1
|
Gomaa B, Abdelhamed H, Banes M, Zinnurine S, Pinchuk L, Lawrence ML. Innate and adaptive immunity gene expression profiles induced by virulent Aeromonas hydrophila infection in the immune-related organs of channel catfish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 162:105276. [PMID: 39341476 DOI: 10.1016/j.dci.2024.105276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Aeromonas hydrophila causes motile Aeromonas septicemia (MAS) in freshwater fish. In recent years, MAS outbreaks due to virulent Aeromonas hydrophila (vAh) have been responsible for large-scale losses within commercial catfish farms in Mississippi and Alabama. The aim of this study was to evaluate immune gene expression in catfish immune-competent tissues during infection with vAh strain ML09-119. Specific pathogen-free catfish fingerlings were intraperitoneally infected with vAh strain ML09-119, and relative expression of thirteen immune-related genes was evaluated from head kidney, spleen, and liver. Our results revealed that vAh was detected 2 h post-infection (hpi) in the head kidney, liver, and spleen. The highest concentration of vAh was detected at 12 hpi, from which point concentrations decreased until clearance at 5 days post-infection (dpi). Gene expression analysis revealed upregulation of pro-inflammatory cytokines and innate immune response (TLR 4 and 5) in the first 24 hpi. Adaptive immune-related genes were upregulated at 7 dpi in the spleen and 14 dpi in the head kidney. Furthermore, immunoglobulin M showed significant upregulation at 14 dpi in the head kidney and 21 dpi in the spleen. In summary, vAh ML09-119 infection induced a strong inflammatory response involving multiple innate immunity genes, proinflammatory cytokines, and chemokines. Surviving catfish were able to clear the infection and produce antibodies and memory cells. Assessment of the immunological response to vAh infection is critical for understanding the pathogen's mechanisms of pathogenesis and developing means for MAS control, including vaccine development and improved treatments.
Collapse
Affiliation(s)
- Basant Gomaa
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Hossam Abdelhamed
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Michelle Banes
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Saida Zinnurine
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Lesya Pinchuk
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Mark L Lawrence
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, MS, 39762, USA.
| |
Collapse
|
2
|
Herbst CH, Bouteau A, Menykő EJ, Qin Z, Gyenge E, Su Q, Cooper V, Mabbott NA, Igyártó BZ. Dendritic cells overcome Cre/Lox induced gene deficiency by siphoning cytosolic material from surrounding cells. iScience 2024; 27:109119. [PMID: 38384841 PMCID: PMC10879714 DOI: 10.1016/j.isci.2024.109119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/10/2024] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
In a previous report, keratinocytes were shown to share their gene expression profile with surrounding Langerhans cells (LCs), influencing LC biology. Here, we investigated whether transferred material could substitute for lost gene products in cells subjected to Cre/Lox conditional gene deletion. We found that in human Langerin-Cre mice, epidermal LCs and CD11b+CD103+ mesenteric DCs overcome gene deletion if the deleted gene was expressed by neighboring cells. The mechanism of material transfer differed from traditional antigen uptake routes, relying on calcium and PI3K, being susceptible to polyguanylic acid inhibition, and remaining unaffected by inflammation. Termed intracellular monitoring, this process was specific to DCs, occurring in all murine DC subsets tested and human monocyte-derived DCs. The transferred material was presented on MHC-I and MHC-II, suggesting a role in regulating immune responses.
Collapse
Affiliation(s)
- Christopher H Herbst
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aurélie Bouteau
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Evelin J Menykő
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Zhen Qin
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ervin Gyenge
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Qingtai Su
- OncoNano Medicine, Inc, Southlake, TX 76092, USA
| | - Vincent Cooper
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Botond Z Igyártó
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Lellahi SM, Azeem W, Hua Y, Gabriel B, Paulsen Rye K, Reikvam H, Kalland KH. GM-CSF, Flt3-L and IL-4 affect viability and function of conventional dendritic cell types 1 and 2. Front Immunol 2023; 13:1058963. [PMID: 36713392 PMCID: PMC9880532 DOI: 10.3389/fimmu.2022.1058963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Conventional type 1 dendritic cells (cDC1) and conventional type 2 dendritic cells (cDC2) have attracted increasing attention as alternatives to monocyte-derived dendritic cells (moDCs) in cancer immunotherapy. Use of cDCs for therapy has been hindered by their low numbers in peripheral blood. In the present study, we found that extensive spontaneous apoptosis and cDC death in culture within 24hrs represent an additional challenge. Different media conditions that maintain cDC viability and function were investigated. CD141+ cDC1 and CD1c+ cDC2 were isolated from healthy blood donor buffy coats. Low viabilities were found with CellGenix DC, RPMI-1640, and X-VIVO 15 standard culture media and with several supplements at 24hrs and 48hrs. Among multiple factors it was found that GM-CSF improved both cDC1 and cDC2 viability, whereas Flt3-L and IL-4 only increased viability of cDC1 and cDC2, respectively. Combinations of these three cytokines improved viability of both cDCs further, both at 24hrs and 48hrs time points. Although these cytokines have been extensively investigated for their role in myeloid cell differentiation, and are also used clinically, their effects on mature cDCs remain incompletely known, in particular effects on pro-inflammatory or tolerogenic cDC features. HLA-DR, CD80, CD83, CD86, PD-L1 and PD-L2 cDC membrane expressions were relatively little affected by GM-CSF, IL-4 and Flt3-L cytokine supplements compared to the strong induction following Toll-like receptor (TLR) stimulation for 24hrs. With minor exceptions the three cytokines appeared to be permissive to the TLR-induced marker expression. Allogeneic mixed leukocyte reaction showed that the cytokines promoted T-cell proliferation and revealed a potential to boost both Th1 and Th2 polarizing cytokines. GM-CSF and Flt3-L and their combination improved the capability of cDC1 for dextran uptake, while in cDC2, dextran capture was improved by GM-CSF. The data suggest that GM-CSF, IL-4 and Flt3-L and combinations might be beneficial for DC viability and function in vitro. Limited viability of cDCs could be a confounding variable experimentally and in immunotherapy.
Collapse
Affiliation(s)
- Seyed Mohammad Lellahi
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Waqas Azeem
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Immunology and Transfusion Medicine, Helse Bergen, Bergen, Norway
| | - Yaping Hua
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Benjamin Gabriel
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Håkon Reikvam
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Karl-Henning Kalland
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
| |
Collapse
|
4
|
Blagovic K, Smith CK, Ramakrishnan A, Moore L, Soto DR, Thompson Z, Stockmann AP, Kruszelnicki S, Thakkar A, Murray J, Torres S, Wondimagegnhu B, Yi R, Dadgar M, Paracha AM, Page C, Clear L, Chaudhry OA, Myint M, Bridgen DT, Gilbert JB, Seidl KJ, Sharei A, Loughhead S, Bernstein H, Yarar D. Engineered red blood cells (activating antigen carriers) drive potent T cell responses and tumor regression in mice. Front Immunol 2022; 13:1015585. [PMID: 36263022 PMCID: PMC9573954 DOI: 10.3389/fimmu.2022.1015585] [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: 08/09/2022] [Accepted: 09/14/2022] [Indexed: 11/22/2022] Open
Abstract
Activation of T cell responses is essential for effective tumor clearance; however, inducing targeted, potent antigen presentation to stimulate T cell responses remains challenging. We generated Activating Antigen Carriers (AACs) by engineering red blood cells (RBCs) to encapsulate relevant tumor antigens and the adjuvant polyinosinic-polycytidylic acid (poly I:C), for use as a tumor-specific cancer vaccine. The processing method and conditions used to create the AACs promote phosphatidylserine exposure on RBCs and thus harness the natural process of aged RBC clearance to enable targeting of the AACs to endogenous professional antigen presenting cells (APCs) without the use of chemicals or viral vectors. AAC uptake, antigen processing, and presentation by APCs drive antigen-specific activation of T cells, both in mouse in vivo and human in vitro systems, promoting polyfunctionality of CD8+ T cells and, in a tumor model, driving high levels of antigen-specific CD8+ T cell infiltration and tumor killing. The efficacy of AAC therapy was further enhanced by combination with the chemotherapeutic agent Cisplatin. In summary, these findings support AACs as a potential vector-free immunotherapy strategy to enable potent antigen presentation and T cell stimulation by endogenous APCs with broad therapeutic potential.
Collapse
|
5
|
A novel platform for the production of autologous human antibodies. Anticancer Drugs 2022; 33:903-912. [PMID: 36136990 DOI: 10.1097/cad.0000000000001380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
At Research Genetic Cancer Centre, we have developed a novel method for the production of human monoclonal antibodies against a specific antigen of our choice (c-met) using isolated human blood cells. By mimicking nature, dendritic, CD4 and CD19 cells from healthy volunteers were driven towards Th2 immunity. Cell activation was succeeded by a cytokine cocktail, and IgG production was promoted by IgG class switching factors. IgG secretion was determined using both enzyme linked immunosorbent assay (ELISA) and Western blot as well as immunoglobulin heavy chain gamma polypeptide gene expression. Secreted antibody was further purified by affinity column chromatography against c-met peptide. Anti-c-met activity was determined using the purified antibody as primary antibody for c-met detection by ELISA, Western blot and flow cytometry. Finally, anti-c-met antibody efficiency was determined by MCF-7 viability assay. Plasma cell formation and IgG secretion took place after 6 days of culture. Plasma cells produced anti-c-met IgG antibody that significantly decreased MCF-7 breast cancer cell proliferation. To our knowledge, this is the first platform of its kind, generating fully human antibodies-on-demand using patient's own cells, bringing personalized, targeted therapy for cancer one step closer.
Collapse
|
6
|
Alotaibi S, Ozkan J, Papas E, Markoulli M. Diurnal Variation of Corneal Dendritic Cell Density. Curr Eye Res 2022; 47:1239-1245. [PMID: 35726825 DOI: 10.1080/02713683.2022.2088799] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Purpose: To measure variation in corneal dendritic cell density, and percentage of mature to total dendritic cells, in healthy individuals during the sleep/wake cycle.Methods: Using in vivo confocal microscopy, images of the subbasal nerve plexus were captured from 19 healthy, noncontact lens wearing participants. The central cornea and inferior whorl were imaged three times (midday, before sleep, upon awakening). Dendritic cell counts from the images were categorized according to perceived maturity (immature vs mature). Dendritic cell density and percentage of mature to total cells were compared between time points.Result: The median and interquartile range (IQR) of total dendritic cell density in the central cornea was 32.0 (7.0-131.3) cells/mm2 at midday, 37.1 (8.2-103.9) cells/mm2 before sleep, and 19.5 (7.0-83.2) cells/mm2 on awakening. Corresponding values for immature cells were 28.1 (5.8-112.5) cells/mm2, 22.3 (7.4-84.0) cells/mm2 and 18.0 (2.9-64.8) cells/mm2, and for mature cells, 3.1 (0.0-6.6) cells/mm2, 2.0 (0.8-16.8) cells/mm2, and 1.6 (0.2-8.2) cells/mm2. At the inferior whorl, total dendritic cell density was 38.5 (18.4-84.5) cells/mm2, 34.4 (9.4-82.3) cell/mm2, and 32.3 (15.2-96.1) cells/mm2. Immature cell density was 32.8 (18.4-80.9) cells/mm2, 34.4 (8.6-81.0) cells/mm2, and 32.3 (12.6-78.5) cells/mm2. Mature cell density was 1.6 (0.0-6.3) cells/mm2, 1.6 (0.0-3.1) cells/mm2, and 1.8 (0.0-6.3) cells/mm2. There was no significant difference between time points for total cell density (p > 0.05), but the percentage of mature cells upon awakening was significantly greater, compared to midday, at the central cornea (p = 0.02).Conclusion: In healthy individuals, overall corneal dendritic cell density is reasonably constant during the sleep/wake cycle, but the relative number of mature cells tends to increase overnight.
Collapse
Affiliation(s)
- Sultan Alotaibi
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia.,Department of Optometry and Vision Science, College of Applied Medical Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Jerome Ozkan
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Eric Papas
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Maria Markoulli
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
7
|
Montesinos CA, Khalid R, Cristea O, Greenberger JS, Epperly MW, Lemon JA, Boreham DR, Popov D, Gorthi G, Ramkumar N, Jones JA. Space Radiation Protection Countermeasures in Microgravity and Planetary Exploration. Life (Basel) 2021; 11:life11080829. [PMID: 34440577 PMCID: PMC8398261 DOI: 10.3390/life11080829] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Space radiation is one of the principal environmental factors limiting the human tolerance for space travel, and therefore a primary risk in need of mitigation strategies to enable crewed exploration of the solar system. METHODS We summarize the current state of knowledge regarding potential means to reduce the biological effects of space radiation. New countermeasure strategies for exploration-class missions are proposed, based on recent advances in nutrition, pharmacologic, and immune science. RESULTS Radiation protection can be categorized into (1) exposure-limiting: shielding and mission duration; (2) countermeasures: radioprotectors, radiomodulators, radiomitigators, and immune-modulation, and; (3) treatment and supportive care for the effects of radiation. Vehicle and mission design can augment the overall exposure. Testing in terrestrial laboratories and earth-based exposure facilities, as well as on the International Space Station (ISS), has demonstrated that dietary and pharmacologic countermeasures can be safe and effective. Immune system modulators are less robustly tested but show promise. Therapies for radiation prodromal syndrome may include pharmacologic agents; and autologous marrow for acute radiation syndrome (ARS). CONCLUSIONS Current radiation protection technology is not yet optimized, but nevertheless offers substantial protection to crews based on Lunar or Mars design reference missions. With additional research and human testing, the space radiation risk can be further mitigated to allow for long-duration exploration of the solar system.
Collapse
Affiliation(s)
| | - Radina Khalid
- School of Engineering, Rice University, Houston, TX 77005, USA;
| | - Octav Cristea
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA; (J.S.G.); (M.W.E.)
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA; (J.S.G.); (M.W.E.)
| | - Jennifer A. Lemon
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; (J.A.L.); (D.R.B.)
| | - Douglas R. Boreham
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; (J.A.L.); (D.R.B.)
| | - Dmitri Popov
- Advanced Medical Technologies and Systems Inc., Richmond Hill, ON L4B 1N1, Canada;
| | | | - Nandita Ramkumar
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeffrey A. Jones
- Center for Space Medicine, Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
8
|
Brandum EP, Jørgensen AS, Rosenkilde MM, Hjortø GM. Dendritic Cells and CCR7 Expression: An Important Factor for Autoimmune Diseases, Chronic Inflammation, and Cancer. Int J Mol Sci 2021; 22:ijms22158340. [PMID: 34361107 PMCID: PMC8348795 DOI: 10.3390/ijms22158340] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
Chemotactic cytokines-chemokines-control immune cell migration in the process of initiation and resolution of inflammatory conditions as part of the body's defense system. Many chemokines also participate in pathological processes leading up to and exacerbating the inflammatory state characterizing chronic inflammatory diseases. In this review, we discuss the role of dendritic cells (DCs) and the central chemokine receptor CCR7 in the initiation and sustainment of selected chronic inflammatory diseases: multiple sclerosis (MS), rheumatoid arthritis (RA), and psoriasis. We revisit the binary role that CCR7 plays in combatting and progressing cancer, and we discuss how CCR7 and DCs can be harnessed for the treatment of cancer. To provide the necessary background, we review the differential roles of the natural ligands of CCR7, CCL19, and CCL21 and how they direct the mobilization of activated DCs to lymphoid organs and control the formation of associated lymphoid tissues (ALTs). We provide an overview of DC subsets and, briefly, elaborate on the different T-cell effector types generated upon DC-T cell priming. In the conclusion, we promote CCR7 as a possible target of future drugs with an antagonistic effect to reduce inflammation in chronic inflammatory diseases and an agonistic effect for boosting the reactivation of the immune system against cancer in cell-based and/or immune checkpoint inhibitor (ICI)-based anti-cancer therapy.
Collapse
|
9
|
Barbet G, Nair-Gupta P, Schotsaert M, Yeung ST, Moretti J, Seyffer F, Metreveli G, Gardner T, Choi A, Tortorella D, Tampé R, Khanna KM, García-Sastre A, Blander JM. TAP dysfunction in dendritic cells enables noncanonical cross-presentation for T cell priming. Nat Immunol 2021; 22:497-509. [PMID: 33790474 PMCID: PMC8981674 DOI: 10.1038/s41590-021-00903-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/22/2021] [Indexed: 02/01/2023]
Abstract
Classic major histocompatibility complex class I (MHC-I) presentation relies on shuttling cytosolic peptides into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP). Viruses disable TAP to block MHC-I presentation and evade cytotoxic CD8+ T cells. Priming CD8+ T cells against these viruses is thought to rely solely on cross-presentation by uninfected TAP-functional dendritic cells. We found that protective CD8+ T cells could be mobilized during viral infection even when TAP was absent in all hematopoietic cells. TAP blockade depleted the endosomal recycling compartment of MHC-I molecules and, as such, impaired Toll-like receptor-regulated cross-presentation. Instead, MHC-I molecules accumulated in the ER-Golgi intermediate compartment (ERGIC), sequestered away from Toll-like receptor control, and coopted ER-SNARE Sec22b-mediated vesicular traffic to intersect with internalized antigen and rescue cross-presentation. Thus, when classic MHC-I presentation and endosomal recycling compartment-dependent cross-presentation are impaired in dendritic cells, cell-autonomous noncanonical cross-presentation relying on ERGIC-derived MHC-I counters TAP dysfunction to nevertheless mediate CD8+ T cell priming.
Collapse
Affiliation(s)
- Gaëtan Barbet
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- The Child Health Institute of New Jersey, and Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Priyanka Nair-Gupta
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Janssen Research and Development LLC, Spring House, PA, USA
| | - Michael Schotsaert
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen T Yeung
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Infectious Disease, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Julien Moretti
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Fabian Seyffer
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Giorgi Metreveli
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Gardner
- Molecular Pharmacology and Chemistry Program, Sloan Kettering Institute, New York, NY, USA
- ArsenalBio, San Francisco, CA, USA
| | - Angela Choi
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Moderna Inc., Cambridge, MA, USA
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kamal M Khanna
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Sadiq BA, Mantel I, Blander JM. A Comprehensive Experimental Guide to Studying Cross-Presentation in Dendritic Cells In Vitro. CURRENT PROTOCOLS IN IMMUNOLOGY 2020; 131:e115. [PMID: 33316130 PMCID: PMC9060150 DOI: 10.1002/cpim.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cross-presentation was first observed serendipitously in the 1970s. The importance of it was quickly realized and subsequently attracted great attention from immunologists. Since then, our knowledge of the ability of certain antigen presenting cells to internalize, process, and load exogenous antigens onto MHC-I molecules to cross-prime CD8+ T cells has increased significantly. Dendritic cells (DCs) are exceptional cross-presenters, thus making them a great tool to study cross-presentation but the relative rarity of DCs in circulation and in tissues makes it challenging to isolate sufficient numbers of cells to study this process in vitro. In this paper, we describe in detail two methods to culture DCs from bone-marrow progenitors and a method to expand the numbers of DCs present in vivo as a source of endogenous bona-fide cross-presenting DCs. We also describe methods to assess cross-presentation by DCs using the activation of primary CD8+ T cells as a readout. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Isolation of bone marrow progenitor cells Basic Protocol 2: In vitro differentiation of dendritic cells with GM-CSF Support Protocol 1: Preparation of conditioned medium from GM-CSF producing J558L cells Basic Protocol 3: In vitro differentiation of dendritic cells with Flt3L Support Protocol 2: Preparation of Flt3L containing medium from B16-Flt3L cells Basic Protocol 4: Expansion of cDC1s in vivo for use in ex vivo experiments Basic Protocol 5: Characterizing resting and activated dendritic cells Basic Protocol 6: Dendritic cell stimulation, antigenic cargo, and fixation Support Protocol 3: Preparation of model antigen coated microbeads Support Protocol 4: Preparation of apoptotic cells Support Protocol 5: Preparation of recombinant bacteria Basic Protocol 7: Immunocytochemistry immunofluorescence (ICC/IF) Support Protocol 6: Preparation of Alcian blue-coated coverslips Basic Protocol 8: CD8+ T cell activation to assess cross-presentation Support Protocol 7: Isolation and labeling of CD8+ T cells with CFSE.
Collapse
Affiliation(s)
- Barzan A. Sadiq
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
| | - Ian Mantel
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, New York
| | - J. Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, New York
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, New York
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, New York
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, New York
| |
Collapse
|
11
|
Sabatino D. Medicinal Chemistry and Methodological Advances in the Development of Peptide-Based Vaccines. J Med Chem 2020; 63:14184-14196. [PMID: 32990437 DOI: 10.1021/acs.jmedchem.0c00848] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The evolution of rapidly proliferating infectious and tumorigenic diseases has resulted in an urgent need to develop new and improved intervention strategies. Among the many therapeutic strategies at our disposal, our immune system remains the gold-standard in disease prevention, diagnosis, and treatment. Vaccines have played an important role in eradicating or mitigating the spread of infectious diseases by bolstering our immunity. Despite their utility, the design and development of new, more effective vaccines remains a public health necessity. Peptide-based vaccines have been developed for a wide range of established and emerging infectious and tumorigenic diseases. New innovations in epitope design and selection, synthesis, and formulation as well as screening techniques against immunological targets have led to more effective peptide vaccines. Current and future work is geared toward the translation of peptide vaccines from preclinical to clinical utility.
Collapse
Affiliation(s)
- David Sabatino
- Department of Chemistry and Biochemistry, Seton Hall University, South Orange, New Jersey 07079, United States
| |
Collapse
|
12
|
Nguyen TL, Yin Y, Choi Y, Jeong JH, Kim J. Enhanced Cancer DNA Vaccine via Direct Transfection to Host Dendritic Cells Recruited in Injectable Scaffolds. ACS NANO 2020; 14:11623-11636. [PMID: 32808762 DOI: 10.1021/acsnano.0c04188] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Deoxyribonucleic acid (DNA) vaccines are a promising cancer immunotherapy approach. However, effective delivery of DNA to antigen-presenting cells (e.g., dendritic cells (DCs)) for the induction of an adaptive immune response is limited. Conventional DNA delivery via intramuscular, intradermal, and subcutaneous injection by hypodermal needles shows a low potency and immunogenicity. Here, we propose the enhanced cancer DNA vaccine by direct transfection to the high number of DCs recruited into the chemoattractant-loaded injectable mesoporous silica microrods (MSRs). Subcutaneous administration of the MSRs mixed with tumor-antigen coding DNA polyplexes resulted in DC recruitment in the macroporous space of the scaffold formed by the spontaneous assembly of high-aspect-ratio MSRs, thereby allowing for enhanced cellular uptake of antigen-coded DNA by host DCs. The MSR scaffolds delivering the DNA vaccine trigger a more robust DC activation, antigen-specific CD8+ T cell response, and Th1 immune response compared to the bolus DNA vaccine. Additionally, the immunological memory can be induced with a single administration of the vaccine. The combination of the vaccination and antiprogrammed cell death-1 antibody significantly eliminates established lung metastasis. These results indicate that MSRs serve as a powerful platform for DNA vaccine delivery to DCs for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Yue Yin
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Youngjin Choi
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
13
|
miR-223-3p Inhibits Antigen Endocytosis and Presentation and Promotes the Tolerogenic Potential of Dendritic Cells through Targeting Mannose Receptor Signaling and Rhob. J Immunol Res 2020; 2020:1379458. [PMID: 32656268 PMCID: PMC7320286 DOI: 10.1155/2020/1379458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background The role of miR-223-3p in dendritic cells (DCs) is unknown. This study is aimed at investigating the effect of miR-223-3p on the antigen uptake and presentation capacities of DCs and the underlying molecular mechanism. Methods FITC-OVA antigen uptake and cell surface markers in bone marrow-derived DCs (BMDCs) were analyzed by flow cytometry. BMDCs were transfected with the miR-223-3p mimic or inhibitor. Cytokine levels were determined by ELISA. CD4+ T cell differentiation was determined by mixed lymphocyte culture assay. Results OVA treatment significantly downregulated miR-223-3p in BMDCs. The miR-223-3p mimic significantly inhibited OVA-induced antigen uptake and surface expression of MHC-II on BMDCs (P < 0.01). The miR-223-3p mimic increased TGF-β1 production in OVA-treated DCs (P < 0.01). Mixed lymphocyte reaction showed that the miR-223-3p mimic significantly promoted Treg cell differentiation. In addition, the miR-223-3p mimic significantly upregulated CD103 in DCs, indicating the promotion of tolerogenic DCs. The miR-223-3p mimic downregulated Rhob protein in OVA-induced DCs. Rhob knockdown significantly suppressed the ability of FITC-OVA endocytosis (P < 0.01) and surface MHC-II molecule expression (P < 0.01) in BMDCs, promoting promoted Treg cell differentiation. Mannose receptor (MR) knockdown significantly upregulated miR-223-3p, downregulated Rhob protein in OVA-treated DCs, inhibited the FITC-OVA endocytosis and surface MHC-II expression in BMDCs, and promoted Treg cell differentiation (all P < 0.01). Conclusion These data suggest that miR-223-3p has an inhibitory effect on the antigen uptake and presentation capacities of BMDCs and promotes Treg cell differentiation, which is, at least partially, through targeting MR signaling and Rhob.
Collapse
|
14
|
Machhi J, Kevadiya BD, Muhammad IK, Herskovitz J, Olson KE, Mosley RL, Gendelman HE. Harnessing regulatory T cell neuroprotective activities for treatment of neurodegenerative disorders. Mol Neurodegener 2020; 15:32. [PMID: 32503641 PMCID: PMC7275301 DOI: 10.1186/s13024-020-00375-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence demonstrates that adaptive immunity influences the pathobiology of neurodegenerative disorders. Misfolded aggregated self-proteins can break immune tolerance leading to the induction of autoreactive effector T cells (Teffs) with associated decreases in anti-inflammatory neuroprotective regulatory T cells (Tregs). An imbalance between Teffs and Tregs leads to microglial activation, inflammation and neuronal injury. The cascade of such a disordered immunity includes the drainage of the aggregated protein antigens into cervical lymph nodes serving to amplify effector immune responses. Both preclinical and clinical studies demonstrate transformation of this altered immunity for therapeutic gain. We posit that the signs and symptoms of common neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and stroke can be attenuated by boosting Treg activities.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Radiology, School of Medicine, Stanford University, Palo Alto, 94304 USA
| | - Ijaz Khan Muhammad
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacy, University of Swabi, Anbar Swabi, 23561 Pakistan
| | - Jonathan Herskovitz
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
15
|
Cho KJ, Ishido S, Eisenlohr LC, Roche PA. Activation of Dendritic Cells Alters the Mechanism of MHC Class II Antigen Presentation to CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:1621-1629. [PMID: 31996461 DOI: 10.4049/jimmunol.1901234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/31/2019] [Indexed: 11/19/2022]
Abstract
Both immature and mature dendritic cells (DCs) can process and present foreign Ags to CD4 T cells; however, the mechanism by which MHC class II (MHC-II) in mature DCs acquires antigenic peptides remains unknown. To address this, we have studied Ag processing and presentation of two distinct CD4 T cell epitopes of the influenza virus hemagglutinin coat protein by both immature and mature mouse DCs. We find that immature DCs almost exclusively use newly synthesized MHC-II targeted to DM+ late endosomes for presentation to influenza virus-specific CD4 T cells. By contrast, mature DCs exclusively use recycling MHC-II that traffics to both early and late endosomes for antigenic peptide binding. Rab11a knockdown partially inhibits recycling of MHC-II in mature DCs and selectively inhibits presentation of an influenza virus hemagglutinin CD4 T cell epitope generated in early endosomes. These studies highlight a "division of labor" in MHC-II peptide binding, in which immature DCs preferentially present Ags acquired in Rab11a- DM+ late endosomes, whereas mature DCs use recycling MHC-II to present antigenic peptides acquired in both Rab11a+ early endosomes and Rab11a- endosomes for CD4 T cell activation.
Collapse
Affiliation(s)
- Kyung-Jin Cho
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, 663-8501 Japan
| | - Laurence C Eisenlohr
- The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104; and.,Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
16
|
Rotavirus VP6 Adjuvant Effect on Norovirus GII.4 Virus-Like Particle Uptake and Presentation by Bone Marrow-Derived Dendritic Cells In Vitro and In Vivo. J Immunol Res 2020; 2020:3194704. [PMID: 32411793 PMCID: PMC7204108 DOI: 10.1155/2020/3194704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/20/2019] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that rotavirus (RV) inner capsid protein VP6 has an adjuvant effect on norovirus (NoV) virus-like particle- (VLP-) induced immune responses and studied the adjuvant mechanism in immortalized cell lines used as antigen-presenting cells (APCs). Here, we investigated the uptake and presentation of RV VP6 and NoV GII.4 VLPs by primary bone marrow-derived dendritic cells (BMDCs). The adjuvant effect of VP6 on GII.4 VLP presentation and NoV-specific immune response induction by BMDC in vivo was also studied. Intracellular staining demonstrated that BMDCs internalized both antigens, but VP6 more efficiently than NoV VLPs. Both antigens were processed and presented to antigen-primed T cells, which responded by robust interferon γ secretion. When GII.4 VLPs and VP6 were mixed in the same pulsing reaction, a subpopulation of the cells had uptaken both antigens. Furthermore, VP6 copulsing increased GII.4 VLP uptake by 37% and activated BMDCs to secrete 2-5-fold increased levels of interleukin 6 and tumor necrosis factor α compared to VLP pulsing alone. When in vitro-pulsed BMDCs were transferred to syngeneic BALB/c mice, VP6 improved NoV-specific antibody responses. The results of this study support the earlier findings of VP6 adjuvant effect in vitro and in vivo.
Collapse
|
17
|
Perez-Zsolt D, Martinez-Picado J, Izquierdo-Useros N. When Dendritic Cells Go Viral: The Role of Siglec-1 in Host Defense and Dissemination of Enveloped Viruses. Viruses 2019; 12:v12010008. [PMID: 31861617 PMCID: PMC7019426 DOI: 10.3390/v12010008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are among the first cells that recognize incoming viruses at the mucosal portals of entry. Initial interaction between DCs and viruses facilitates cell activation and migration to secondary lymphoid tissues, where these antigen presenting cells (APCs) prime specific adaptive immune responses. Some viruses, however, have evolved strategies to subvert the migratory capacity of DCs as a way to disseminate infection systemically. Here we focus on the role of Siglec-1, a sialic acid-binding type I lectin receptor potently upregulated by type I interferons on DCs, that acts as a double edge sword, containing viral replication through the induction of antiviral immunity, but also favoring viral spread within tissues. Such is the case for distant enveloped viruses like human immunodeficiency virus (HIV)-1 or Ebola virus (EBOV), which incorporate sialic acid-containing gangliosides on their viral membrane and are effectively recognized by Siglec-1. Here we review how Siglec-1 is highly induced on the surface of human DCs upon viral infection, the way this impacts different antigen presentation pathways, and how enveloped viruses have evolved to exploit these APC functions as a potent dissemination strategy in different anatomical compartments.
Collapse
Affiliation(s)
- Daniel Perez-Zsolt
- IrsiCaixa AIDS Research Institute, Ctra. de Canyet s/n, 08916 Badalona, Spain;
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Badalona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Ctra. de Canyet s/n, 08916 Badalona, Spain;
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Badalona, Spain
- Chair in Infectious Diseases and Immunity, Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (J.M.-P.); (N.I.-U.)
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Ctra. de Canyet s/n, 08916 Badalona, Spain;
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, 08916 Badalona, Spain
- Correspondence: (J.M.-P.); (N.I.-U.)
| |
Collapse
|
18
|
Hipolito VEB, Diaz JA, Tandoc KV, Oertlin C, Ristau J, Chauhan N, Saric A, Mclaughlan S, Larsson O, Topisirovic I, Botelho RJ. Enhanced translation expands the endo-lysosome size and promotes antigen presentation during phagocyte activation. PLoS Biol 2019; 17:e3000535. [PMID: 31800587 PMCID: PMC6913987 DOI: 10.1371/journal.pbio.3000535] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/16/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
The mechanisms that govern organelle adaptation and remodelling remain poorly defined. The endo-lysosomal system degrades cargo from various routes, including endocytosis, phagocytosis, and autophagy. For phagocytes, endosomes and lysosomes (endo-lysosomes) are kingpin organelles because they are essential to kill pathogens and process and present antigens. During phagocyte activation, endo-lysosomes undergo a morphological transformation, going from a collection of dozens of globular structures to a tubular network in a process that requires the phosphatidylinositol-3-kinase-AKT-mechanistic target of rapamycin (mTOR) signalling pathway. Here, we show that the endo-lysosomal system undergoes an expansion in volume and holding capacity during phagocyte activation within 2 h of lipopolysaccharides (LPS) stimulation. Endo-lysosomal expansion was paralleled by an increase in lysosomal protein levels, but this was unexpectedly largely independent of the transcription factor EB (TFEB) and transcription factor E3 (TFE3), which are known to scale up lysosome biogenesis. Instead, we demonstrate a hitherto unappreciated mechanism of acute organelle expansion via mTOR Complex 1 (mTORC1)-dependent increase in translation, which appears to be mediated by both S6Ks and 4E-BPs. Moreover, we show that stimulation of RAW 264.7 macrophage cell line with LPS alters translation of a subset but not all of mRNAs encoding endo-lysosomal proteins, thereby suggesting that endo-lysosome expansion is accompanied by functional remodelling. Importantly, mTORC1-dependent increase in translation activity was necessary for efficient and rapid antigen presentation by dendritic cells. Collectively, we identified a previously unknown and functionally relevant mechanism for endo-lysosome expansion that relies on mTORC1-dependent translation to stimulate endo-lysosome biogenesis in response to an infection signal. Activation of phagocytes rapidly expands the endo-lysosomal system and promotes antigen presentation. Endo-lysosome expansion was driven by mTORC1-dependent enhanced translation, revealing regulated translation as a mechanism to remodel membrane organelles in response to external signals and stresses.
Collapse
Affiliation(s)
- Victoria E. B. Hipolito
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Jacqueline A. Diaz
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Kristofferson V. Tandoc
- Department of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- The Lady Davis Institute, Jewish General Hospital, Montréal, Quebec, Canada
| | - Christian Oertlin
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Johannes Ristau
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Neha Chauhan
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Amra Saric
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Shannon Mclaughlan
- The Lady Davis Institute, Jewish General Hospital, Montréal, Quebec, Canada
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ivan Topisirovic
- Department of Experimental Medicine, McGill University, Montréal, Quebec, Canada
- The Lady Davis Institute, Jewish General Hospital, Montréal, Quebec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Roberto J. Botelho
- Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
19
|
Yang Y, Nam GH, Kim GB, Kim YK, Kim IS. Intrinsic cancer vaccination. Adv Drug Deliv Rev 2019; 151-152:2-22. [PMID: 31132376 DOI: 10.1016/j.addr.2019.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022]
Abstract
Immunotherapy is revolutionizing the treatment of cancer, and the current immunotherapeutics have remarkably improved the outcomes for some cancer patients. However, we still need answers for patients with immunologically cold tumors that do not benefit from the current immunotherapy treatments. Here, we suggest a novel strategy that is based on using a very old and sophisticated system for cancer immunotherapy, namely "intrinsic cancer vaccination", which seeks to awaken our own immune system to activate tumor-specific T cells. To do this, we must take advantage of the genetic instability of cancer cells and the expression of cancer cell neoantigens to trigger immunity against cancer cells. It will be necessary to not only enhance the phagocytosis of cancer cells by antigen presenting cells but also induce immunogenic cancer cell death and the subsequent immunogenic clearance, cross-priming and generation of tumor-specific T cells. This strategy will allow us to avoid using known tumor-specific antigens, ex vivo manipulation or adoptive cell therapy; rather, we will efficiently present cancer cell neoantigens to our immune system and propagate the cancer-immunity cycle. This strategy simply follows the natural cycle of cancer-immunity from its very first step, and therefore could be combined with any other treatment modality to yield enhanced efficacy.
Collapse
Affiliation(s)
- Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Gi-Hoon Nam
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Gi Beom Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yoon Kyoung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.
| |
Collapse
|
20
|
Kotsias F, Cebrian I, Alloatti A. Antigen processing and presentation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:69-121. [PMID: 31810556 DOI: 10.1016/bs.ircmb.2019.07.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells are at the center of immune responses. They are defined by their ability to sense the environment, take up and process antigen, migrate to secondary lymphoid organs, where they present antigens to the adaptive immune system. In particular, they present lipids and proteins from pathogens, which they encountered in peripheral tissues, to T cells in order to induce a specific effector immune response. These complex antigens need to be broken down into peptides of a certain length in association with Major Histocompatibility Complex (MHC) molecules. Presentation of MHC/antigen complexes alongside costimulatory molecules and secretion of proinflammatory cytokines will induce an appropriate immune response. This interaction between dendritic cells and T cells takes place at defined locations within secondary lymphoid organs. In this review, we discuss the current knowledge and recent advances on the cellular and molecular mechanisms that underlie antigen processing and the subsequent presentation to T lymphocytes.
Collapse
Affiliation(s)
- Fiorella Kotsias
- Cátedra de Virología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina; Instituto de Investigaciones en Producción Animal (INPA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Ignacio Cebrian
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET/Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Andrés Alloatti
- Facultad de Ciencias Médicas, Instituto de Inmunología Clínica y Experimental de Rosario (IDICER)-CONICET/Universidad Nacional de Rosario, Rosario, Argentina.
| |
Collapse
|
21
|
Hossain MK, Vartak A, Sucheck SJ, Wall KA. Liposomal Fc Domain Conjugated to a Cancer Vaccine Enhances Both Humoral and Cellular Immunity. ACS OMEGA 2019; 4:5204-5208. [PMID: 30949616 PMCID: PMC6441943 DOI: 10.1021/acsomega.9b00029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/27/2019] [Indexed: 06/09/2023]
Abstract
Targeted delivery of antigens to antigen-presenting cells (APCs) by utilizing natural anticarbohydrate antibodies is a promising approach for selective uptake and enhanced antigen presentation. Previously, we reported that in the presence of a natural antibody, anti-rhamnose antibody (anti-Rha), the bacterial sugar rhamnose conjugated with liposomal cancer antigen MUC1-Tn enhances antigen presentation by APCs such as dendritic cells by targeting Fc gamma receptors. The idea was to utilize the natural human anti-Rha antibodies present in human serum for targeted delivery of cancer-specific antigens. Recently, we found that the IgG3 antibody isotype was the most prevalent anti-Rha antibody generated in mice immunized with rhamnose-Ficoll (Rha-Ficoll) antigen. In this manuscript, we have conjugated the murine IgG3-Fc with a MUC1-containing cancer vaccine and compared the humoral and cellular immune response to this vaccine with one targeted via the human anti-Rha antibody and to the MUC1 vaccine alone. This Fc approach enhanced antibody production and T-cell proliferation almost to the same level as using the anti-Rha antibody. These results suggest that targeting Fc directly to dendritic cells can be an alternative approach to human anti-Rha for generating effective antigen-primed T-cells.
Collapse
Affiliation(s)
- Md Kamal Hossain
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Abhishek Vartak
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Steven J. Sucheck
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Katherine A. Wall
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| |
Collapse
|
22
|
Rossi A, Dupaty L, Aillot L, Zhang L, Gallien C, Hallek M, Odenthal M, Adriouch S, Salvetti A, Büning H. Vector uncoating limits adeno-associated viral vector-mediated transduction of human dendritic cells and vector immunogenicity. Sci Rep 2019; 9:3631. [PMID: 30842485 PMCID: PMC6403382 DOI: 10.1038/s41598-019-40071-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/05/2019] [Indexed: 12/28/2022] Open
Abstract
AAV vectors poorly transduce Dendritic cells (DC), a feature invoked to explain AAV's low immunogenicity. However, the reason for this non-permissiveness remained elusive. Here, we performed an in-depth analysis using human monocyte-derived immature DC (iDC) as model. iDC internalized AAV vectors of various serotypes, but even the most efficient serotype failed to transduce iDC above background. Since AAV vectors reached the cell nucleus, we hypothesized that AAV's intracellular processing occurs suboptimal. On this basis, we screened an AAV peptide display library for capsid variants more suitable for DC transduction and identified the I/VSS family which transduced DC with efficiencies of up to 38%. This property correlated with an improved vector uncoating. To determine the consequence of this novel feature for AAV's in vivo performance, we engineered one of the lead candidates to express a cytoplasmic form of ovalbumin, a highly immunogenic model antigen, and assayed transduction efficiency as well as immunogenicity. The capsid variant clearly outperformed the parental serotype in muscle transduction and in inducing antigen-specific humoral and T cell responses as well as anti-capsid CD8+ T cells. Hence, vector uncoating represents a major barrier hampering AAV vector-mediated transduction of DC and impacts on its use as vaccine platform.
Collapse
Affiliation(s)
- Axel Rossi
- International Center for Research in Infectiology (CIRI), INSERM U1111 - Université claude Bernard Lyon 1, CNRS UMR5308, Ecole Normale Supérieur de Lyon, Université de Lyon, Lyon, France
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Léa Dupaty
- Normandie Univ, UNIROUEN, INSERM, U1234, Physiopathologie et biothérapies des maladies inflammatoires et autoimmunes (PANTHER), 76000, Rouen, France
| | - Ludovic Aillot
- International Center for Research in Infectiology (CIRI), INSERM U1111 - Université claude Bernard Lyon 1, CNRS UMR5308, Ecole Normale Supérieur de Lyon, Université de Lyon, Lyon, France
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR5206, Lyon, France
| | - Liang Zhang
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Célia Gallien
- International Center for Research in Infectiology (CIRI), INSERM U1111 - Université claude Bernard Lyon 1, CNRS UMR5308, Ecole Normale Supérieur de Lyon, Université de Lyon, Lyon, France
| | - Michael Hallek
- Clinic I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | | | - Sahil Adriouch
- Normandie Univ, UNIROUEN, INSERM, U1234, Physiopathologie et biothérapies des maladies inflammatoires et autoimmunes (PANTHER), 76000, Rouen, France.
| | - Anna Salvetti
- International Center for Research in Infectiology (CIRI), INSERM U1111 - Université claude Bernard Lyon 1, CNRS UMR5308, Ecole Normale Supérieur de Lyon, Université de Lyon, Lyon, France.
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR5206, Lyon, France.
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover, Germany.
| |
Collapse
|
23
|
Class II MHC antigen processing in immune tolerance and inflammation. Immunogenetics 2018; 71:171-187. [PMID: 30421030 DOI: 10.1007/s00251-018-1095-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/31/2018] [Indexed: 01/22/2023]
Abstract
Presentation of peptide antigens by MHC-II proteins is prerequisite to effective CD4 T cell tolerance to self and to recognition of foreign antigens. Antigen uptake and processing pathways as well as expression of the peptide exchange factors HLA-DM and HLA-DO differ among the various professional and non-professional antigen-presenting cells and are modulated by cell developmental state and activation. Recent studies have highlighted the importance of these cell-specific factors in controlling the source and breadth of peptides presented by MHC-II under different conditions. During inflammation, increased presentation of selected self-peptides has implications for maintenance of peripheral tolerance and autoimmunity.
Collapse
|
24
|
Kim WS, Shin MK, Shin SJ. MAP1981c, a Putative Nucleic Acid-Binding Protein, Produced by Mycobacterium avium subsp. paratuberculosis, Induces Maturation of Dendritic Cells and Th1-Polarization. Front Cell Infect Microbiol 2018; 8:206. [PMID: 29977867 PMCID: PMC6021526 DOI: 10.3389/fcimb.2018.00206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/05/2018] [Indexed: 12/23/2022] Open
Abstract
Mycobacterium avium subsp. paratuberculosis (MAP) is the causative pathogen of chronic granulomatous enteropathy (Johne's disease) in animals, and has been focused on its association with various autoimmune diseases in humans, including Crohn's disease. The discovery of novel mycobacterial antigens and exploring their role in host immunity can contribute to the advancement of effective defense strategies including vaccines and diagnostic tools. In a preliminary study, we identified cellular extract proteins of MAP that strongly react with the blood of patients with Crohn's disease. In particular, MAP1981c, a putative nucleic acid-binding protein, showed high expression levels and strong reactivity to IgG and IgM in the sera of patients. Here, we investigated the immunological features of MAP1981c and focused on its interaction with dendritic cells (DCs), confirming its immunomodulatory ability. MAP1981c was shown to recognize Toll-like receptor (TLR) 4, and induce DC maturation and activation by increasing the expression of co-stimulatory (CD80 and CD86) and MHC class I/II molecules and the secretion of pro-inflammatory cytokines (IL-6, IL-1β, and TNF-α) in DCs. This DC activation by MAP1981c was mediated by downstream signaling of TLR4 via MyD88- and TRIF-, MAP kinase-, and NF-κB-dependent signaling pathways. In addition, MAP1981c-treated DCs activated naïve T cells and induced the differentiation of CD4+ and CD8+ T cells to express T-bet, IFN-γ, and/or IL-2, but not GATA-3 and IL-4, thus indicating that MAP1981c contributes to Th1-type immune responses both in vitro and in vivo. Taken together, these results suggest that MAP1981c is a novel immunocompetent antigen that induces DC maturation and a Th1-biased response upon DC activation, suggesting that MAP1981c can be an effective vaccine and diagnostic target.
Collapse
Affiliation(s)
- Woo Sik Kim
- Department of Microbiology and Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology and Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
25
|
Abstract
Antigen cross-presentation is an adaptation of the cellular process of loading MHC-I molecules with endogenous peptides during their biosynthesis within the endoplasmic reticulum. Cross-presented peptides derive from internalized proteins, microbial pathogens, and transformed or dying cells. The physical separation of internalized cargo from the endoplasmic reticulum, where the machinery for assembling peptide-MHC-I complexes resides, poses a challenge. To solve this problem, deliberate rewiring of organelle communication within cells is necessary to prepare for cross-presentation, and different endocytic receptors and vesicular traffic patterns customize the emergent cross-presentation compartment to the nature of the peptide source. Three distinct pathways of vesicular traffic converge to form the ideal cross-presentation compartment, each regulated differently to supply a unique component that enables cross-presentation of a diverse repertoire of peptides. Delivery of centerpiece MHC-I molecules is the critical step regulated by microbe-sensitive Toll-like receptors. Defining the subcellular sources of MHC-I and identifying sites of peptide loading during cross-presentation remain key challenges.
Collapse
Affiliation(s)
- J Magarian Blander
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; .,Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
26
|
Sachamitr P, Leishman AJ, Davies TJ, Fairchild PJ. Directed Differentiation of Human Induced Pluripotent Stem Cells into Dendritic Cells Displaying Tolerogenic Properties and Resembling the CD141 + Subset. Front Immunol 2018; 8:1935. [PMID: 29358940 PMCID: PMC5766641 DOI: 10.3389/fimmu.2017.01935] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) has begun to revolutionize cell therapy by providing a convenient source of rare cell types not normally available from patients in sufficient numbers for therapeutic purposes. In particular, the development of protocols for the differentiation of populations of leukocytes as diverse as naïve T cells, macrophages, and natural killer cells provides opportunities for their scale-up and quality control prior to administration. One population of leukocytes whose therapeutic potential has yet to be explored is the subset of conventional dendritic cells (DCs) defined by their surface expression of CD141. While these cells stimulate cytotoxic T cells in response to inflammation through the cross-presentation of viral and tumor-associated antigens in an MHC class I-restricted manner, under steady-state conditions CD141+ DCs resident in interstitial tissues are focused on the maintenance of homeostasis through the induction of tolerance to local antigens. Here, we describe protocols for the directed differentiation of human iPSCs into a mixed population of CD11c+ DCs through the spontaneous formation of embryoid bodies and exposure to a cocktail of growth factors, the scheduled withdrawal of which serves to guide the process of differentiation. Furthermore, we describe the enrichment of DCs expressing CD141 through depletion of CD1c+ cells, thereby obtaining a population of “untouched” DCs unaffected by cross-linking of surface CD141. The resulting cells display characteristic phagocytic and endocytic capacity and acquire an immunostimulatory phenotype following exposure to inflammatory cytokines and toll-like receptor agonists. Nevertheless, under steady-state conditions, these cells share some of the tolerogenic properties of tissue-resident CD141+ DCs, which may be further reinforced by exposure to a range of pharmacological agents including interleukin-10, rapamycin, dexamethasone, and 1α,25-dihydoxyvitamin D3. Our protocols therefore provide access to a novel source of DCs analogous to the CD141+ subset under steady-state conditions in vivo and may, therefore, find utility in the treatment of a range of disease states requiring the establishment of immunological tolerance.
Collapse
Affiliation(s)
- Patty Sachamitr
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Alison J Leishman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Timothy J Davies
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul J Fairchild
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Domogalla MP, Rostan PV, Raker VK, Steinbrink K. Tolerance through Education: How Tolerogenic Dendritic Cells Shape Immunity. Front Immunol 2017; 8:1764. [PMID: 29375543 PMCID: PMC5770648 DOI: 10.3389/fimmu.2017.01764] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/27/2017] [Indexed: 12/27/2022] Open
Abstract
Dendritic cells (DCs) are central players in the initiation and control of responses, regulating the balance between tolerance and immunity. Tolerogenic DCs are essential in the maintenance of central and peripheral tolerance by induction of clonal T cell deletion and T cell anergy, inhibition of memory and effector T cell responses, and generation and activation of regulatory T cells. Therefore, tolerogenic DCs are promising candidates for specific cellular therapy of allergic and autoimmune diseases and for treatment of transplant rejection. Studies performed in rodents have demonstrated the efficacy and feasibility of tolerogenic DCs for tolerance induction in various inflammatory diseases. In the last years, numerous protocols for the generation of human monocyte-derived tolerogenic DCs have been established and some first phase I trials have been conducted in patients suffering from autoimmune disorders, demonstrating the safety and efficiency of this cell-based immunotherapy. This review gives an overview about methods and protocols for the generation of human tolerogenic DCs and their mechanisms of tolerance induction with the focus on interleukin-10-modulated DCs. In addition, we will discuss the prerequisites for optimal clinical grade tolerogenic DC subsets and results of clinical trials with tolerogenic DCs in autoimmune diseases.
Collapse
Affiliation(s)
- Matthias P Domogalla
- Department of Dermatology, Division for Experimental and Translational Research, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Patricia V Rostan
- Department of Dermatology, Division for Experimental and Translational Research, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Verena K Raker
- Department of Dermatology, Division for Experimental and Translational Research, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, Division for Experimental and Translational Research, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
28
|
Pierini S, Perales-Linares R, Uribe-Herranz M, Pol JG, Zitvogel L, Kroemer G, Facciabene A, Galluzzi L. Trial watch: DNA-based vaccines for oncological indications. Oncoimmunology 2017; 6:e1398878. [PMID: 29209575 DOI: 10.1080/2162402x.2017.1398878] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/24/2017] [Indexed: 12/16/2022] Open
Abstract
DNA-based vaccination is a promising approach to cancer immunotherapy. DNA-based vaccines specific for tumor-associated antigens (TAAs) are indeed relatively simple to produce, cost-efficient and well tolerated. However, the clinical efficacy of DNA-based vaccines for cancer therapy is considerably limited by central and peripheral tolerance. During the past decade, considerable efforts have been devoted to the development and characterization of novel DNA-based vaccines that would circumvent this obstacle. In this setting, particular attention has been dedicated to the route of administration, expression of modified TAAs, co-expression of immunostimulatory molecules, and co-delivery of immune checkpoint blockers. Here, we review preclinical and clinical progress on DNA-based vaccines for cancer therapy.
Collapse
Affiliation(s)
- Stefano Pierini
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renzo Perales-Linares
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mireia Uribe-Herranz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan G Pol
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Andrea Facciabene
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Ovarian Cancer Research Center (OCRC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
29
|
Weng TY, Li CJ, Li CY, Hung YH, Yen MC, Chang YW, Chen YH, Chen YL, Hsu HP, Chang JY, Lai MD. Skin Delivery of Clec4a Small Hairpin RNA Elicited an Effective Antitumor Response by Enhancing CD8 + Immunity In Vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:419-427. [PMID: 29246320 PMCID: PMC5701806 DOI: 10.1016/j.omtn.2017.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 10/21/2017] [Accepted: 10/22/2017] [Indexed: 12/01/2022]
Abstract
Clec4a has been reported to be an immune suppressor of dendritic cells (DCs), but its potential role in cancer therapy remains to be elucidated. The present study investigated whether downregulating the expression of Clec4a via skin delivery of small hairpin RNA (shRNA) using a gene gun produced stronger host immunity and inhibited tumor progression in animal models. Administration of Clec4a2 shRNA delayed tumor growth in both mouse bladder and lung tumor-bearing mouse models. The result was further confirmed with a compensation experiment showing that the antitumor effects induced by Clec4a2 shRNA were restored by co-injection of a plasmid expressing exogenous Clec4a2. Increased numbers of infiltrating CD4+ and CD8+ T cells at tumor sites were observed in mice treated with Clec4a2 shRNA. Splenocytes from mice with Clec4a2 shRNA administration exhibited stronger cytotoxic activity compared with splenocytes from control mice. CD8-deletion in vivo abrogated the antitumor effects elicited by Clec4a2 shRNA. Additionally, shClec4a enhanced the antitumor effects of the Neu DNA vaccine in the MBT-2 tumor model. In summary, the findings provide evidence that silencing of Clec4a2 expression via skin delivery of shRNA produces an effective antitumor response and that Clec4a2 shRNA may have therapeutic potential as an adjuvant for cancer immunotherapy.
Collapse
Affiliation(s)
- Tzu-Yang Weng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan; Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Chia-Jung Li
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Chung-Yen Li
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Yu-Hsuan Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 68, Jhonghua 3rd Rd., Cianjin District, Kaohsiung City 80145, Taiwan
| | - Yu-Wei Chang
- Department of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Yu-Hung Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan; Department of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Yi-Ling Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, No. 60, Sec. 1, Erren Rd., Rende District, Tainan City 717, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institute, No. 367, Sheng-li Rd., North District, Tainan City 704, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City 701, Taiwan.
| |
Collapse
|
30
|
Blander JM. The comings and goings of MHC class I molecules herald a new dawn in cross-presentation. Immunol Rev 2017; 272:65-79. [PMID: 27319343 DOI: 10.1111/imr.12428] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MHC class I (MHC-I) molecules are the centerpieces of cross-presentation. They are loaded with peptides derived from exogenous sources and displayed on the plasma membrane to communicate with CD8 T cells, relaying a message of tolerance or attack. The study of cross-presentation has been focused on the relative contributions of the vacuolar versus cytosolic pathways of antigen processing and the location where MHC-I molecules are loaded. While vacuolar processing generates peptides loaded onto vacuolar MHC-I molecules, how and where exogenous peptides generated by the proteasome and transported by TAP meet MHC-I molecules for loading has been a matter of debate. The source and trafficking of MHC-I molecules in dendritic cells have largely been ignored under the expectation that these molecules came from the Endoplasmic reticulum (ER) or the plasma membrane. New studies reveal a concentrated pool of MHC-I molecules in the endocytic recycling compartment (ERC). These pools are rapidly mobilized to phagosomes carrying microbial antigens, and in a signal-dependent manner under the control of Toll-like receptors. The phagosome becomes a dynamic hub receiving traffic from multiple sources, the ER-Golgi intermediate compartment for delivering the peptide-loading machinery and the ERC for deploying MHC-I molecules that alert CD8 T cells of infection.
Collapse
Affiliation(s)
- J Magarian Blander
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
31
|
Alloatti A, Kotsias F, Magalhaes JG, Amigorena S. Dendritic cell maturation and cross-presentation: timing matters! Immunol Rev 2017; 272:97-108. [PMID: 27319345 PMCID: PMC6680313 DOI: 10.1111/imr.12432] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As a population, dendritic cells (DCs) appear to be the best cross‐presenters of internalized antigens on major histocompatibility complex class I molecules in the mouse. To do this, DCs have developed a number of unique and dedicated means to control their endocytic and phagocytic pathways: among them, the capacity to limit acidification of their phagosomes, to prevent proteolytic degradation, to delay fusion of phagosomes to lysosomes, to recruit ER proteins to phagosomes, and to export phagocytosed antigens to the cytosol. The regulation of phagocytic functions, and thereby of antigen processing and presentation by innate signaling, represents a critical level of integration of adaptive and innate immune responses. Understanding how innate signals control antigen cross‐presentation is critical to define effective vaccination strategies for CD8+ T‐cell responses.
Collapse
Affiliation(s)
- Andrés Alloatti
- Institut Curie, PSL Research University, INSERM U932, Paris Cedex 05, France
| | - Fiorella Kotsias
- Institut Curie, PSL Research University, INSERM U932, Paris Cedex 05, France.,Department of Virology, Faculty of Veterinary Sciences, University of Buenos Aires, Buenos Aires, and CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Argentina
| | | | - Sebastian Amigorena
- Institut Curie, PSL Research University, INSERM U932, Paris Cedex 05, France
| |
Collapse
|
32
|
Abstract
Unlike B cells, CD8-positive and CD4-positive T cells of the adaptive immune system do not recognize intact foreign proteins but instead recognize polypeptide fragments of potential antigens. These antigenic peptides are expressed on the surface of antigen presenting cells bound to MHC class I and MHC class II proteins. Here, we review the basics of antigen acquisition by antigen presenting cells, antigen proteolysis into polypeptide fragments, antigenic peptide binding to MHC proteins, and surface display of both MHC class I-peptide and MHC class II-peptide complexes.
Collapse
|
33
|
Yokota-Nakatsuma A, Ohoka Y, Takeuchi H, Song SY, Iwata M. Beta 1-integrin ligation and TLR ligation enhance GM-CSF-induced ALDH1A2 expression in dendritic cells, but differentially regulate their anti-inflammatory properties. Sci Rep 2016; 6:37914. [PMID: 27897208 PMCID: PMC5126582 DOI: 10.1038/srep37914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022] Open
Abstract
Retinoic acid (RA)–producing CD103+ mature dendritic cells (DCs) in mesenteric lymph nodes (MLNs) play crucial roles in gut immunity. GM-CSF and RA contribute to the expression of the RA-producing enzyme ALDH1A2. However, additional signals appeared to be required for inducing ALDH1A2high mature DCs from immature DCs. We found here that TLR ligands (Ls) and immobilized E-cadherin could provide such signals in FLT3-L–generated bone marrow (BM)–derived DCs after treatment with GM-CSF and the RA receptor agonist Am80. The TLR-L-treated DCs produced proinflammatory cytokines unlike normal ALDH1A2high MLN-DCs, whereas the E-cadherin-treated DCs did not. Immobilized VCAM-1 and semaphorin 7 A exerted effects similar to those of E-cadherin. Soluble anti-integrin β1 antibodies or inhibitors of integrin signaling molecules suppressed the effects of these immobilized proteins, whereas immobilized anti-integrin β1 antibodies enhanced the GM-CSF/Am80-induced ALDH1A2 expression without inducing proinflammatory cytokines. Sequential stimulation of splenic pre-DCs with GM-CSF/Am80 and immobilized E-cadherin or anti-integrin β1 antibody also induced differentiation to mature DCs with high ALDH activity. The E-cadherin-treated BM-DCs induced gut-tropic Foxp3+ T cells and alleviated DSS–induced colitis, whereas the TLR-L-treated DCs aggravated DSS–induced colitis. The results suggest that integrin β1-mediated signals contribute to the differentiation and maturation of RA-producing anti-inflammatory DCs.
Collapse
Affiliation(s)
- Aya Yokota-Nakatsuma
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| | - Yoshiharu Ohoka
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| | - Hajime Takeuchi
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| | - Si-Young Song
- Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan.,Institute of Neuroscience, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan
| | - Makoto Iwata
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Shido, Sanuki-shi, Kagawa, Japan.,Japan Science and Technology Agency, CREST, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
34
|
Calmette J, Bertrand M, Vétillard M, Ellouze M, Flint S, Nicolas V, Biola-Vidamment A, Pallardy M, Morand E, Bachelerie F, Godot V, Schlecht-Louf G. Glucocorticoid-Induced Leucine Zipper Protein Controls Macropinocytosis in Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:4247-4256. [PMID: 27793999 DOI: 10.4049/jimmunol.1600561] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 09/27/2016] [Indexed: 12/25/2022]
Abstract
Ag sampling is a key process in dendritic cell (DC) biology. DCs use constitutive macropinocytosis, receptor-mediated endocytosis, and phagocytosis to capture exogenous Ags for presentation to T cells. We investigated the mechanisms that regulate Ag uptake by DCs in the steady-state and after a short-term LPS exposure in vitro and in vivo. We show that the glucocorticoid-induced leucine zipper protein (GILZ), already known to regulate effector versus regulatory T cell activation by DCs, selectively limits macropinocytosis, but not receptor-mediated phagocytosis, in immature and recently activated DCs. In vivo, the GILZ-mediated inhibition of Ag uptake is restricted to the CD8α+ DC subset, which expresses the highest GILZ level among splenic DC subsets. In recently activated DCs, we further establish that GILZ limits p38 MAPK phosphorylation, providing a possible mechanism for GILZ-mediated macropinocytosis control. Finally, our results demonstrate that the modulation of Ag uptake by GILZ does not result in altered Ag presentation to CD4 T cells but impacts the efficiency of cross-presentation to CD8 T cells. Altogether, our results identify GILZ as an endogenous inhibitor of macropinocytosis in DCs, the action of which contributes to the fine-tuning of Ag cross-presentation.
Collapse
Affiliation(s)
- Joseph Calmette
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Clamart 92140, France
| | - Matthieu Bertrand
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Clamart 92140, France
| | - Mathias Vétillard
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Clamart 92140, France
| | - Mehdi Ellouze
- UMR955, Team 16, Institut de Recherche Vaccinal, INSERM, Université Paris Est Créteil, Créteil 94010, France
| | - Shaun Flint
- Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Valérie Nicolas
- Institut Paris-Sud d'Innovation Thérapeutique, SFR-UMS, Chatenay Malabry 92296, France
| | - Armelle Biola-Vidamment
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Chatenay Malabry 92296, France; and
| | - Marc Pallardy
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Chatenay Malabry 92296, France; and
| | - Eric Morand
- Southern Clinical School, Monash University Faculty of Medicine, Nursing, and Health Sciences, Clayton, Victoria 3168, Australia
| | - Françoise Bachelerie
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Clamart 92140, France
| | - Véronique Godot
- UMR955, Team 16, Institut de Recherche Vaccinal, INSERM, Université Paris Est Créteil, Créteil 94010, France
| | - Géraldine Schlecht-Louf
- UMR996-Inflammation, Chimiokines et Immunopathologie, INSERM, Université Paris-Sud, Université Paris-Saclay, Clamart 92140, France;
| |
Collapse
|
35
|
Mitochondrial respiratory-chain adaptations in macrophages contribute to antibacterial host defense. Nat Immunol 2016; 17:1037-1045. [PMID: 27348412 DOI: 10.1038/ni.3509] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/03/2016] [Indexed: 12/15/2022]
Abstract
Macrophages tightly scale their core metabolism after being activated, but the precise regulation of the mitochondrial electron-transport chain (ETC) and its functional implications are currently unknown. Here we found that recognition of live bacteria by macrophages transiently decreased assembly of the ETC complex I (CI) and CI-containing super-complexes and switched the relative contributions of CI and CII to mitochondrial respiration. This was mediated by phagosomal NADPH oxidase and the reactive oxygen species (ROS)-dependent tyrosine kinase Fgr. It required Toll-like receptor signaling and the NLRP3 inflammasome, which were both connected to bacterial viability-specific immune responses. Inhibition of CII during infection with Escherichia coli normalized serum concentrations of interleukin 1β (IL-1β) and IL-10 to those in mice treated with dead bacteria and impaired control of bacteria. We have thus identified ETC adaptations as an early immunological-metabolic checkpoint that adjusts innate immune responses to bacterial infection.
Collapse
|
36
|
Qian Y, Jin H, Qiao S, Dai Y, Huang C, Lu L, Luo Q, Zhang Z. Targeting dendritic cells in lymph node with an antigen peptide-based nanovaccine for cancer immunotherapy. Biomaterials 2016; 98:171-83. [PMID: 27192420 DOI: 10.1016/j.biomaterials.2016.05.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/16/2016] [Accepted: 05/03/2016] [Indexed: 12/21/2022]
Abstract
The design of peptide-based subunit vaccine formulations for the direct delivery of tumor antigen peptides (Aps) to dendritic cells (DCs) localized within draining lymph nodes (DLNs) is challenging. Mature DCs (mDCs) are abundantly distributed within DLNs but have dramatically reduced endocytic uptake and antigen-processing abilities, so their role as potential vaccine targets has been largely overlooked. Here we report an ultra-small biocompatible nanovaccine (α-Ap-FNP) functionalized by avidly targeting delivery of Ap via the scavenger receptor class B1 (SR-B1) pathway to mDCs. The self-assembly, small size (∼30 nm), SR-B1-targeting and optical properties of α-Ap-FNP resulted in its efficient Ap loading, substantial LN accumulation, targeting of mDCs and enhanced Ap presentation, and fluorescence trafficking, respectively. We also demonstrate that the α-Ap-FNP can be either used alone or encapsulated with CpG oligodeoxynucleotide as a prophylactic and therapeutic vaccine. Thus, the excellent properties of α-Ap-FNP provide it potential for clinical applications as a potent nanovaccine for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuan Qian
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Honglin Jin
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China; Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Sha Qiao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yanfeng Dai
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chuan Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China; MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
37
|
Coria LM, Ibañez AE, Tkach M, Sabbione F, Bruno L, Carabajal MV, Berguer PM, Barrionuevo P, Schillaci R, Trevani AS, Giambartolomei GH, Pasquevich KA, Cassataro J. A Brucella spp. Protease Inhibitor Limits Antigen Lysosomal Proteolysis, Increases Cross-Presentation, and Enhances CD8+ T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2016; 196:4014-29. [PMID: 27084100 DOI: 10.4049/jimmunol.1501188] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 03/15/2016] [Indexed: 01/18/2023]
Abstract
In this study, we demonstrate that the unlipidated (U) outer membrane protein (Omp) 19 from Brucella spp. is a competitive inhibitor of human cathepsin L. U-Omp19 inhibits lysosome cathepsins and APC-derived microsome activity in vitro and partially inhibits lysosomal cathepsin L activity within live APCs. Codelivery of U-Omp19 with the Ag can reduce intracellular Ag digestion and increases Ag half-life in dendritic cells (DCs). U-Omp19 retains the Ag in Lamp-2(+) compartments after its internalization and promotes a sustained expression of MHC class I/peptide complexes in the cell surface of DCs. Consequently, U-Omp19 enhances Ag cross-presentation by DCs to CD8(+) T cells. U-Omp19 s.c. delivery induces the recruitment of CD11c(+)CD8α(+) DCs and monocytes to lymph nodes whereas it partially limits in vivo Ag proteolysis inside DCs. Accordingly, this protein is able to induce CD8(+) T cell responses in vivo against codelivered Ag. Antitumor responses were elicited after U-Omp19 coadministration, increasing survival of mice in a murine melanoma challenge model. Collectively, these results indicate that a cysteine protease inhibitor from bacterial origin could be a suitable component of vaccine formulations against tumors.
Collapse
Affiliation(s)
- Lorena M Coria
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Andrés E Ibañez
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Mercedes Tkach
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1428 Buenos Aires, Argentina
| | - Florencia Sabbione
- Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Laura Bruno
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Marianela V Carabajal
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Paula M Berguer
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1405 Buenos Aires, Argentina; and
| | - Paula Barrionuevo
- Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Roxana Schillaci
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1428 Buenos Aires, Argentina
| | - Analía S Trevani
- Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo, Laboratorio de Inmunogenética, Hospital de Clínicas "José de San Martín," Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad de Buenos Aires, 1120 Buenos Aires, Argentina
| | - Karina A Pasquevich
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina;
| |
Collapse
|
38
|
Zavašnik-Bergant T, Bergant Marušič M. Exogenous Thyropin from p41 Invariant Chain Diminishes Cysteine Protease Activity and Affects IL-12 Secretion during Maturation of Human Dendritic Cells. PLoS One 2016; 11:e0150815. [PMID: 26960148 PMCID: PMC4784741 DOI: 10.1371/journal.pone.0150815] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 02/19/2016] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DC) play a pivotal role as antigen presenting cells (APC) and their maturation is crucial for effectively eliciting an antigen-specific immune response. The p41 splice variant of MHC class II-associated chaperone, called invariant chain p41 Ii, contains an amino acid sequence, the p41 fragment, which is a thyropin-type inhibitor of proteolytic enzymes. The effects of exogenous p41 fragment and related thyropin inhibitors acting on human immune cells have not been reported yet. In this study we demonstrate that exogenous p41 fragment can enter the endocytic pathway of targeted human immature DC. Internalized p41 fragment has contributed to the total amount of the immunogold labelled p41 Ii-specific epitope, as quantified by transmission electron microscopy, in particular in late endocytic compartments with multivesicular morphology where antigen processing and binding to MHC II take place. In cell lysates of treated immature DC, diminished enzymatic activity of cysteine proteases has been confirmed. Internalized exogenous p41 fragment did not affect the perinuclear clustering of acidic cathepsin S-positive vesicles typical of mature DC. p41 fragment is shown to interfere with the nuclear translocation of NF-κB p65 subunit in LPS-stimulated DC. p41 fragment is also shown to reduce the secretion of interleukin-12 (IL-12/p70) during the subsequent maturation of treated DC. The inhibition of proteolytic activity of lysosomal cysteine proteases in immature DC and the diminished capability of DC to produce IL-12 upon their subsequent maturation support the immunomodulatory potential of the examined thyropin from p41 Ii.
Collapse
Affiliation(s)
- Tina Zavašnik-Bergant
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- * E-mail:
| | | |
Collapse
|
39
|
Coria LM, Ibañez AE, Pasquevich KA, Cobiello PLG, Frank FM, Giambartolomei GH, Cassataro J. Brucella abortus Omp19 recombinant protein subcutaneously co-delivered with an antigen enhances antigen-specific T helper 1 memory responses and induces protection against parasite challenge. Vaccine 2015; 34:430-437. [PMID: 26707377 DOI: 10.1016/j.vaccine.2015.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/27/2015] [Accepted: 12/09/2015] [Indexed: 01/18/2023]
Abstract
The discovery of effective adjuvants for many vaccines especially those with limited commercial appeal, such as vaccines to poverty-related diseases, is required. In this work, we demonstrated that subcutaneous co-administration of mice with the outer membrane protein U-Omp19 from Brucella spp. plus OVA as antigen (Ag) increases Ag-specific T cell proliferation and T helper (Th) 1 immune responses in vitro and in vivo. U-Omp19 treated dendritic cells promote IFN-γ production by specific CD4(+) T cells and increases T cell proliferation. U-Omp19 co-administration induces the production of Ag specific effector memory T cell populations (CD4(+) CD44(high) CD62L(low) T cells). Finally, subcutaneous co-administration of U-Omp19 with Trypanosoma cruzi Ags confers protection against virulent parasite challenge, reducing parasitemia and weight loss while increasing mice survival. These results indicate that the bacterial protein U-Omp19 when delivered subcutaneously could be a suitable component of vaccine formulations against infectious diseases requiring Th1 immune responses.
Collapse
Affiliation(s)
- Lorena M Coria
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM), CONICET, Buenos Aires, Argentina
| | - Andrés E Ibañez
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM), CONICET, Buenos Aires, Argentina
| | - Karina A Pasquevich
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM), CONICET, Buenos Aires, Argentina
| | - Paula L González Cobiello
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Fernanda M Frank
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Laboratorio de Inmunogenética, Hospital de Clínicas "José de San Martín", CONICET-UBA, Buenos Aires, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM), CONICET, Buenos Aires, Argentina.
| |
Collapse
|
40
|
The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 2015; 15:203-16. [PMID: 25720354 DOI: 10.1038/nri3818] [Citation(s) in RCA: 662] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antigenic peptide-loaded MHC class II molecules (peptide-MHC class II) are constitutively expressed on the surface of professional antigen-presenting cells (APCs), including dendritic cells, B cells, macrophages and thymic epithelial cells, and are presented to antigen-specific CD4(+) T cells. The mechanisms of antigen uptake, the nature of the antigen processing compartments and the lifetime of cell surface peptide-MHC class II complexes can vary depending on the type of APC. It is likely that these differences are important for the function of each distinct APC subset in the generation of effective adaptive immune responses. In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide-MHC class II complexes, the intracellular trafficking of peptide-MHC class II complexes to the APC plasma membrane and their ultimate degradation.
Collapse
|
41
|
Nair-Gupta P, Baccarini A, Tung N, Seyffer F, Florey O, Huang Y, Banerjee M, Overholtzer M, Roche PA, Tampé R, Brown BD, Amsen D, Whiteheart SW, Blander JM. TLR signals induce phagosomal MHC-I delivery from the endosomal recycling compartment to allow cross-presentation. Cell 2015; 158:506-21. [PMID: 25083866 DOI: 10.1016/j.cell.2014.04.054] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 01/08/2014] [Accepted: 04/23/2014] [Indexed: 11/16/2022]
Abstract
Adaptation of the endoplasmic reticulum (ER) pathway for MHC class I (MHC-I) presentation in dendritic cells enables cross-presentation of peptides derived from phagocytosed microbes, infected cells, or tumor cells to CD8 T cells. How these peptides intersect with MHC-I molecules remains poorly understood. Here, we show that MHC-I selectively accumulate within phagosomes carrying microbial components, which engage Toll-like receptor (TLR) signaling. Although cross-presentation requires Sec22b-mediated phagosomal recruitment of the peptide loading complex from the ER-Golgi intermediate compartment (ERGIC), this step is independent of TLR signaling and does not deliver MHC-I. Instead, MHC-I are recruited from an endosomal recycling compartment (ERC), which is marked by Rab11a, VAMP3/cellubrevin, and VAMP8/endobrevin and holds large reserves of MHC-I. While Rab11a activity stocks ERC stores with MHC-I, MyD88-dependent TLR signals drive IκB-kinase (IKK)2-mediated phosphorylation of phagosome-associated SNAP23. Phospho-SNAP23 stabilizes SNARE complexes orchestrating ERC-phagosome fusion, enrichment of phagosomes with ERC-derived MHC-I, and subsequent cross-presentation during infection.
Collapse
Affiliation(s)
- Priyanka Nair-Gupta
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alessia Baccarini
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Navpreet Tung
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fabian Seyffer
- Institute of Biochemistry, Biocenter, Cluster of Excellence-Macromolecular Complexes, Goethe-University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
| | - Oliver Florey
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yunjie Huang
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Meenakshi Banerjee
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paul A Roche
- Experimental Cell Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Cluster of Excellence-Macromolecular Complexes, Goethe-University Frankfurt, Max-von-Laue Strasse 9, 60438 Frankfurt am Main, Germany
| | - Brian D Brown
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derk Amsen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Plesmanlaan 125, 1066CX Amsterdam, the Netherlands
| | - Sidney W Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - J Magarian Blander
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
42
|
Liu Z, Roche PA. Macropinocytosis in phagocytes: regulation of MHC class-II-restricted antigen presentation in dendritic cells. Front Physiol 2015; 6:1. [PMID: 25688210 PMCID: PMC4311620 DOI: 10.3389/fphys.2015.00001] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/05/2015] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) are outstanding antigen presenting cells (APCs) due to their robust ability to internalize extracellular antigens using endocytic processes such as receptor-mediated endocytosis, phagocytosis, and macropinocytosis. Macropinocytosis mediates the non-specific uptake of soluble antigens and occurs in DCs constitutively. Macropinocytosis plays a key role in DC-mediated antigen presentation to T cells against pathogens and the efficiency of macropinocytosis in antigen capture is regulated during the process of DC maturation. Here, we review the methods to study macropinocytosis, describe our current knowledge of the regulatory mechanisms of antigen uptake via macropinocytosis and the intracellular trafficking route followed by macropinocytosed antigens, and discuss the significance of macropinocytosis for DC function.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health Bethesda, MD, USA
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
43
|
Kobayashi M, Sakabe T, Chiba A, Nakajima A, Okamoto M, Shimodaira S, Yonemitsu Y, Shibamoto Y, Suzuki N, Nagaya M. Therapeutic effect of intratumoral injections of dendritic cells for locally recurrent gastric cancer: a case report. World J Surg Oncol 2014; 12:390. [PMID: 25526950 PMCID: PMC4320508 DOI: 10.1186/1477-7819-12-390] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 12/03/2014] [Indexed: 01/17/2023] Open
Abstract
An 80-year-old man with a history of gastric cancer and pulmonary emphysema underwent a distal gastrectomy for gastric cancer in 1997. In 2010, an endoscopic examination revealed a depressed-type lesion at the oral side of the anastomosis, which was diagnosed as signet-ring adenocarcinoma. Surgical management was considered, but was rejected because of obstructive and restrictive respiratory events. Chemotherapy was terminated because of adverse events. Endoscopy was used to administer intratumoral injections of dendritic cells (DCs) targeting synthesized peptides of Wilms tumor 1 (WT1) and mucin 1, cell-surface associated (MUC1). An immunohistochemical analysis of the tumor samples indicated positivity for WT1 and MUC1. One month after seven cycles of DC had been administered (between November 2010 and April 2011), no suspicious lesions were evident, and his biopsy results were normal. The patient has been in remission for 30 months. Intratumoral injections of DCs showed therapeutic effects in this patient, who could not undergo endoscopic submucosal dissection or surgery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Masaki Nagaya
- Seren Clinic Tokyo, Isokai, 2-10-2 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan.
| | | |
Collapse
|
44
|
Vega-Ramos J, Roquilly A, Zhan Y, Young LJ, Mintern JD, Villadangos JA. Inflammation Conditions Mature Dendritic Cells To Retain the Capacity To Present New Antigens but with Altered Cytokine Secretion Function. THE JOURNAL OF IMMUNOLOGY 2014; 193:3851-9. [DOI: 10.4049/jimmunol.1303215] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
45
|
Izquierdo-Useros N, Lorizate M, McLaren PJ, Telenti A, Kräusslich HG, Martinez-Picado J. HIV-1 capture and transmission by dendritic cells: the role of viral glycolipids and the cellular receptor Siglec-1. PLoS Pathog 2014; 10:e1004146. [PMID: 25033082 PMCID: PMC4102576 DOI: 10.1371/journal.ppat.1004146] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) are essential in order to combat invading viruses and trigger antiviral responses. Paradoxically, in the case of HIV-1, DCs might contribute to viral pathogenesis through trans-infection, a mechanism that promotes viral capture and transmission to target cells, especially after DC maturation. In this review, we highlight recent evidence identifying sialyllactose-containing gangliosides in the viral membrane and the cellular lectin Siglec-1 as critical determinants for HIV-1 capture and storage by mature DCs and for DC-mediated trans-infection of T cells. In contrast, DC-SIGN, long considered to be the main receptor for DC capture of HIV-1, plays a minor role in mature DC-mediated HIV-1 capture and trans-infection.
Collapse
Affiliation(s)
- Nuria Izquierdo-Useros
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- * E-mail: (NIU); (HGK); (JMP)
| | - Maier Lorizate
- Unidad de Biofisica (CSIC-UPV/EHU) and Departamento de Bioquímica, Universidad del Pais Vasco, Bilbao, Spain
| | - Paul J. McLaren
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Amalio Telenti
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Universitätsklinikum Heidelberg, Heidelberg, Germany
- * E-mail: (NIU); (HGK); (JMP)
| | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
- Universitat de Vic–Universitat Central de Catalunya (UVic-UCC), Vic, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- * E-mail: (NIU); (HGK); (JMP)
| |
Collapse
|
46
|
Abstract
Despite significant effort, the development of effective vaccines inducing strong and durable T-cell responses against intracellular pathogens and cancer cells has remained a challenge. The initiation of effector CD8+ T-cell responses requires the presentation of peptides derived from internalized antigen on class I major histocompatibility complex molecules by dendritic cells (DCs) in a process called cross-presentation. A current strategy to enhance the effectiveness of vaccination is to deliver antigens directly to DCs. This is done via selective targeting of antigen using monoclonal antibodies directed against endocytic receptors on the surface of the DCs. In this review, we will discuss considerations relevant to the design of such vaccines: the existence of DC subsets with specialized functions, the impact of the antigen intracellular trafficking on cross-presentation, and the influence of maturation signals received by DCs on the outcome of the immune response.
Collapse
Affiliation(s)
- Lillian Cohn
- Laboratory of Molecular Immunology, Rockefeller University , New York, NY , USA
| | | |
Collapse
|
47
|
McNally A, McNally M, Galea R, Thomas R, Steptoe RJ. Immunogenic, but not steady-state, antigen presentation permits regulatory T-cells to control CD8+ T-cell effector differentiation by IL-2 modulation. PLoS One 2014; 9:e85455. [PMID: 24454872 PMCID: PMC3890313 DOI: 10.1371/journal.pone.0085455] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 12/04/2013] [Indexed: 12/13/2022] Open
Abstract
Absorption of IL-2 is one proposed mechanism of CD4+CD25+FoxP3+ regulatory T cell (Treg) suppression. Direct in vivo experimental evidence for this has recently been obtained. While modulation of IL-2 bioavailability controls CD8+ T-cell effector differentiation under strongly immunogenic conditions it is not known whether Treg modulate CD8+ T cell responses through this mechanism under steady-state conditions. Here we assess this using a mouse model in which dendritic cells (DC) are manipulated to present cognate antigen to CD8+ T cells either in the steady-state or after activation. Our observations show that Treg exert a check on expansion and effector differentiation of CD8+ T cells under strongly immunogenic conditions associated with TLR ligand activation of DC, and this is mediated by limiting IL-2 availability. In contrast, when DC remain unactivated, depletion of Treg has little apparent effect on effector differentiation or IL-2 homeostasis. We conclude that while modulation of IL-2 homeostasis is an important mechanism through which Treg control CD8+ effector differentiation under immunogenic conditions, this mechanism plays little role in modulating CD8+ T-cell differentiation under steady-state conditions.
Collapse
Affiliation(s)
- Alice McNally
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Michael McNally
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Ryan Galea
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Ranjeny Thomas
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Raymond J. Steptoe
- UQ Diamantina Institute, University of Queensland, Brisbane, Australia
- * E-mail:
| |
Collapse
|
48
|
Weng TY, Huang SS, Yen MC, Lin CC, Chen YL, Lin CM, Chen WC, Wang CY, Chang JY, Lai MD. A novel cancer therapeutic using thrombospondin 1 in dendritic cells. Mol Ther 2013; 22:292-302. [PMID: 24127010 DOI: 10.1038/mt.2013.236] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 09/27/2013] [Indexed: 12/12/2022] Open
Abstract
Induction of thrombospondin 1 (TSP-1) is generally assumed to suppress tumor growth through inhibiting angiogenesis; however, it is less clear how TSP-1 in dendritic cells (DCs) influences tumor progression. We investigated tumor growth and immune mechanism by downregulation of TSP-1 in dendritic cells. Administration of TSP-1 small hairpin RNA (shRNA) through the skin produced anticancer therapeutic effects. Tumor-infiltrating CD4(+) and CD8(+) T cells were increased after the administration of TSP-1 shRNA. The expression of interleukin-12 and interferon-γ in the lymph nodes was enhanced by injection of TSP-1 shRNA. Lymphocytes from the mice injected with TSP-1 shRNA selectively killed the tumor cells, and the cytotoxicity of lymphocytes was abolished by depletion of CD8(+) T cells. Injection of CD11c(+) TSP-1-knockout (TSP-1-KO) bone marrow-derived DCs (BMDCs) delayed tumor growth in tumor-bearing mice. Similarly, antitumor activity induced by TSP-1-KO BMDCs was abrogated by depletion of CD8(+) T cells. In contrast, the administration of shRNAs targeting TSP-2, another TSP family member, did not extend the survival of tumor-bearing mice. Finally, TSP-1 shRNA functioned as an immunotherapeutic adjuvant to augment the therapeutic efficacy of Neu DNA vaccination. Collectively, the downregulation of TSP-1 in DCs produces an effective antitumor response that is opposite to the protumor effects by silencing of TSP-1 within tumor cells.
Collapse
Affiliation(s)
- Tzu-Yang Weng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Shien Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Chi Yen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ling Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chiu-Mei Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Ching Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jang-Yang Chang
- National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
49
|
Wagner CS, Grotzke J, Cresswell P. Intracellular regulation of cross-presentation during dendritic cell maturation. PLoS One 2013; 8:e76801. [PMID: 24098562 PMCID: PMC3789698 DOI: 10.1371/journal.pone.0076801] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/03/2013] [Indexed: 11/19/2022] Open
Abstract
We have investigated the effect of different maturation stimuli on the ability of mature dendritic cells (DCs) to cross-present newly acquired particulate antigens. Cross-presentation was impaired in DCs matured by treatment with TNF-α, CpG and LPS, but was less affected upon CD40L-induced maturation. The difference could not be explained by decreased antigen uptake or translocation into the cytosol, but decreased cross-presentation ability did correlate with increased phagosomal/lysosomal acidification. Nevertheless, intra-phagosomal degradation of OVA was not increased in matured samples, suggesting that decreasing phagosomal pH may also regulate cross-presentation by a mechanism other than enhancing degradation.
Collapse
Affiliation(s)
- Claudia S. Wagner
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jeff Grotzke
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Peter Cresswell
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
50
|
Vega-Ramos J, Villadangos JA. Consequences of direct and indirect activation of dendritic cells on antigen presentation: Functional implications and clinical considerations. Mol Immunol 2013. [DOI: 10.1016/j.molimm.2012.10.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|