmaceutics 2020; 12:pharmaceutics12020153. [PMID: 32070016 PMCID: PMC7076524 DOI: 10.3390/pharmaceutics12020153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/14/2023] Open
Abstract
While the success of dendritic cell (DC) vaccination largely depends on cross-presentation (CP) efficiency, the precise molecular mechanism of CP is not yet characterized. Recent research revealed that endoplasmic reticulum (ER)-associated degradation (ERAD), which was first identified as part of the protein quality control system in the ER, plays a pivotal role in the processing of extracellular proteins in CP. The discovery of ERAD-dependent processing strongly suggests that the properties of extracellular antigens are one of the keys to effective DC vaccination, in addition to DC subsets and the maturation of these cells. In this review, we address recent advances in CP, focusing on the molecular mechanisms of the ERAD-dependent processing of extracellular proteins. As ERAD itself and the ERAD-dependent processing in CP share cellular machinery, enhancing the recognition of extracellular proteins, such as the ERAD substrate, by ex vivo methods may serve to improve the efficacy of DC vaccination.
Collapse
Affiliation(s)
- Jun Imai
- Correspondence: ; Tel.: +81-27-352-1180
| | | | | |
Collapse
|
18
|
Johnson BD, Geldenhuys WJ, Hazlehurst LA. The Role of ERO1α in Modulating Cancer Progression and Immune Escape. JOURNAL OF CANCER IMMUNOLOGY 2020; 2:103-115. [PMID: 33615311 PMCID: PMC7894644 DOI: 10.33696/cancerimmunol.2.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum oxidoreductin-1 alpha (ERO1α) was originally shown to be an endoplasmic reticulum (ER) resident protein undergoing oxidative cycles in concert with protein disulfide isomerase (PDI) to promote proper protein folding and to maintain homeostasis within the ER. ERO1α belongs to the flavoprotein family containing a flavin adenine dinucleotide utilized in transferring of electrons during oxidation-reduction cycles. This family is used to maintain redox potentials and protein homeostasis within the ER. ERO1α's location and function has since been shown to exist beyond the ER. Originally thought to exist solely in the ER, it has since been found to exist in the golgi apparatus, as well as in exosomes purified from patient samples. Besides aiding in protein folding of transmembrane and secretory proteins in conjunction with PDI, ERO1α is also known for formation of de novo disulfide bridges. Public databases, such as the Cancer Genome Atlas (TCGA) and The Protein Atlas, reveal ERO1α as a poor prognostic marker in multiple disease settings. Recent evidence indicates that ERO1α expression in tumor cells is a critical determinant of metastasis. However, the impact of increased ERO1α expression in tumor cells extends into the tumor microenvironment. Secretory proteins requiring ERO1α expression for proper folding have been implicated as being involved in immune escape through promotion of upregulation of programmed death ligand-1 (PD-L1) and stimulation of polymorphonuclear myeloid derived suppressor cells (PMN-MDSC's) via secretion of granulocytic colony stimulating factor (G-CSF). Hereby, ERO1α plays a pivotal role in cancer progression and potentially immune escape; making ERO1α an emerging attractive putative target for the treatment of cancer.
Collapse
Affiliation(s)
| | - Werner J. Geldenhuys
- WVU School of Pharmacy, Morgantown, WV, 25606, USA
- WVU Neuroscience Institute, Morgantown, WV, 25606, USA
| | - Lori A. Hazlehurst
- WVU Cancer Institute, Morgantown, WV 26506, USA
- WVU School of Pharmacy, Morgantown, WV, 25606, USA
| |
Collapse
|
19
|
Nowakowska-Gołacka J, Sominka H, Sowa-Rogozińska N, Słomińska-Wojewódzka M. Toxins Utilize the Endoplasmic Reticulum-Associated Protein Degradation Pathway in Their Intoxication Process. Int J Mol Sci 2019; 20:E1307. [PMID: 30875878 PMCID: PMC6471375 DOI: 10.3390/ijms20061307] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/08/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022] Open
Abstract
Several bacterial and plant AB-toxins are delivered by retrograde vesicular transport to the endoplasmic reticulum (ER), where the enzymatically active A subunit is disassembled from the holotoxin and transported to the cytosol. In this process, toxins subvert the ER-associated degradation (ERAD) pathway. ERAD is an important part of cellular regulatory mechanism that targets misfolded proteins to the ER channels, prior to their retrotranslocation to the cytosol, ubiquitination and subsequent degradation by a protein-degrading complex, the proteasome. In this article, we present an overview of current understanding of the ERAD-dependent transport of AB-toxins to the cytosol. We describe important components of ERAD and discuss their significance for toxin transport. Toxin recognition and disassembly in the ER, transport through ER translocons and finally cytosolic events that instead of overall proteasomal degradation provide proper folding and cytotoxic activity of AB-toxins are discussed as well. We also comment on recent reports presenting medical applications for toxin transport through the ER channels.
Collapse
Affiliation(s)
- Jowita Nowakowska-Gołacka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Hanna Sominka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Natalia Sowa-Rogozińska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Monika Słomińska-Wojewódzka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| |
Collapse
|
20
|
Gros M, Amigorena S. Regulation of Antigen Export to the Cytosol During Cross-Presentation. Front Immunol 2019; 10:41. [PMID: 30745902 PMCID: PMC6360170 DOI: 10.3389/fimmu.2019.00041] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/09/2019] [Indexed: 02/02/2023] Open
Abstract
Cross-priming refers to the induction of primary cytotoxic CD8+ T cell responses to antigens that are not expressed in antigen presenting cells (APCs) responsible for T cell priming. Cross-priming is achieved through cross-presentation of exogenous antigens derived from tumors, extracellular pathogens or infected neighboring cells on Major Histocompatibility Complex (MHC) class I molecules. Despite extensive research efforts to understand the intracellular pathways involved in antigen cross-presentation, certain critical steps remain elusive and controversial. Here we review recent advances on antigen cross-presentation, focusing on the mechanisms involved in antigen export to the cytosol, a crucial step of this pathway.
Collapse
|
21
|
Embgenbroich M, Burgdorf S. Current Concepts of Antigen Cross-Presentation. Front Immunol 2018; 9:1643. [PMID: 30061897 PMCID: PMC6054923 DOI: 10.3389/fimmu.2018.01643] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells have the ability to efficiently present internalized antigens on major histocompatibility complex (MHC) I molecules. This process is termed cross-presentation and is important role in the generation of an immune response against viruses and tumors, after vaccinations or in the induction of immune tolerance. The molecular mechanisms enabling cross-presentation have been topic of intense debate since many years. However, a clear view on these mechanisms remains difficult, partially due to important remaining questions, controversial results and discussions. Here, we give an overview of the current concepts of antigen cross-presentation and focus on a description of the major cross-presentation pathways, the role of retarded antigen degradation for efficient cross-presentation, the dislocation of antigens from endosomal compartment into the cytosol, the reverse transport of proteasome-derived peptides for loading on MHC I and the translocation of the cross-presentation machinery from the ER to endosomes. We try to highlight recent advances, discuss some of the controversial data and point out some of the major open questions in the field.
Collapse
Affiliation(s)
- Maria Embgenbroich
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Sven Burgdorf
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
22
|
Van Puyenbroeck V, Vermeire K. Inhibitors of protein translocation across membranes of the secretory pathway: novel antimicrobial and anticancer agents. Cell Mol Life Sci 2018; 75:1541-1558. [PMID: 29305616 PMCID: PMC5897483 DOI: 10.1007/s00018-017-2743-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/15/2017] [Accepted: 12/27/2017] [Indexed: 12/22/2022]
Abstract
Proteins routed to the secretory pathway start their journey by being transported across biological membranes, such as the endoplasmic reticulum. The essential nature of this protein translocation process has led to the evolution of several factors that specifically target the translocon and block translocation. In this review, various translocation pathways are discussed together with known inhibitors of translocation. Properties of signal peptide-specific systems are highlighted for the development of new therapeutic and antimicrobial applications, as compounds can target signal peptides from either host cells or pathogens and thereby selectively prevent translocation of those specific proteins. Broad inhibition of translocation is also an interesting target for the development of new anticancer drugs because cancer cells heavily depend on efficient protein translocation into the endoplasmic reticulum to support their fast growth.
Collapse
Affiliation(s)
- Victor Van Puyenbroeck
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000, Leuven, Belgium
| | - Kurt Vermeire
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
23
|
Abele R, Tampé R. Moving the Cellular Peptidome by Transporters. Front Cell Dev Biol 2018; 6:43. [PMID: 29761100 PMCID: PMC5937356 DOI: 10.3389/fcell.2018.00043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/03/2018] [Indexed: 12/18/2022] Open
Abstract
Living matter is defined by metastability, implying a tightly balanced synthesis and turnover of cellular components. The first step of eukaryotic protein degradation via the ubiquitin-proteasome system (UPS) leads to peptides, which are subsequently degraded to single amino acids by an armada of proteases. A small fraction of peptides, however, escapes further cytosolic destruction and is transported by ATP-binding cassette (ABC) transporters into the endoplasmic reticulum (ER) and lysosomes. The ER-resident heterodimeric transporter associated with antigen processing (TAP) is a crucial component in adaptive immunity for the transport and loading of peptides onto major histocompatibility complex class I (MHC I) molecules. Although the function of the lysosomal resident homodimeric TAPL-like (TAPL) remains, until today, only loosely defined, an involvement in immune defense is anticipated since it is highly expressed in dendritic cells and macrophages. Here, we compare the gene organization and the function of single domains of both peptide transporters. We highlight the structural organization, the modes of substrate binding and translocation as well as physiological functions of both organellar transporters.
Collapse
Affiliation(s)
- Rupert Abele
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt, Germany.,Cluster of Excellence - Macromolecular Complexes, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
24
|
Bridges RJ, Bradbury NA. Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator and Drugs: Insights from Cellular Trafficking. Handb Exp Pharmacol 2018; 245:385-425. [PMID: 29460152 DOI: 10.1007/164_2018_103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The eukaryotic cell is organized into membrane-delineated compartments that are characterized by specific cadres of proteins sustaining biochemically distinct cellular processes. The appropriate subcellular localization of proteins is key to proper organelle function and provides a physiological context for cellular processes. Disruption of normal trafficking pathways for proteins is seen in several genetic diseases, where a protein's absence for a specific subcellular compartment leads to organelle disruption, and in the context of an individual, a disruption of normal physiology. Importantly, several drug therapies can also alter protein trafficking, causing unwanted side effects. Thus, a deeper understanding of trafficking pathways needs to be appreciated as novel therapeutic modalities are proposed. Despite the promising efficacy of novel therapeutic agents, the intracellular bioavailability of these compounds has proved to be a potential barrier, leading to failures in treatments for various diseases and disorders. While endocytosis of drug moieties provides an efficient means of getting material into cells, the subsequent release and endosomal escape of materials into the cytosol where they need to act has been a barrier. An understanding of cellular protein/lipid trafficking pathways has opened up strategies for increasing drug bioavailability. Approaches to enhance endosomal exit have greatly increased the cytosolic bioavailability of drugs and will provide a means of investigating previous drugs that may have been shelved due to their low cytosolic concentration.
Collapse
Affiliation(s)
- Robert J Bridges
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA.
| |
Collapse
|
25
|
Chernukha IM, Fedulova LV, Vasilevskaya ER, Kotenkova EA. Comparative study of biocorrective protein-peptide agent to improve quality and safety of livestock products. POTRAVINARSTVO 2017. [DOI: 10.5219/590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Water with modified isotopic composition (D/H = 40 ppm) as a solubilizing agent for biologically active substances extraction from immune organs Sus Scrofa increases protein yield in thymus to 20%, in spleen up to 38%, mesenteric lymph nodes up to 35% in comparison with distilled water (D/H = 150 ppm). It was found a significant amount of neurotransmitter amino acids, such as aspartic and glutamic acids, glycine (thymus - 10.5; 13.7; 7.6%; spleen - 12.2; 10.7; 7.8% lymph nodes - 11.0; 13.3; 11.1%, respectively) having the immunological and adaptogenic activity, in extracts of Sus scrofa immunocompetent organs (thymus, spleen, mesenteric lymph nodes). Application of water with modified isotopic composition as solubilizing agent (D / H = 40 ppm) for extraction of immunocompetent organs' biologically active compounds led to increased amino acid content, including hydrophilic amino acids, in thymus extracts - up to 22%, in spleen extracts - up to 15% , in lymph nodes exctracts - up to 8%, in comparison to distilled water (D / H = 150 ppm). Peptide profile analysis revealed positive effect of water (D/H = 40 ppm) on a quality protein content extracts in molecular weight range from 10 to 20 kDa and from 30 to 45 kDa and peptide composition (2 kDa), at the same time quantitative content of compounds adaptogenic and immunocorrective action increased. Reducing of deuterium content in the solubilizing agent enhances quantitative amino acid content, i.e. extraction in an aqueous- salt solution based on deuterium depleted water of animal tissues with a high content of amino acids with hydrophilic radicals proceeded more completely.
Collapse
|
26
|
Sokolova E, Guryev E, Yudintsev A, Vodeneev V, Deyev S, Balalaeva I. HER2-specific recombinant immunotoxin 4D5scFv-PE40 passes through retrograde trafficking route and forces cells to enter apoptosis. Oncotarget 2017; 8:22048-22058. [PMID: 28423549 PMCID: PMC5400645 DOI: 10.18632/oncotarget.15833] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/06/2017] [Indexed: 01/15/2023] Open
Abstract
Immunotoxin 4D5scFv-PE40 is a recombinant protein that comprises 4D5scFv antibody as a targeting module and fragment of Pseudomonas exotoxin A as an effector (toxic) one. The immunotoxin has shown pronounced antitumor effect on cancer cells overexpressing HER2 receptor in vitro and on HER2-positive experimental tumors in vivo. We clarified the mechanism of 4D5scFv-PE40 activity that is of particular importance in the case of targeted therapeutic agent aimed at personalizing treatment of disease in relation to molecular genetic characteristics of each patient. After specific binding to HER2 on the cell surface and clathrin-mediated endocytosis the immunotoxin passes through retrograde trafficking route. During this route the immunotoxin molecule is supposed to undergo enzymatic processing that ends in separation of C-terminal and N-terminal fragments of the immunotoxin. Finally, C-terminal functionally active fragment of 4D5scFv-PE40 arrests protein synthesis in cytoplasm followed by cell death via apoptosis.
Collapse
Affiliation(s)
- Evgeniya Sokolova
- Institute of Biology and Biomedicine, Lobachevsky University, Nizhny Novgorod 603950, Russia
- Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| | - Evgeniy Guryev
- Institute of Biology and Biomedicine, Lobachevsky University, Nizhny Novgorod 603950, Russia
| | - Andrey Yudintsev
- Institute of Biology and Biomedicine, Lobachevsky University, Nizhny Novgorod 603950, Russia
| | - Vladimir Vodeneev
- Institute of Biology and Biomedicine, Lobachevsky University, Nizhny Novgorod 603950, Russia
| | - Sergey Deyev
- Institute of Biology and Biomedicine, Lobachevsky University, Nizhny Novgorod 603950, Russia
- Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| | - Irina Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky University, Nizhny Novgorod 603950, Russia
- Institute of Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
27
|
Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer. Toxins (Basel) 2016; 8:toxins8100274. [PMID: 27669301 PMCID: PMC5086635 DOI: 10.3390/toxins8100274] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer, yet no effective therapeutics exist. This review provides an overview of the recent development of recombinant immunotoxins for the treatment of glypican-3 (GPC3) expressing HCC. GPC3 is a cell surface heparan sulfate proteoglycan that is overexpressed in HCC, but is absent from normal adult human tissues. Treatment of HCC with anti-GPC3 immunotoxins represents a new therapeutic option. Using phage display and hybridoma technologies, three high affinity antibodies (HN3, HS20 and YP7) have been generated against GPC3. Two of these antibodies (HN3 and HS20) have demonstrated the ability to inhibit Wnt/Yap signaling, leading to a reduction in liver cancer cell proliferation. By combining the HN3 antibody capable of inhibiting Wnt/Yap signaling with the protein synthesis inhibitory domain of the Pseudomonas exotoxin, a recombinant immunotoxin that exhibits a dual inhibitory mechanism was generated. This immunotoxin was found to be highly effective in the treatment of human HCCs in mouse xenograft models. Engineering of the toxin fragment to reduce the level of immunogenicity is currently being explored. The development of immunotoxins provides opportunities for novel liver cancer therapies.
Collapse
|
28
|
Sokolova E, Proshkina G, Kutova O, Shilova O, Ryabova A, Schulga A, Stremovskiy O, Zdobnova T, Balalaeva I, Deyev S. Recombinant targeted toxin based on HER2-specific DARPin possesses a strong selective cytotoxic effect in vitro and a potent antitumor activity in vivo. J Control Release 2016; 233:48-56. [PMID: 27178808 DOI: 10.1016/j.jconrel.2016.05.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 01/10/2023]
Abstract
DARPins fused with other proteins are promising non-immunoglobulin scaffolds for specific binding to target cells. In this study HER2-specific DARPin (DARPin_9-29) was used as a tumor-targeting moiety for the delivery of a cytotoxic agent - the fragment of Pseudomonas aeruginosa exotoxin A. It was determined that DARPin-PE40 possesses a considerable cytotoxic activity and induces apoptosis in HER2-positive cells. Cytotoxic effect of DARPin-PE40 strongly correlates with the HER2 expression level. The effect of intravenous administration of DARPin-PE40 was tested in the xenograft model of breast cancer. It was shown that treatment of animals with DARPin-PE40 caused strong and prolonged suppression of xenograft tumor growth.
Collapse
Affiliation(s)
- Evgeniya Sokolova
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia; Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia
| | - Galina Proshkina
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Olga Kutova
- Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia
| | - Olga Shilova
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Anastasiya Ryabova
- Prokhorov General Physics Institute, Russian Academy of Sciences, 38 Vavilova St, Moscow 119991, Russia; National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 31 Kashirskoe highway, Moscow 115409, Russia
| | - Alexey Schulga
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Oleg Stremovskiy
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Tatiana Zdobnova
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia; Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia
| | - Irina Balalaeva
- Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia
| | - Sergey Deyev
- M.M. Shemyakin and Y.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia; Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia.
| |
Collapse
|
29
|
Autophagy and proteasome interconnect to coordinate cross-presentation through MHC class I pathway in B cells. Immunol Cell Biol 2016; 94:964-974. [PMID: 27297581 DOI: 10.1038/icb.2016.59] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/31/2022]
Abstract
Cross-presentation of exogenous protein antigens by B cells through the major histocompatibility complex (MHC) class I pathway in lymphoid malignancies, and transplant setting has been recognised as an important mediator of immune pathogenesis and T cell-mediated immune regulation. However, the precise mechanism of cross-presentation of exogenous antigens in B cells has remained unresolved. Here we have delineated a novel pathway for cross-presentation in B cells, which involves synergistic cooperation of the proteasome and autophagy. After endocytosis, protein antigen is processed through an autophagy- and proteasome-dependent pathway and CD8+ T-cell epitopes are loaded onto MHC class I molecules within the autophagolysomal compartment rather than the conventional secretory pathway, which requires transporters associated with antigen processing-dependent transport. Interestingly, this cross-presentation was critically dependent on valosin-containing protein (VCP)/p97 ATPase through its participation in autophagy. Loss of VCP/p97 ATPase was coincident with accumulation of LC3-II and marked reduction in antigen presentation. These observations provide unique insight on how the autophagy and proteasomal degradation systems interconnect to coordinate MHC class I-restricted cross-presentation in B cells.
Collapse
|
30
|
Michalska M, Wolf P. Pseudomonas Exotoxin A: optimized by evolution for effective killing. Front Microbiol 2015; 6:963. [PMID: 26441897 PMCID: PMC4584936 DOI: 10.3389/fmicb.2015.00963] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/31/2015] [Indexed: 11/13/2022] Open
Abstract
Pseudomonas Exotoxin A (PE) is the most toxic virulence factor of the pathogenic bacterium Pseudomonas aeruginosa. This review describes current knowledge about the intoxication pathways of PE. Moreover, PE represents a remarkable example for pathoadaptive evolution, how bacterial molecules have been structurally and functionally optimized under evolutionary pressure to effectively impair and kill their host cells.
Collapse
Affiliation(s)
- Marta Michalska
- Department of Urology, Medical Center, University of Freiburg Freiburg, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center, University of Freiburg Freiburg, Germany
| |
Collapse
|
31
|
Kalies KU, Römisch K. Inhibitors of Protein Translocation Across the ER Membrane. Traffic 2015; 16:1027-38. [PMID: 26122014 DOI: 10.1111/tra.12308] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 12/21/2022]
Abstract
Protein translocation into the endoplasmic reticulum (ER) constitutes the first step of protein secretion. ER protein import is essential in all eukaryotic cells and is particularly critical in fast-growing tumour cells. Thus, the process can serve as target both for potential cancer drugs and for bacterial virulence factors. Inhibitors of protein transport across the ER membrane range from broad-spectrum to highly substrate-specific and can interfere with virtually any stage of this multistep process, and even with transport of endocytosed antigens into the cytosol for cross-presentation.
Collapse
Affiliation(s)
- Kai-Uwe Kalies
- Institute of Biology, CSCM, University of Lübeck, Lübeck, Germany
| | - Karin Römisch
- Department of Microbiology, Faculty of Natural Sciences and Technology VIII, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
32
|
Gutiérrez-Martínez E, Planès R, Anselmi G, Reynolds M, Menezes S, Adiko AC, Saveanu L, Guermonprez P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol 2015; 6:363. [PMID: 26236315 PMCID: PMC4505393 DOI: 10.3389/fimmu.2015.00363] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/05/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have the unique ability to pick up dead cells carrying antigens in tissue and migrate to the lymph nodes where they can cross-present cell-associated antigens by MHC class I to CD8+ T cells. There is strong in vivo evidence that the mouse XCR1+ DCs subset acts as a key player in this process. The intracellular processes underlying cross-presentation remain controversial and several pathways have been proposed. Indeed, a wide number of studies have addressed the cellular process of cross-presentation in vitro using a variety of sources of antigen and antigen-presenting cells. Here, we review the in vivo and in vitro evidence supporting the current mechanistic models and disscuss their physiological relevance to the cross-presentation of cell-associated antigens by DCs subsets.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Remi Planès
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Matthew Reynolds
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Aimé Cézaire Adiko
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Loredana Saveanu
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| |
Collapse
|
33
|
Baleeiro RB, Rietscher R, Diedrich A, Czaplewska JA, Lehr CM, Scherließ R, Hanefeld A, Gottschaldt M, Walden P. Spatial separation of the processing and MHC class I loading compartments for cross-presentation of the tumor-associated antigen HER2/ neu by human dendritic cells. Oncoimmunology 2015; 4:e1047585. [PMID: 26985398 DOI: 10.1080/2162402x.2015.1047585] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/20/2015] [Accepted: 04/27/2015] [Indexed: 10/23/2022] Open
Abstract
Cross-presentation is the process by which professional antigen presenting cells (APCs) (B cells, dendritic cells (DCs) and macrophages) present endocytosed antigens (Ags) via MHC-I to CD8+ T cells. This process is crucial for induction of adaptive immune responses against tumors and infected cells. The pathways and cellular compartments involved in cross-presentation are unresolved and controversial. Among the cells with cross-presenting capacity, DCs are the most efficient, which was proposed to depend on prevention of endosomal acidification to block degradation of the epitopes. Contrary to this view, we show in this report that some cargoes induce strong endosomal acidification following uptake by human DCs, while others not. Moreover, processing of the tumor-associated antigen HER2/neu delivered in nanoparticles (NP) for cross-presentation of the epitope HER2/neu369-377 on HLA-A2 depended on endosomal acidification and cathepsin activity as well as proteasomes, and newly synthesized HLA class I. However, the HLA-A*0201/HER2/neu369-377 complexes were not found in the endoplasmic reticulum (ER) nor in endolysosomes but in hitherto not described vesicles. The data thus indicate spatial separation of antigen processing and loading of MHC-I for cross-presentation: antigen processing occurs in the uptake compartment and the cytosol whereas MHC-I loading with peptide takes place in a distinct subcellular compartment. The findings further elucidate the cellular pathways involved in the cross-presentation of a full-length, clinically relevant tumor-associated antigen by human DCs, and the impact of the vaccine formulation on antigen processing and CD8+ T cell induction.
Collapse
Affiliation(s)
- Renato B Baleeiro
- Department of Dermatology; Venerology and Allergology; Charité - Universitätsmedizin Berlin ; Berlin, Germany
| | - René Rietscher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS); Helmholtz Centre for Infection Research; Saarland University ; Saarbrücken, Germany
| | - Andrea Diedrich
- Department of Pharmaceutics and Biopharmaceutics; Kiel University ; Kiel, Germany
| | - Justyna A Czaplewska
- Laboratory of Organic and Macromolecular Chemistry (IOMC); Friedrich Schiller University Jena; Jena, Germany; Jena Center for Soft Matter (JCSM); Friedrich Schiller University Jena; Jena, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS); Helmholtz Centre for Infection Research; Saarland University; Saarbrücken, Germany; Biopharmaceutics and Pharmaceutical Technology; Saarland University; Saarbrücken, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics; Kiel University ; Kiel, Germany
| | | | - Michael Gottschaldt
- Laboratory of Organic and Macromolecular Chemistry (IOMC); Friedrich Schiller University Jena; Jena, Germany; Jena Center for Soft Matter (JCSM); Friedrich Schiller University Jena; Jena, Germany
| | - Peter Walden
- Department of Dermatology; Venerology and Allergology; Charité - Universitätsmedizin Berlin ; Berlin, Germany
| |
Collapse
|
34
|
Grotzke JE, Cresswell P. Are ERAD components involved in cross-presentation? Mol Immunol 2015; 68:112-5. [PMID: 26005101 DOI: 10.1016/j.molimm.2015.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 04/29/2015] [Accepted: 05/03/2015] [Indexed: 10/23/2022]
Abstract
A long unanswered question in the antigen presentation field is how exogenous antigens cross-presented by Major Histocompatibility Complex class I (MHC-I) molecules to CD8(+) T cells are translocated into the cytosol. Here we discuss the known mechanisms involved in this process with a focus on the hypothesized role of the machinery that functions in endoplasmic reticulum-associated degradation (ERAD). Other potential mechanisms of antigen entry to the cytosol are also discussed.
Collapse
Affiliation(s)
- Jeff E Grotzke
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Peter Cresswell
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
35
|
van de Weijer ML, Luteijn RD, Wiertz EJHJ. Viral immune evasion: Lessons in MHC class I antigen presentation. Semin Immunol 2015; 27:125-37. [PMID: 25887630 DOI: 10.1016/j.smim.2015.03.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/13/2015] [Indexed: 12/19/2022]
Abstract
The MHC class I antigen presentation pathway enables cells infected with intracellular pathogens to signal the presence of the invader to the immune system. Cytotoxic T lymphocytes are able to eliminate the infected cells through recognition of pathogen-derived peptides presented by MHC class I molecules at the cell surface. In the course of evolution, many viruses have acquired inhibitors that target essential stages of the MHC class I antigen presentation pathway. Studies on these immune evasion proteins reveal fascinating strategies used by viruses to elude the immune system. Viral immunoevasins also constitute great research tools that facilitate functional studies on the MHC class I antigen presentation pathway, allowing the investigation of less well understood routes, such as TAP-independent antigen presentation and cross-presentation of exogenous proteins. Viral immunoevasins have also helped to unravel more general cellular processes. For instance, basic principles of ER-associated protein degradation via the ubiquitin-proteasome pathway have been resolved using virus-induced degradation of MHC class I as a model. This review highlights how viral immunoevasins have increased our understanding of MHC class I-restricted antigen presentation.
Collapse
Affiliation(s)
| | - Rutger D Luteijn
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Emmanuel J H J Wiertz
- Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands.
| |
Collapse
|
36
|
Verdurmen WPR, Luginbühl M, Honegger A, Plückthun A. Efficient cell-specific uptake of binding proteins into the cytoplasm through engineered modular transport systems. J Control Release 2015; 200:13-22. [PMID: 25526701 DOI: 10.1016/j.jconrel.2014.12.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 12/13/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Abstract
Through advances in protein scaffold engineering and selection technologies, highly specific binding proteins, which fold under reducing conditions, can be generated against virtually all targets. Despite tremendous therapeutic opportunities, intracellular applications are hindered by difficulties associated with achieving cytosolic delivery, compounded by even correctly measuring it. Here, we addressed cytosolic delivery systematically through the development of a biotin ligase-based assay that objectively quantifies cytosolic delivery in a generic fashion. We developed modular transport systems that consist of a designed ankyrin repeat protein (DARPin) for receptor targeting and a different DARPin for intracellular recognition and a bacterial toxin-derived component for cytosolic translocation. We show that both anthrax pores and the translocation domain of Pseudomonas exotoxin A (ETA) efficiently deliver DARPins into the cytosol. We found that the cargo must not exceed a threshold thermodynamic stability for anthrax pores, which can be addressed by engineering, while the ETA pathway does not appear to have this restriction.
Collapse
Affiliation(s)
- Wouter P R Verdurmen
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| | - Manuel Luginbühl
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| | - Annemarie Honegger
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| | - Andreas Plückthun
- Dept of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057 Zurich, Switzerland.
| |
Collapse
|
37
|
Fischbach H, Döring M, Nikles D, Lehnert E, Baldauf C, Kalinke U, Tampé R. Ultrasensitive quantification of TAP-dependent antigen compartmentalization in scarce primary immune cell subsets. Nat Commun 2015; 6:6199. [PMID: 25656091 PMCID: PMC4347055 DOI: 10.1038/ncomms7199] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/05/2015] [Indexed: 11/09/2022] Open
Abstract
Presentation of peptides on major histocompatibility complex class I (MHC I) is essential for the establishment and maintenance of self-tolerance, priming of antigen-specific CD8(+) T cells and the exertion of several T-cell effector functions. Cytosolic proteasomes continuously degrade proteins into peptides, which are actively transported across the endoplasmic reticulum (ER) membrane by the transporter associated with antigen processing (TAP). In the ER lumen antigenic peptides are loaded onto MHC I, which is displayed on the cell surface. Here we describe an innovative flow cytometric approach to monitor time-resolved ER compartmentalization of antigenic peptides. This assay allows the analysis of distinct primary human immune cell subsets at reporter peptide concentrations of 1 nM. Thus, this ultrasensitive method for the first time permits quantification of TAP activity under close to physiological conditions in scarce primary cell subsets such as antigen cross-presenting dendritic cells.
Collapse
Affiliation(s)
- Hanna Fischbach
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue Str. 9, 60438 Frankfurt/Main, Germany
| | - Marius Döring
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz-Centre for Infection Research and the Hannover Medical School, Feodor-Lynen Str. 7-9, 30625 Hannover, Germany
| | - Daphne Nikles
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue Str. 9, 60438 Frankfurt/Main, Germany
| | - Elisa Lehnert
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue Str. 9, 60438 Frankfurt/Main, Germany
| | - Christoph Baldauf
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue Str. 9, 60438 Frankfurt/Main, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz-Centre for Infection Research and the Hannover Medical School, Feodor-Lynen Str. 7-9, 30625 Hannover, Germany
| | - Robert Tampé
- 1] Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue Str. 9, 60438 Frankfurt/Main, Germany [2] Cluster of Excellence Frankfurt - Macromolecular Complexes, Goethe-University Frankfurt, Max-von-Laue Str. 9, 60438 Frankfurt/Main, Germany
| |
Collapse
|
38
|
Lin J, Eggensperger S, Hank S, Wycisk AI, Wieneke R, Mayerhofer PU, Tampé R. A negative feedback modulator of antigen processing evolved from a frameshift in the cowpox virus genome. PLoS Pathog 2014; 10:e1004554. [PMID: 25503639 PMCID: PMC4263761 DOI: 10.1371/journal.ppat.1004554] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 11/04/2014] [Indexed: 12/31/2022] Open
Abstract
Coevolution of viruses and their hosts represents a dynamic molecular battle between the immune system and viral factors that mediate immune evasion. After the abandonment of smallpox vaccination, cowpox virus infections are an emerging zoonotic health threat, especially for immunocompromised patients. Here we delineate the mechanistic basis of how cowpox viral CPXV012 interferes with MHC class I antigen processing. This type II membrane protein inhibits the coreTAP complex at the step after peptide binding and peptide-induced conformational change, in blocking ATP binding and hydrolysis. Distinct from other immune evasion mechanisms, TAP inhibition is mediated by a short ER-lumenal fragment of CPXV012, which results from a frameshift in the cowpox virus genome. Tethered to the ER membrane, this fragment mimics a high ER-lumenal peptide concentration, thus provoking a trans-inhibition of antigen translocation as supply for MHC I loading. These findings illuminate the evolution of viral immune modulators and the basis of a fine-balanced regulation of antigen processing. Virus-infected or malignant transformed cells are eliminated by cytotoxic T lymphocytes, which recognize antigenic peptide epitopes in complex with major histocompatibility complex class I (MHC I) molecules at the cell surface. The majority of such peptides are derived from proteasomal degradation in the cytosol and are then translocated into the ER lumen in an energy-consuming reaction via the transporter associated with antigen processing (TAP), which delivers the peptides onto MHC I molecules as final acceptors. Viruses have evolved sophisticated strategies to escape this immune surveillance. Here we show that the cowpox viral protein CPXV012 inhibits the ER peptide translocation machinery by allosterically blocking ATP binding and hydrolysis by TAP. The short ER resident active domain of the viral protein evolved from a reading frame shift in the cowpox virus genome and exploits the ER-lumenal negative feedback peptide sensor of TAP. This CPXV012-induced conformational arrest of TAP is signaled by a unique communication across the ER membrane to the cytosolic motor domains of the peptide pump. Furthermore, this study provides the rare opportunity to decipher on a molecular level how nature plays hide and seek with a pathogen and its host.
Collapse
Affiliation(s)
- Jiacheng Lin
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
| | - Sabine Eggensperger
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
| | - Susanne Hank
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
| | - Agnes I. Wycisk
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
| | - Peter U. Mayerhofer
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
- * E-mail: (PUM); (RT)
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe-University Frankfurt, Frankfurt, Germany
- Cluster of Excellence – Macromolecular Complexes, Goethe-University Frankfurt, Frankfurt, Germany
- * E-mail: (PUM); (RT)
| |
Collapse
|
39
|
Mechanistic determinants of the directionality and energetics of active export by a heterodimeric ABC transporter. Nat Commun 2014; 5:5419. [PMID: 25377891 PMCID: PMC4242082 DOI: 10.1038/ncomms6419] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/29/2014] [Indexed: 11/09/2022] Open
Abstract
The ATP-binding cassette (ABC) transporter associated with antigen processing (TAP) participates in immune surveillance by moving proteasomal products into the endoplasmic reticulum (ER) lumen for major histocompatibility complex class I loading and cell surface presentation to cytotoxic T cells. Here we delineate the mechanistic basis for antigen translocation. Notably, TAP works as a molecular diode, translocating peptide substrates against the gradient in a strict unidirectional way. We reveal the importance of the D-loop at the dimer interface of the two nucleotide-binding domains (NBDs) in coupling substrate translocation with ATP hydrolysis and defining transport vectoriality. Substitution of the conserved aspartate, which coordinates the ATP-binding site, decreases NBD dimerization affinity and turns the unidirectional primary active pump into a passive bidirectional nucleotide-gated facilitator. Thus, ATP hydrolysis is not required for translocation per se, but is essential for both active and unidirectional transport. Our data provide detailed mechanistic insight into how heterodimeric ABC exporters operate.
Collapse
|
40
|
Antignani A, Sarnovsky R, FitzGerald DJ. ABT-737 promotes the dislocation of ER luminal proteins to the cytosol, including pseudomonas exotoxin. Mol Cancer Ther 2014; 13:1655-63. [PMID: 24739394 DOI: 10.1158/1535-7163.mct-13-0998] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Impaired apoptosis is often a key element in tumor development. Therefore, drugs mimicking prosurvival antagonists offer promise as cancer therapeutics. When ABT-737, a BH3-only mimetic, was added to KB3-1 human cervical adenocarcinoma cells, we noted an induction of an endoplasmic reticulum (ER) stress response and the dislocation of ER luminal proteins, including chaperones, to the cell cytosol. Furthermore, when immunotoxin (antibody-toxin chimeric molecule) and ABT-737 combinations were added to cells, there was enhanced toxin-mediated inhibition of protein synthesis, consistent with enhanced translocation of toxin to the cytosol. A similar enhancement was not seen with thapsigargin, suggesting that ER stress alone was not responsible for enhanced translocation. Cytosol preparations from ABT-737-treated but not from thapsigargin-treated cells revealed the presence of greater amounts of processed 37-kDa toxin fragment compared with the addition of immunotoxin alone. As early as 4 hours after the addition of ABT-737 and immunotoxin, there was release of mitochondrial cytochrome c and activation of caspase-3/7 indicating that the combination caused apoptotic cell death. These results were reflected in decreased cellular ATP levels that were noted with combinations of ABT-737 and immunotoxin but not with either agent alone or with combinations of thapsigargin and immunotoxin. We conclude that ABT-737 increases ER permeability, promoting the dislocation of toxin from the ER to the cytosol resulting in early apoptotic cell death. These mechanistic insights suggest why this class of BH3-only mimetic synergizes in a particular way with Pseudomonas exotoxin-based immunotoxins.
Collapse
Affiliation(s)
- Antonella Antignani
- Authors' Affiliation: Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Robert Sarnovsky
- Authors' Affiliation: Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - David J FitzGerald
- Authors' Affiliation: Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
41
|
The ins-and-outs of endosomal antigens for cross-presentation. Curr Opin Immunol 2014; 26:63-8. [DOI: 10.1016/j.coi.2013.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 10/30/2013] [Accepted: 11/06/2013] [Indexed: 01/28/2023]
|
42
|
Sachamitr P, Fairchild PJ. Cross presentation of antigen by dendritic cells: mechanisms and implications for immunotherapy. Expert Rev Clin Immunol 2014; 8:547-55. [DOI: 10.1586/eci.12.45] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Teter K. Toxin instability and its role in toxin translocation from the endoplasmic reticulum to the cytosol. Biomolecules 2013; 3:997-1029. [PMID: 24970201 PMCID: PMC4030972 DOI: 10.3390/biom3040997] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/26/2013] [Accepted: 11/27/2013] [Indexed: 12/21/2022] Open
Abstract
AB toxins enter a host cell by receptor-mediated endocytosis. The catalytic A chain then crosses the endosome or endoplasmic reticulum (ER) membrane to reach its cytosolic target. Dissociation of the A chain from the cell-binding B chain occurs before or during translocation to the cytosol, and only the A chain enters the cytosol. In some cases, AB subunit dissociation is facilitated by the unique physiology and function of the ER. The A chains of these ER-translocating toxins are stable within the architecture of the AB holotoxin, but toxin disassembly results in spontaneous or assisted unfolding of the isolated A chain. This unfolding event places the A chain in a translocation-competent conformation that promotes its export to the cytosol through the quality control mechanism of ER-associated degradation. A lack of lysine residues for ubiquitin conjugation protects the exported A chain from degradation by the ubiquitin-proteasome system, and an interaction with host factors allows the cytosolic toxin to regain a folded, active state. The intrinsic instability of the toxin A chain thus influences multiple steps of the intoxication process. This review will focus on the host-toxin interactions involved with A chain unfolding in the ER and A chain refolding in the cytosol.
Collapse
Affiliation(s)
- Ken Teter
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 12722 Research Parkway, Orlando, FL 32826, USA.
| |
Collapse
|
44
|
van Kasteren SI, Overkleeft H, Ovaa H, Neefjes J. Chemical biology of antigen presentation by MHC molecules. Curr Opin Immunol 2013; 26:21-31. [PMID: 24556397 DOI: 10.1016/j.coi.2013.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 10/03/2013] [Indexed: 11/25/2022]
Abstract
MHC class I and MHC class II molecules present peptides to the immune system to drive proper T cell responses. Pharmacological modulation of T-cell responses can offer treatment options for a range of immune-related diseases. Pharmacological downregulation of MHC molecules may find application in treatment of auto-immunity and transplantation rejection while pharmacological activation of antigen presentation would support immune responses to infection and cancer. Since the cell biology of MHC class I and MHC class II antigen presentation is understood in great detail, many potential targets for manipulation have been defined over the years. Here, we discuss how antigen presentation by MHC molecules can be modulated by pharmacological agents and how chemistry can further support the study of antigen presentation in general. The chemical biology of antigen presentation by MHC molecules shows surprising options for immune modulation and the development of future therapies.
Collapse
Affiliation(s)
- Sander I van Kasteren
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands
| | - Hermen Overkleeft
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands
| | - Huib Ovaa
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands
| | - Jacques Neefjes
- The Netherlands Cancer Institute, Division of Cell Biology, Plesmanlaan 121, Amsterdam, Netherlands; Leiden Institute of Chemistry (LIC), Division of Bio-Organic Chemistry, Einsteinweg 55, Leiden, Netherlands.
| |
Collapse
|
45
|
Schäuble N, Cavalié A, Zimmermann R, Jung M. Interaction of Pseudomonas aeruginosa Exotoxin A with the human Sec61 complex suppresses passive calcium efflux from the endoplasmic reticulum. Channels (Austin) 2013; 8:76-83. [PMID: 24088629 PMCID: PMC4048345 DOI: 10.4161/chan.26526] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 11/19/2022] Open
Abstract
According to live-cell calcium-imaging experiments, the Sec61 complex is a passive calcium-leak channel in the human endoplasmic reticulum (ER) membrane that is regulated by ER luminal immunoglobulin heavy chain binding protein (BiP) and cytosolic Ca(2+)-calmodulin. In single channel measurements, the open Sec61 complex is Ca(2+) permeable. It can be closed not only by interaction with BiP or Ca(2+)-calmodulin, but also with Pseudomonas aeruginosa Exotoxin A which can enter human cells by retrograde transport. Exotoxin A has been shown to interact with the Sec61 complex and, thereby, inhibit ER export of immunogenic peptides into the cytosol. Here, we show that Exotoxin A also inhibits passive Ca(2+) leakage from the ER in human cells, and we characterized the N-terminus of the Sec61 α-subunit as the relevant binding site for Exotoxin A.
Collapse
Affiliation(s)
- Nico Schäuble
- Medical Biochemistry and Molecular Biology; Saarland University; Homburg, Germany
| | - Adolfo Cavalié
- Experimental and Clinical Pharmacology and Toxicology; Saarland University; Homburg, Germany
| | - Richard Zimmermann
- Medical Biochemistry and Molecular Biology; Saarland University; Homburg, Germany
| | - Martin Jung
- Medical Biochemistry and Molecular Biology; Saarland University; Homburg, Germany
| |
Collapse
|
46
|
Assaying peptide translocation by the peptide transporter TAP. Methods Mol Biol 2013. [PMID: 23329478 DOI: 10.1007/978-1-62703-218-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
MHC class I molecules display peptides at the cell surface that are mostly derived from cytosolic or nuclear proteins. Since peptide loading of MHC class I molecules occurs in the ER lumen, cytosolic peptides have to pass the ER membrane. The peptide transporter TAP translocates peptides over this ER membrane which is critical for successful MHC class I antigen presentation. How peptide translocation by TAP can be assayed and inhibitors of chemical or viral origin can be identified, will be described here.
Collapse
|
47
|
Crespo MI, Zacca ER, Núñez NG, Ranocchia RP, Maccioni M, Maletto BA, Pistoresi-Palencia MC, Morón G. TLR7 triggering with polyuridylic acid promotes cross-presentation in CD8α+ conventional dendritic cells by enhancing antigen preservation and MHC class I antigen permanence on the dendritic cell surface. THE JOURNAL OF IMMUNOLOGY 2013; 190:948-60. [PMID: 23284054 DOI: 10.4049/jimmunol.1102725] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
ssRNA can interact with dendritic cells (DCs) through binding to TLR7, inducing secretion of proinflammatory cytokines and type I IFN. Triggering TLR7 enhances cross-priming of CD8(+) T cells, which requires cross-presentation of exogenous Ag to DCs. However, how TLR triggering can affect Ag cross-presentation is still not clear. Using OVA as an Ag model, we observed that stimulation of TLR7 in DCs by polyuridylic acid (polyU), a synthetic ssRNA analog, generates a strong specific cytotoxic response in C57BL/6 mice. PolyU stimulate CD8α(+) DCs to cross-prime naive CD8(+) T cells in a type I IFN-dependent fashion. This enhanced cross-priming is accompanied by a higher density of OVA(256-264)/H-2K(b) complexes on CD8α(+) DCs treated with polyU, as well as by upregulation of costimulatory molecules and increased secretion of proinflammatory cytokines by DCs. Cross-priming of CD8(+) T cells by DCs treated with polyU requires proteasome and Ag translocation to cytosol through the Sec61 channel in DCs. The observed enhancement in OVA cross-presentation with polyU in DCs could be mediated by a limited Ag degradation in endophagosomal compartments and a higher permanence of OVA peptide/MHC class I complexes on DCs. These observations clearly reveal that key steps of Ag processing for cross-presentation can be modulated by TLR ligands, opening new avenues for understanding their mechanisms as adjuvants of the immune response.
Collapse
Affiliation(s)
- María I Crespo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Behar SM. Antigen-specific CD8(+) T cells and protective immunity to tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:141-63. [PMID: 23468108 DOI: 10.1007/978-1-4614-6111-1_8] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The continuing HIV/AIDS epidemic and the spread of multi-drug resistant Mycobacterium tuberculosis has led to the perpetuation of the worldwide tuberculosis epidemic. While M. bovis BCG is widely used as a vaccine, it lacks efficacy in preventing pulmonary tuberculosis in adults [1]. To combat this ongoing scourge, vaccine development for tuberculosis is a global priority. Most infected individuals develop long-lived protective immunity, which controls and contains M. tuberculosis in a T cell-dependent manner. An effective T cells response determines whether the infection resolves or develops into clinically evident disease. Consequently, there is great interest in determining which T cells subsets mediate anti-mycobacterial immunity, delineating their effector functions, and evaluating whether vaccination can elicit these T cells subsets and induce protective immunity. CD4(+) T cells are critical for resistance to M. tuberculosis in both humans and rodent models. CD4(+) T cells are required to control the initial infection as well as to prevent recrudescence in both humans and mice [2]. While it is generally accepted that class II MHC-restricted CD4(+) T cells are essential for immunity to tuberculosis, M. tuberculosis infection elicits CD8(+) T cells responses in both people and in experimental animals. CD8(+) T cells are also recruited to the lung during M. tuberculosis infection and are found in the granulomas of infected people. Thus, how CD8(+) T cells contribute to overall immunity to tuberculosis and whether antigens recognized by CD8(+) T cells would enhance the efficacy of vaccine strategies continue to be important questions.
Collapse
Affiliation(s)
- Samuel M Behar
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
49
|
Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev 2012; 92:537-76. [PMID: 22535891 DOI: 10.1152/physrev.00027.2011] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Protein folding is a complex, error-prone process that often results in an irreparable protein by-product. These by-products can be recognized by cellular quality control machineries and targeted for proteasome-dependent degradation. The folding of proteins in the secretory pathway adds another layer to the protein folding "problem," as the endoplasmic reticulum maintains a unique chemical environment within the cell. In fact, a growing number of diseases are attributed to defects in secretory protein folding, and many of these by-products are targeted for a process known as endoplasmic reticulum-associated degradation (ERAD). Since its discovery, research on the mechanisms underlying the ERAD pathway has provided new insights into how ERAD contributes to human health during both normal and diseases states. Links between ERAD and disease are evidenced from the loss of protein function as a result of degradation, chronic cellular stress when ERAD fails to keep up with misfolded protein production, and the ability of some pathogens to coopt the ERAD pathway. The growing number of ERAD substrates has also illuminated the differences in the machineries used to recognize and degrade a vast array of potential clients for this pathway. Despite all that is known about ERAD, many questions remain, and new paradigms will likely emerge. Clearly, the key to successful disease treatment lies within defining the molecular details of the ERAD pathway and in understanding how this conserved pathway selects and degrades an innumerable cast of substrates.
Collapse
Affiliation(s)
- Christopher J Guerriero
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
50
|
Wagner CS, Grotzke JE, Cresswell P. Intracellular events regulating cross-presentation. Front Immunol 2012; 3:138. [PMID: 22675326 PMCID: PMC3366438 DOI: 10.3389/fimmu.2012.00138] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 05/14/2012] [Indexed: 01/07/2023] Open
Abstract
Cross-presentation plays a fundamental role in the induction of CD8-T cell immunity. However, although more than three decades have passed since its discovery, surprisingly little is known about the exact mechanisms involved. Here we give an overview of the components involved at different stages of this process. First, antigens must be internalized into the cross-presenting cell. The involvement of different receptors, method of antigen uptake, and nature of the antigen can influence intracellular trafficking and access to the cross-presentation pathway. Once antigens access the endocytic system, different requirements for endosomal/phagosomal processing arise, such as proteolysis and reduction of disulfide bonds. The majority of cross-presented peptides are generated by proteasomal degradation. Therefore, antigens must cross a membrane barrier in a manner analogous to the fate of misfolded proteins in the endoplasmic reticulum (ER) that are retrotranslocated into the cytosol for degradation. Indeed, some components of the ER-associated degradation machinery have been implicated in cross-presentation. Further complicating the matter, endosomal and phagosomal compartments have been suggested as alternative sites to the ER for loading of peptides on major histocompatibility complex class I molecules. Finally, the antigen presenting cells involved, particularly dendritic cell subsets and their state of maturation, influence the efficiency of cross-presentation.
Collapse
Affiliation(s)
- Claudia S Wagner
- Department of Immunobiology, Yale University Medical Center, New Haven, CT, USA
| | | | | |
Collapse
|