1
|
Bruun N, Laursen MF, Carmelo R, Christensen E, Jensen TS, Christiansen G, Birkelund S, Agger R, Kofod-Olsen E. Novel nucleotide-packaging vaccine delivers antigen and poly(I:C) to dendritic cells and generate a potent antibody response in vivo. Vaccine 2024; 42:2909-2918. [PMID: 38538405 DOI: 10.1016/j.vaccine.2024.03.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/10/2024] [Accepted: 03/22/2024] [Indexed: 04/16/2024]
Abstract
An issue with many current vaccines is the dependency on broadly inflammatory adjuvants, such as aluminum hydroxide or aluminum salts that affect many immune- and non-immune cells. These adjuvants are not necessarily activating all antigen-presenting cells (APCs) that take up the antigen and most likely they also activate APCs with no antigen uptake, as well as many non-immune cells. Conjugation of antigen and adjuvant would enable the use of smaller amounts of adjuvant and avoid unnecessary tissue damage and activation of bystander cells. It would ensure that all APCs that take up the antigen would also become activated and avoid that immature and non-activated APCs present the antigen to T cells without a co-stimulatory signal, leading to tolerogenesis. We have developed a novel vaccine that co-deliver antigen and a nucleotide adjuvant to the same APC and lead to a strong activation response in dendritic cells and macrophages. The vaccine is constructed as a fusion-protein with an antigen fused to the DNA/RNA-binding domain from the Hc2 protein from Chlamydia trachomatis. We have found that the fusion protein is able to package polyinosinic:polycytidylic acid (poly(I:C)) or dsDNA into small particles. These particles were taken up by macrophages and dendritic cells and led to strong activation and maturation of these cells. Immunization of mice with the fusion protein packaged poly(I:C) led to a stronger antibody response compared to immunization with a combination of poly(I:C) and antigen without the Hc2 DNA/RNA-binding domain.
Collapse
Affiliation(s)
- Natasja Bruun
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Marlene F Laursen
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Rita Carmelo
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Esben Christensen
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Trine S Jensen
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Gunna Christiansen
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Svend Birkelund
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Ralf Agger
- Aalborg University, Department of Health Science and Technology, Denmark
| | - Emil Kofod-Olsen
- Aalborg University, Department of Health Science and Technology, Denmark.
| |
Collapse
|
2
|
Komiya E, Takamatsu S, Miura D, Tsukakoshi K, Tsugawa W, Sode K, Ikebukuro K, Asano R. Exploration and Application of DNA-Binding Proteins to Make a Versatile DNA-Protein Covalent-Linking Patch (D-Pclip): The Case of a Biosensing Element. J Am Chem Soc 2024; 146:4087-4097. [PMID: 38295327 PMCID: PMC10870700 DOI: 10.1021/jacs.3c12668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/02/2024]
Abstract
DNA-protein complexes are attractive components with broad applications in various research fields, such as DNA aptamer-enzyme complexes as biosensing elements. However, noncovalent DNA-protein complexes often decrease detection sensitivity because they are highly susceptible to environmental conditions. In this study, we developed a versatile DNA-protein covalent-linking patch (D-Pclip) for fabricating covalent and stoichiometric DNA-protein complexes. We comprehensively explored the database to determine the DNA-binding ability of the candidates and selected UdgX as the only uracil-DNA glycosylase known to form covalent bonds with DNA via uracil, with a binding efficiency >90%. We integrated a SpyTag/SpyCatcher protein-coupling system into UdgX to create a universal and convenient D-Pclip. The usability of D-Pclip was shown by preparing a stoichiometric model complex of a hemoglobin (Hb)-binding aptamer and glucose oxidase (GOx) by mixing at 4 °C. The prepared aptamer-GOx complexes detected Hb in a dose-dependent manner within the clinically required detection range in buffer and human serum without any washing procedures. D-Pclip covalently connects any uracil-inserted DNA sequence and any SpyCatcher-fused protein stoichiometrically; therefore, it has a high potential for various applications.
Collapse
Affiliation(s)
- Erika Komiya
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Shouhei Takamatsu
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Daimei Miura
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kaori Tsukakoshi
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Wakako Tsugawa
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Koji Sode
- Joint
Department of Biomedical Engineering, University
of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27599, United States
- Institute
of Global Innovation Research, Tokyo University
of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu, Tokyo 183-8509, Japan
| | - Kazunori Ikebukuro
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Ryutaro Asano
- Department
of Biotechnology and Life Science, Tokyo
University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
- Institute
of Global Innovation Research, Tokyo University
of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu, Tokyo 183-8509, Japan
| |
Collapse
|
3
|
Zhou JY, Sarkar MK, Okamura K, Harris JE, Gudjonsson JE, Fitzgerald KA. Activation of the NLRP1 inflammasome in human keratinocytes by the dsDNA mimetic poly(dA:dT). Proc Natl Acad Sci U S A 2023; 120:e2213777120. [PMID: 36693106 PMCID: PMC9945980 DOI: 10.1073/pnas.2213777120] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The accrual of cytosolic DNA leads to transcription of type I IFNs, proteolytic maturation of the IL-1 family of cytokines, and pyroptotic cell death. Caspase-1 cleaves pro-IL1β to generate mature bioactive cytokine and gasdermin D which facilitates IL-1 release and pyroptotic cell death. Absent in melanoma-2 (AIM2) is a sensor of dsDNA leading to caspase-1 activation, although in human monocytes, cGAS-STING acting upstream of NLRP3 mediates the dsDNA-activated inflammasome response. In healthy human keratinocytes, AIM2 is not expressed yet caspase-1 is activated by the synthetic dsDNA mimetic poly(dA:dT). Here, we show that this response is not mediated by either AIM2 or the cGAS-STING-NLRP3 pathway and is instead dependent on NLRP1. Poly(dA:dT) is unique in its ability to activate NLRP1, as conventional linear dsDNAs fail to elicit NLRP1 activation. DsRNA was recently shown to activate NLRP1 and prior work has shown that poly(dA:dT) is transcribed into an RNA intermediate that stimulates the RNA sensor RIG-I. However, poly(dA:dT)-dependent RNA intermediates are insufficient to activate NLRP1. Instead, poly(dA:dT) results in oxidative nucleic acid damage and cellular stress, events which activate MAP3 kinases including ZAKα that converge on p38 to activate NLRP1. Collectively, this work defines a new activator of NLRP1, broadening our understanding of sensors that recognize poly(dA:dT) and advances the understanding of the immunostimulatory potential of this potent adjuvant.
Collapse
Affiliation(s)
- Jeffrey Y. Zhou
- aDivision of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Mrinal K. Sarkar
- bDepartment of Dermatology, University of Michigan, Ann Arbor, MI48109
| | - Ken Okamura
- cDepartment of Dermatology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - John E. Harris
- cDepartment of Dermatology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | | | - Katherine A. Fitzgerald
- aDivision of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA01605
- 1To whom correspondence may be addressed.
| |
Collapse
|
4
|
Stoitzner P, Romani N, Rademacher C, Probst HC, Mahnke K. Antigen targeting to dendritic cells: Still a place in future immunotherapy? Eur J Immunol 2022; 52:1909-1924. [PMID: 35598160 PMCID: PMC10084009 DOI: 10.1002/eji.202149515] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/01/2022] [Accepted: 05/20/2022] [Indexed: 12/16/2022]
Abstract
The hallmark of DCs is their potent and outstanding capacity to activate naive resting T cells. As such, DCs are the sentinels of the immune system and instrumental for the induction of immune responses. This is one of the reasons, why DCs became the focus of immunotherapeutical strategies to fight infections, cancer, and autoimmunity. Besides the exploration of adoptive DC-therapy for which DCs are generated from monocytes or purified in large numbers from the blood, alternative approaches were developed such as antigen targeting of DCs. The idea behind this strategy is that DCs resident in patients' lymphoid organs or peripheral tissues can be directly loaded with antigens in situ. The proof of principle came from mouse models; subsequent translational studies confirmed the potential of this therapy. The first clinical trials demonstrated feasibility and the induction of T-cell immunity in patients. This review will cover: (i) the historical aspects of antigen targeting, (ii) briefly summarize the biology of DCs and the immunological functions upon which this concept rests, (iii) give an overview on attempts to target DC receptors with antibodies or (glycosylated) ligands, and finally, (iv) discuss the translation of antigen targeting into clinical therapy.
Collapse
Affiliation(s)
- Patrizia Stoitzner
- Department of Dermatology, Venereology, and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology, Venereology, and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Rademacher
- Department of Microbiology, Immunology and Genetics, University of Vienna, Vienna, Austria.,Institute of Immunology, University Medical Center Mainz, Mainz, Germany
| | - Hans Christian Probst
- Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany.,Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Kelemen E, Ádám É, Sági SM, Göblös A, Kemény L, Bata-Csörgő Z, Széll M, Danis J. Psoriasis-Associated Inflammatory Conditions Induce IL-23 mRNA Expression in Normal Human Epidermal Keratinocytes. Int J Mol Sci 2022; 23:540. [PMID: 35008970 PMCID: PMC8745281 DOI: 10.3390/ijms23010540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/14/2022] Open
Abstract
Psoriasis is a multifactorial, chronic inflammatory skin disease, the development of which is affected by both genetic and environmental factors. Cytosolic nucleic acid fragments, recognized as pathogen- and danger-associated molecular patterns, are highly abundant in psoriatic skin. It is known that psoriatic skin exhibits increased levels of IL-23 compared to healthy skin. However, the relationship between free nucleic acid levels and IL-23 expression has not been clarified yet. To examine a molecular mechanism by which nucleic acids potentially modulate IL-23 levels, an in vitro system was developed to investigate the IL-23 mRNA expression of normal human epidermal keratinocytes under psoriasis-like circumstances. This system was established using synthetic nucleic acid analogues (poly(dA:dT) and poly(I:C)). Signaling pathways, receptor involvement and the effect of PRINS, a long non-coding RNA previously identified and characterized by our research group, were analyzed to better understand the regulation of IL-23 in keratinocytes. Our results indicate that free nucleic acids regulate epithelial IL-23 mRNA expression through the TLR3 receptor and specific signaling pathways, thereby, contributing to the development of an inflammatory milieu favorable for the appearance of psoriatic symptoms. A moderate negative correlation was confirmed between the nucleic-acid-induced IL-23 mRNA level and the rate of its decrease upon PRINS overexpression.
Collapse
Affiliation(s)
- Evelyn Kelemen
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary;
| | - Éva Ádám
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary;
| | - Stella Márta Sági
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
| | - Anikó Göblös
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
| | - Márta Széll
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary;
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
| | - Judit Danis
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| |
Collapse
|
6
|
Harnessing the Complete Repertoire of Conventional Dendritic Cell Functions for Cancer Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12070663. [PMID: 32674488 PMCID: PMC7408110 DOI: 10.3390/pharmaceutics12070663] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
The onset of checkpoint inhibition revolutionized the treatment of cancer. However, studies from the last decade suggested that the sole enhancement of T cell functionality might not suffice to fight malignancies in all individuals. Dendritic cells (DCs) are not only part of the innate immune system, but also generals of adaptive immunity and they orchestrate the de novo induction of tolerogenic and immunogenic T cell responses. Thus, combinatorial approaches addressing DCs and T cells in parallel represent an attractive strategy to achieve higher response rates across patients. However, this requires profound knowledge about the dynamic interplay of DCs, T cells, other immune and tumor cells. Here, we summarize the DC subsets present in mice and men and highlight conserved and divergent characteristics between different subsets and species. Thereby, we supply a resource of the molecular players involved in key functional features of DCs ranging from their sentinel function, the translation of the sensed environment at the DC:T cell interface to the resulting specialized T cell effector modules, as well as the influence of the tumor microenvironment on the DC function. As of today, mostly monocyte derived dendritic cells (moDCs) are used in autologous cell therapies after tumor antigen loading. While showing encouraging results in a fraction of patients, the overall clinical response rate is still not optimal. By disentangling the general aspects of DC biology, we provide rationales for the design of next generation DC vaccines enabling to exploit and manipulate the described pathways for the purpose of cancer immunotherapy in vivo. Finally, we discuss how DC-based vaccines might synergize with checkpoint inhibition in the treatment of malignant diseases.
Collapse
|
7
|
Lee J, Tatsumi A, Tsukakoshi K, Wilson ED, Abe K, Sode K, Ikebukuro K. Application of a Glucose Dehydrogenase-Fused with Zinc Finger Protein to Label DNA Aptamers for the Electrochemical Detection of VEGF. SENSORS 2020; 20:s20143878. [PMID: 32664558 PMCID: PMC7411789 DOI: 10.3390/s20143878] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022]
Abstract
Aptamer-based electrochemical sensors have gained attention in the context of developing a diagnostic biomarker detection method because of their rapid response, miniaturization ability, stability, and design flexibility. In such detection systems, enzymes are often used as labels to amplify the electrochemical signal. We have focused on glucose dehydrogenase (GDH) as a labeling enzyme for electrochemical detection owing to its high enzymatic activity, availability, and well-established electrochemical principle and platform. However, it is difficult and laborious to obtain one to one labeling of a GDH-aptamer complex with conventional chemical conjugation methods. In this study, we used GDH that was genetically fused to a DNA binding protein, i.e., zinc finger protein (ZF). Fused GDH can be attached to an aptamer spontaneously and site specifically in a buffer by exploiting the sequence-specific binding ability of ZF. Using such a fusion protein, we labeled a vascular endothelial growth factor (VEGF)-binding aptamer with GDH and detected the target electrochemically. As a result, upon the addition of glucose, the GDH labeled on the aptamer generated an amperometric signal, and the current response increased dependent on the VEGF concentration. Eventually, the developed electrochemical sensor proved to detect VEGF levels as low as 105 pM, thereby successfully demonstrating the concept of using ZF-fused GDH to enzymatically label aptamers.
Collapse
Affiliation(s)
- Jinhee Lee
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA; (J.L.); (E.D.W.); (K.S.)
| | - Atsuro Tatsumi
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan; (A.T.); (K.T.); (K.A.)
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan; (A.T.); (K.T.); (K.A.)
| | - Ellie D. Wilson
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA; (J.L.); (E.D.W.); (K.S.)
| | - Koichi Abe
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan; (A.T.); (K.T.); (K.A.)
| | - Koji Sode
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA; (J.L.); (E.D.W.); (K.S.)
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan; (A.T.); (K.T.); (K.A.)
- Correspondence: ; Tel.: +81-42-388-7030
| |
Collapse
|
8
|
Baldin AV, Savvateeva LV, Bazhin AV, Zamyatnin AA. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020; 12:cancers12030590. [PMID: 32150821 PMCID: PMC7139354 DOI: 10.3390/cancers12030590] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) have shown great potential as a component or target in the landscape of cancer immunotherapy. Different in vivo and ex vivo strategies of DC vaccine generation with different outcomes have been proposed. Numerous clinical trials have demonstrated their efficacy and safety in cancer patients. However, there is no consensus regarding which DC-based vaccine generation method is preferable. A problem of result comparison between trials in which different DC-loading or -targeting approaches have been applied remains. The employment of different DC generation and maturation methods, antigens and administration routes from trial to trial also limits the objective comparison of DC vaccines. In the present review, we discuss different methods of DC vaccine generation. We conclude that standardized trial designs, treatment settings and outcome assessment criteria will help to determine which DC vaccine generation approach should be applied in certain cancer cases. This will result in a reduction in alternatives in the selection of preferable DC-based vaccine tactics in patient. Moreover, it has become clear that the application of a DC vaccine alone is not sufficient and combination immunotherapy with recent advances, such as immune checkpoint inhibitors, should be employed to achieve a better clinical response and outcome.
Collapse
Affiliation(s)
- Alexey V. Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplant Surgery, Ludwig-Maximilians University of Munich, 81377 Munich, Germany;
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (A.V.B.); (L.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Department of Cell Signaling, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +74-956-229-843
| |
Collapse
|
9
|
Gale EC, Roth GA, Smith AAA, Alcántara‐Hernández M, Idoyaga J, Appel EA. A Nanoparticle Platform for Improved Potency, Stability, and Adjuvanticity of Poly(I:C). ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Emily C. Gale
- Department of Materials Science and EngineeringStanford University Stanford CA 94305 USA
- Department of BiochemistryStanford University School of Medicine Stanford CA 94305 USA
| | - Gillie A. Roth
- Department of Materials Science and EngineeringStanford University Stanford CA 94305 USA
- Department of BioengineeringStanford University Stanford CA 94305 USA
| | - Anton A. A. Smith
- Department of Materials Science and EngineeringStanford University Stanford CA 94305 USA
| | - Marcela Alcántara‐Hernández
- Department of Microbiology and ImmunologyStanford University School of Medicine Stanford CA 94305 USA
- Program in ImmunologyStanford University School of Medicine Stanford CA 94305 USA
| | - Juliana Idoyaga
- Department of Microbiology and ImmunologyStanford University School of Medicine Stanford CA 94305 USA
- Institute for ImmunityTransplantation and InfectionStanford University School of Medicine Stanford CA 94305 USA
| | - Eric A. Appel
- Department of Materials Science and EngineeringStanford University Stanford CA 94305 USA
- Department of BioengineeringStanford University Stanford CA 94305 USA
- Institute for ImmunityTransplantation and InfectionStanford University School of Medicine Stanford CA 94305 USA
| |
Collapse
|
10
|
Liszczak G, Muir TW. Barcoding mit Nukleinsäuren: Anwendung der DNA‐Sequenzierung als molekulares Zählwerk. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201808956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Glen Liszczak
- Department of ChemistryPrinceton University Princeton NJ 08544 USA
- Aktuelle Adresse: Department of BiochemistryUT Southwestern Medical Center Dallas TX 75390 USA
| | - Tom W. Muir
- Department of ChemistryPrinceton University Princeton NJ 08544 USA
| |
Collapse
|
11
|
Liszczak G, Muir TW. Nucleic Acid-Barcoding Technologies: Converting DNA Sequencing into a Broad-Spectrum Molecular Counter. Angew Chem Int Ed Engl 2019; 58:4144-4162. [PMID: 30153374 DOI: 10.1002/anie.201808956] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Indexed: 12/17/2022]
Abstract
The emergence of high-throughput DNA sequencing technologies sparked a revolution in the field of genomics that has rippled into many branches of the life and physical sciences. The remarkable sensitivity, specificity, throughput, and multiplexing capacity that are inherent to parallel DNA sequencing have since motivated its use as a broad-spectrum molecular counter. A key aspect of extrapolating DNA sequencing to non-traditional applications is the need to append nucleic-acid barcodes to entities of interest. In this review, we describe the chemical and biochemical approaches that have enabled nucleic-acid barcoding of proteinaceous and non-proteinaceous materials and provide examples of downstream technologies that have been made possible by DNA-encoded molecules. As commercially available high-throughput sequencers were first released less than 15 years ago, we believe related applications will continue to mature and close by proposing new frontiers to support this assertion.
Collapse
Affiliation(s)
- Glen Liszczak
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA.,Present address: Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tom W Muir
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA
| |
Collapse
|
12
|
CD11c-targeted Delivery of DNA to Dendritic Cells Leads to cGAS- and STING-dependent Maturation. J Immunother 2018; 41:9-18. [PMID: 29189388 DOI: 10.1097/cji.0000000000000195] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immunotherapeutic activation of tumor-specific T cells has proven to be an interesting approach in anticancer treatment. Particularly, anti-CTLA-4 and anti-PD-1/PD-L1 treatment looks promising, and conceivably, even better clinical results might be obtained if such treatment could be combined with boosting the existing tumor-specific T-cell response. One way to achieve this could be by increasing the level of maturation of dendritic cells locally and in the draining lymph nodes. When exposed to cancer cells, dendritic cells may spontaneously mature because of danger-associated molecular patterns derived from the tumor cells. Double-stranded DNA play a particularly important role in the activation of the dendritic cells, through engagement of intracellular DNA-sensors, and signaling through the adaptor protein STING. In the present study, we have investigated the maturational response of human monocyte-derived dendritic cells (moDC) and human monocytic THP-1 cells to targeted and untargeted DNA. We used an anti-CD11c antibody conjugated with double-stranded DNA to analyze the maturation status of human moDCs, as well as maturation using a cGAS KO and STING KO THP-1 cell maturation model. We found that dendritic cells can mature after exposure to cytoplasmic double-stranded DNA delivered through CD11c-mediated endocytosis. Moreover, we show that THP-1 cells matured using IL-4, GM-CSF, and ionomycin upregulate DC-maturation markers after CD11c-targeted delivery of double-stranded DNA. This upregulation is completely abrogated in cGAS KO and STING KO cells.
Collapse
|
13
|
Yan X, Zhang H, Wang Z, Peng H, Tao J, Li XF, Chris Le X. Quantitative synthesis of protein-DNA conjugates with 1 : 1 stoichiometry. Chem Commun (Camb) 2018; 54:7491-7494. [PMID: 29917030 DOI: 10.1039/c8cc03268h] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We describe here a binding-facilitated reaction strategy, enabling quantitative conjugation of DNA to native proteins with a desirable 1 : 1 stoichiometry. The technique takes advantage of the iterative affinity interaction and covalent binding processes to achieve complete conjugation. The complete conjugation obviates the need for separation of the protein-DNA conjugates as required by other DNA-protein conjugation methods.
Collapse
Affiliation(s)
- Xiaowen Yan
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| | - Hongquan Zhang
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| | - Zhixin Wang
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| | - Hanyong Peng
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| | - Jeffrey Tao
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| | - Xing-Fang Li
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| | - X Chris Le
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G2G3, Canada.
| |
Collapse
|
14
|
Danis J, Janovák L, Gubán B, Göblös A, Szabó K, Kemény L, Bata-Csörgő Z, Széll M. Differential Inflammatory-Response Kinetics of Human Keratinocytes upon Cytosolic RNA- and DNA-Fragment Induction. Int J Mol Sci 2018. [PMID: 29518010 PMCID: PMC5877635 DOI: 10.3390/ijms19030774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Keratinocytes are non-professional immune cells contributing actively to innate immune responses partially by reacting to a wide range of molecular patterns by activating pattern recognition receptors. Cytosolic nucleotide fragments as pathogen- or self-derived trigger factors are activating inflammasomes and inducing anti-viral signal transduction pathways as well as inducing expression of inflammatory cytokines. We aimed to compare the induced inflammatory reactions in three keratinocyte cell types—normal human epidermal keratinocytes, the HaCaT cell line and the HPV-KER cell line—upon exposure to the synthetic RNA and DNA analogues poly(I:C) and poly(dA:dT) to reveal the underlying signaling events. Both agents induced the expression of interleukin-6 and tumor necrosis factor α in all cell types; however, notable kinetic and expression level differences were found. Western blot analysis revealed rapid activation of the nuclear factor κB (NF-κB), mitogen activated protein kinase and signal transducers of activator of transcription (STAT) signal transduction pathways in keratinocytes upon poly(I:C) treatment, while poly(dA:dT) induced slower activation. Inhibition of NF-κB, p38, STAT-1 and STAT-3 signaling resulted in decreased cytokine expression, whereas inhibition of mitogen-activated protein kinase kinase 1/2 (MEK1/2) signaling showed a negative feedback role in both poly(I:C)- and poly(dA:dT)-induced cytokine expression. Based on our in vitro results nucleotide fragments are able to induce inflammatory reactions in keratinocytes, but with different rate and kinetics of cytokine expression, explained by faster activation of signaling routes by poly(I:C) than poly(dA:dT).
Collapse
Affiliation(s)
- Judit Danis
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.J.); (B.G.); (A.G.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, 6720 Szeged, Hungary; (K.S.); (M.S.)
- Correspondence: ; Tel.: +36-62-54-52-78
| | - Luca Janovák
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.J.); (B.G.); (A.G.); (L.K.); (Z.B.-C.)
| | - Barbara Gubán
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.J.); (B.G.); (A.G.); (L.K.); (Z.B.-C.)
| | - Anikó Göblös
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.J.); (B.G.); (A.G.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, 6720 Szeged, Hungary; (K.S.); (M.S.)
| | - Kornélia Szabó
- MTA-SZTE Dermatological Research Group, 6720 Szeged, Hungary; (K.S.); (M.S.)
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.J.); (B.G.); (A.G.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, 6720 Szeged, Hungary; (K.S.); (M.S.)
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.J.); (B.G.); (A.G.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, 6720 Szeged, Hungary; (K.S.); (M.S.)
| | - Márta Széll
- MTA-SZTE Dermatological Research Group, 6720 Szeged, Hungary; (K.S.); (M.S.)
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
15
|
Gramespacher JA, Stevens AJ, Thompson RE, Muir TW. Improved protein splicing using embedded split inteins. Protein Sci 2018; 27:614-619. [PMID: 29226478 PMCID: PMC5818749 DOI: 10.1002/pro.3357] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 01/24/2023]
Abstract
Naturally split inteins mediate a traceless protein ligation process known as protein trans-splicing (PTS). Although frequently used in protein engineering applications, the efficiency of PTS can be reduced by the tendency of some split intein fusion constructs to aggregate; a consequence of the fragmented nature of the split intein itself or the polypeptide to which it is fused (the extein). Here, we report a strategy to help address this liability. This involves embedding the split intein within a protein sequence designed to stabilize either the intein fragment itself or the appended extein. We expect this approach to increase the scope of PTS-based protein engineering efforts.
Collapse
Affiliation(s)
| | - Adam J. Stevens
- Department of ChemistryPrinceton University, Frick Laboratory, PrincetonNew Jersey
| | - Robert E. Thompson
- Department of ChemistryPrinceton University, Frick Laboratory, PrincetonNew Jersey
| | - Tom W. Muir
- Department of ChemistryPrinceton University, Frick Laboratory, PrincetonNew Jersey
| |
Collapse
|
16
|
Frutos S, Jordan JB, Bio MM, Muir TW, Thiel OR, Vila-Perelló M. Access to site-specific Fc-cRGD peptide conjugates through streamlined expressed protein ligation. Org Biomol Chem 2018; 14:9549-9553. [PMID: 27722696 DOI: 10.1039/c6ob01833e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An ideal drug should be highly effective, non-toxic and be delivered by a convenient and painless single dose. We are still far from such optimal treatment but peptides, with their high target selectivity and low toxicity profiles, provide a very attractive platform from which to strive towards it. One of the major limitations of peptide drugs is their high clearance rates, which limit dosage regimen options. Conjugation to antibody Fc domains is a viable strategy to improve peptide stability by increasing their hydrodynamic radius and hijacking the Fc recycling pathway. We report the use of a split-intein based semi-synthetic approach to site-specifically conjugate a synthetic integrin binding peptide to an Fc domain. The strategy described here allows conjugating synthetic peptides to Fc domains, which is not possible via genetic methods, fully maintaining the ability of both the Fc domain and the bioactive peptide to interact with their binding partners.
Collapse
Affiliation(s)
- S Frutos
- ProteoDesign S.L., Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| | - J B Jordan
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - M M Bio
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - T W Muir
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - O R Thiel
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - M Vila-Perelló
- ProteoDesign S.L., Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| |
Collapse
|
17
|
van Dinther D, Stolk DA, van de Ven R, van Kooyk Y, de Gruijl TD, den Haan JMM. Targeting C-type lectin receptors: a high-carbohydrate diet for dendritic cells to improve cancer vaccines. J Leukoc Biol 2017; 102:1017-1034. [PMID: 28729358 PMCID: PMC5597514 DOI: 10.1189/jlb.5mr0217-059rr] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 12/23/2022] Open
Abstract
There is a growing understanding of why certain patients do or do not respond to checkpoint inhibition therapy. This opens new opportunities to reconsider and redevelop vaccine strategies to prime an anticancer immune response. Combination of such vaccines with checkpoint inhibitors will both provide the fuel and release the brake for an efficient anticancer response. Here, we discuss vaccine strategies that use C-type lectin receptor (CLR) targeting of APCs, such as dendritic cells and macrophages. APCs are a necessity for the priming of antigen-specific cytotoxic and helper T cells. Because CLRs are natural carbohydrate-recognition receptors highly expressed by multiple subsets of APCs and involved in uptake and processing of Ags for presentation, these receptors seem particularly interesting for targeting purposes.
Collapse
Affiliation(s)
- Dieke van Dinther
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Dorian A Stolk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Rieneke van de Ven
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Joke M M den Haan
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| |
Collapse
|
18
|
Abstract
Adjuvants have been deliberately added to vaccines since the 1920's when alum was discovered to boost antibody responses, leading to better protection. The first adjuvants were discovered by accident and were used in the safer but less immunogenic subunit vaccines, supposedly by providing an antigen depot to extend antigen presentation. Since that time, much has been discovered about how these adjuvants impact cells at the tissue site to activate innate immune responses, mobilize dendritic cells and drive adaptive immunity. New approaches to vaccine construction for infectious diseases that have so far not been well addressed by conventional vaccines often attempt to induce antibodies, polyfunctional CD4+ T cells and CD8+ CTLs. The discovery of pattern recognition receptors and ligands that drive desired T cell responses has led to development of novel adjuvant strategies using immunomodulatory agents to direct appropriate immune responses.
Collapse
Affiliation(s)
- Amy S McKee
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Philippa Marrack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Biomedical Research, National Jewish Health, 1400, Jackson St., Denver, CO 80206, USA
| |
Collapse
|
19
|
Johannssen T, Lepenies B. Glycan-Based Cell Targeting To Modulate Immune Responses. Trends Biotechnol 2016; 35:334-346. [PMID: 28277249 DOI: 10.1016/j.tibtech.2016.10.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023]
Abstract
Glycosylation is an integral post-translational modification present in more than half of all eukaryotic proteins. It affects key protein functions, including folding, stability, and immunogenicity. Glycoengineering approaches, such as the use of bacterial N-glycosylation systems, or expression systems, including yeasts, insect cells, and mammalian cells, have enabled access to defined and homogenous glycoproteins. Given that glycan structures on proteins can be recognized by host lectin receptors, they may facilitate cell-specific targeting and immune modulation. Myeloid C-type lectin receptors (CLRs) expressed by antigen-presenting cells are attractive targets to shape immune responses. Multivalent glycan display on nanoparticles, liposomes, or dendrimers has successfully enabled CLR targeting. In this review, we discuss novel strategies to access defined glycan structures and highlight CLR targeting approaches for immune modulation.
Collapse
Affiliation(s)
- Timo Johannssen
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Am Mühlenberg 1, 14476 Potsdam, Germany; Freie Universität Berlin, Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Arnimallee 22, 14195 Berlin, Germany; University of Veterinary Medicine Hannover, Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany
| | - Bernd Lepenies
- University of Veterinary Medicine Hannover, Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
20
|
Akinbobuyi B, Wang L, Upchurch KC, Byrd MR, Chang CA, Quintana JM, Petersen RE, Seifert ZJ, Boquin JR, Oh S, Kane RR. Synthesis and immunostimulatory activity of substituted TLR7 agonists. Bioorg Med Chem Lett 2016; 26:4246-9. [PMID: 27476423 DOI: 10.1016/j.bmcl.2016.07.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 11/18/2022]
Abstract
Fifteen new substituted adenines were synthesized as potential TLR7 agonists. These compounds, along with 9 previously reported compounds, were analyzed for TLR7 activity and for the selective stimulation of B cell proliferation. Several functionalized derivatives exhibit significant activity, suggesting their potential for use as vaccine adjuvants.
Collapse
Affiliation(s)
- Babatope Akinbobuyi
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Lei Wang
- Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA
| | - Katherine C Upchurch
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA; Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA
| | - Matthew R Byrd
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Charles A Chang
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA
| | - Jeremy M Quintana
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Rachel E Petersen
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Zacharie J Seifert
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - José R Boquin
- Department of Chemistry, Augustana College, 639 38th Street, Rock Island, IL 61201, USA
| | - SangKon Oh
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA; Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA
| | - Robert R Kane
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA; Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA; Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA.
| |
Collapse
|
21
|
Stevens AJ, Brown ZZ, Shah NH, Sekar G, Cowburn D, Muir TW. Design of a Split Intein with Exceptional Protein Splicing Activity. J Am Chem Soc 2016; 138:2162-5. [PMID: 26854538 PMCID: PMC4894280 DOI: 10.1021/jacs.5b13528] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein trans-splicing (PTS) by split inteins has found widespread use in chemical biology and biotechnology. Herein, we describe the use of a consensus design approach to engineer a split intein with enhanced stability and activity that make it more robust than any known PTS system. Using batch mutagenesis, we first conduct a detailed analysis of the difference in splicing rates between the Npu (fast) and Ssp (slow) split inteins of the DnaE family and find that most impactful residues lie on the second shell of the protein, directly adjacent to the active site. These residues are then used to generate an alignment of 73 naturally occurring DnaE inteins that are predicted to be fast. The consensus sequence from this alignment (Cfa) demonstrates both rapid protein splicing and unprecedented thermal and chaotropic stability. Moreover, when fused to various proteins including antibody heavy chains, the N-terminal fragment of Cfa exhibits increased expression levels relative to other N-intein fusions. The durability and efficiency of Cfa should improve current intein based technologies and may provide a platform for the development of new protein chemistry techniques.
Collapse
Affiliation(s)
- Adam J Stevens
- Department of Chemistry, Princeton University , Frick Laboratory, Princeton, New Jersey 08544, United States
| | - Zachary Z Brown
- Department of Chemistry, Princeton University , Frick Laboratory, Princeton, New Jersey 08544, United States
| | - Neel H Shah
- Department of Chemistry, Princeton University , Frick Laboratory, Princeton, New Jersey 08544, United States
| | - Giridhar Sekar
- Department of Biochemistry, Albert Einstein College of Medicine , Bronx, New York 10461, United States
| | - David Cowburn
- Department of Biochemistry, Albert Einstein College of Medicine , Bronx, New York 10461, United States
| | - Tom W Muir
- Department of Chemistry, Princeton University , Frick Laboratory, Princeton, New Jersey 08544, United States
| |
Collapse
|
22
|
Jahan ST, Haddadi A. Investigation and optimization of formulation parameters on preparation of targeted anti-CD205 tailored PLGA nanoparticles. Int J Nanomedicine 2015; 10:7371-84. [PMID: 26677326 PMCID: PMC4677653 DOI: 10.2147/ijn.s90866] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The purpose of this study was to assess the effect of various formulation parameters on anti-CD205 antibody decorated poly(d, l-lactide co-glycolide) (PLGA) nanoparticles (NPs) in terms of their ability to target dendritic cells (DCs). In brief, emulsification solvent evaporation technique was adapted to design NP formulations using two different viscosity grades (low and high) of both ester and carboxylic acid terminated PLGA. Incorporation of ligand was achieved following physical adsorption or chemical conjugation processes. The physicochemical characterizations of formulations were executed to assess the effects of different solvents (chloroform and ethyl acetate), stabilizer percentage, polymer types, polymer viscosities, ligand-NP bonding types, cross-linkers, and cryoprotectants (sucrose and trehalose). Modification of any of these parameters shows significant improvement of physicochemical properties of NPs. Ethyl acetate was the solvent of choice for the formulations to ensure better emulsion formation. Infrared spectroscopy confirmed the presence of anti-CD205 antibody in the NP formulation. Finally, cytotoxicity assay confirmed the safety profile of the NPs for DCs. Thus, ligand modified structurally concealed PLGA NPs is a promising delivery tool for targeting DCs in vivo.
Collapse
Affiliation(s)
- Sheikh Tasnim Jahan
- Division of Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Azita Haddadi
- Division of Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
23
|
pH-Dependent recognition of apoptotic and necrotic cells by the human dendritic cell receptor DEC205. Proc Natl Acad Sci U S A 2015; 112:7237-42. [PMID: 26039988 DOI: 10.1073/pnas.1505924112] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells play important roles in regulating innate and adaptive immune responses. DEC205 (CD205) is one of the major endocytotic receptors on dendritic cells and has been widely used for vaccine generation against viruses and tumors. However, little is known about its structure and functional mechanism. Here we determine the structure of the human DEC205 ectodomain by cryoelectron microscopy. The structure shows that the 12 extracellular domains form a compact double ring-shaped conformation at acidic pH and become extended at basic pH. Biochemical data indicate that the pH-dependent conformational change of DEC205 is correlated with ligand binding and release. DEC205 only binds to apoptotic and necrotic cells at acidic pH, whereas live cells cannot be recognized by DEC205 at either acidic or basic conditions. These results suggest that DEC205 is an immune receptor that recognizes apoptotic and necrotic cells specifically through a pH-dependent mechanism.
Collapse
|
24
|
Sehgal K, Dhodapkar KM, Dhodapkar MV. Targeting human dendritic cells in situ to improve vaccines. Immunol Lett 2014; 162:59-67. [PMID: 25072116 DOI: 10.1016/j.imlet.2014.07.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) provide a critical link between innate and adaptive immunity. The potent antigen presenting properties of DCs makes them a valuable target for the delivery of immunogenic cargo. Recent clinical studies describing in situ DC targeting with antibody-mediated targeting of DC receptor through DEC-205 provide new opportunities for the clinical application of DC-targeted vaccines. Further advances with nanoparticle vectors which can encapsulate antigens and adjuvants within the same compartment and be targeted against diverse DC subsets also represent an attractive strategy for targeting DCs. This review provides a brief summary of the rationale behind targeting dendritic cells in situ, the existing pre-clinical and clinical data on these vaccines and challenges faced by the next generation DC-targeted vaccines.
Collapse
Affiliation(s)
- Kartik Sehgal
- Department of Medicine, Yale University, New Haven, CT, United States
| | | | | |
Collapse
|
25
|
Template-directed covalent conjugation of DNA to native antibodies, transferrin and other metal-binding proteins. Nat Chem 2014; 6:804-9. [PMID: 25143216 DOI: 10.1038/nchem.2003] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 06/11/2014] [Indexed: 12/20/2022]
Abstract
DNA-protein conjugates are important in bioanalytical chemistry, molecular diagnostics and bionanotechnology, as the DNA provides a unique handle to identify, functionalize or otherwise manipulate proteins. To maintain protein activity, conjugation of a single DNA handle to a specific location on the protein is often needed. However, preparing such high-quality site-specific conjugates often requires genetically engineered proteins, which is a laborious and technically challenging approach. Here we demonstrate a simpler method to create site-selective DNA-protein conjugates. Using a guiding DNA strand modified with a metal-binding functionality, we directed a second DNA strand to the vicinity of a metal-binding site of His6-tagged or wild-type metal-binding proteins, such as serotransferrin, where it subsequently reacted with lysine residues at that site. This method, DNA-templated protein conjugation, facilitates the production of site-selective protein conjugates, and also conjugation to IgG1 antibodies via a histidine cluster in the constant domain.
Collapse
|
26
|
Reva IV, Reva GV, Yamamoto T, Girya OY, Grakhova NV, Maloman NY, Danilenko MV. Distribution of antigen-presenting cells CD68 in papillomavirus infection in the skin. Bull Exp Biol Med 2014; 157:56-61. [PMID: 24915947 DOI: 10.1007/s10517-014-2491-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Indexed: 10/25/2022]
Abstract
We analyzed local reactions of immune homeostasis in the human skin, in particular, effector immune cells CD68 responsible for antigen presentation, during human papillomavirus infection. Under conditions of long-term papillomavirus infection, CD68 markers were identifi ed only in the connective tissue of the skin (derma) and were completely absent in the epidermis, where they were found during physiological and reparative regeneration after thermal injury. We concluded that hypertrophy of the epidermis and connective tissue of the dermal papillary layer in human papillomavirus infection is related to the absence of CD68 immune cells in the epithelial plate and their accumulation in the connective tissue adjacent to the basement membrane of the epidermis. The possibility of epithelium contamination with the virus depends on local immune homeostasis. Therefore, induction of proper CD68 distribution in appropriate structures can contribute to normalization of epithelial-connective tissue interactions.
Collapse
Affiliation(s)
- I V Reva
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia,
| | | | | | | | | | | | | |
Collapse
|
27
|
Mancini RJ, Stutts L, Ryu KA, Tom JK, Esser-Kahn AP. Directing the immune system with chemical compounds. ACS Chem Biol 2014; 9:1075-85. [PMID: 24690004 PMCID: PMC5674983 DOI: 10.1021/cb500079s] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Agonists of immune cell receptors direct innate and adaptive immunity. These agonists range in size and complexity from small molecules to large macromolecules. Here, agonists of a class of immune cell receptors known as the Toll-like receptors (TLRs) are highlighted focusing on the distinctive molecular moieties that pertain to receptor binding and activation. How the structure and combined chemical signals translate into a variety of immune responses remain major questions in the field. In this structure-focused review, we outline potential areas where the tools of chemical biology could help decipher the emerging molecular codes that direct immune stimulation.
Collapse
Affiliation(s)
- Rock J Mancini
- Department of Chemistry, Chemical Engineering and Materials Science, and Biomedical Engineering, University of California Irvine , 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | | | | | | | | |
Collapse
|
28
|
Chiang MJ, Holbert MA, Kalin JH, Ahn YH, Giddens J, Amin MN, Taylor MS, Collins SL, Chan-Li Y, Waickman A, Hsiao PY, Bolduc D, Leahy DJ, Horton MR, Wang LX, Powell JD, Cole PA. An Fc domain protein-small molecule conjugate as an enhanced immunomodulator. J Am Chem Soc 2014; 136:3370-3. [PMID: 24533830 PMCID: PMC3954559 DOI: 10.1021/ja5006674] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Proteins as well as small molecules have demonstrated success as therapeutic agents, but their pharmacologic properties sometimes fall short against particular drug targets. Although the adenosine 2a receptor (A(2A)R) has been identified as a promising target for immunotherapy, small molecule A(2A)R agonists have suffered from short pharmacokinetic half-lives and the potential for toxicity by modulating nonimmune pathways. To overcome these limitations, we have tethered the A(2A)R agonist CGS-21680 to the immunoglobulin Fc domain using expressed protein ligation with Sf9 cell secreted protein. The protein small molecule conjugate Fc-CGS retained potent Fc receptor and A(2A)R interactions and showed superior properties as a therapeutic for the treatment of a mouse model of autoimmune pneumonitis. This approach may provide a general strategy for optimizing small molecule therapeutics.
Collapse
Affiliation(s)
- Meng-Jung Chiang
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Although major advances in the care of cancer patients over the past several decades have resulted in improved survival, infectious complications remain a significant cause of morbidity and mortality. To successfully identify, treat, and prevent infections, a comprehensive understanding of risk factors that predispose to infection and of commonly encountered pathogens is necessary. In addition, clinicians must keep abreast of the changing epidemiology of infections in this population. As therapeutic modalities continue to evolve, as established pathogens become increasingly drug resistant, and as new pathogens are discovered, successful management of infections will continue to present challenges in the years to come.
Collapse
Affiliation(s)
- Valentina Stosor
- Div. Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| | - Teresa R. Zembower
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois USA
| |
Collapse
|
30
|
Xue M, Zhu L, Meng Y, Wang L, Sun H, Wang F, Wang E, Shan F. Detailed modulation of phenotypes and functions of bone marrow dendritic cells (BMDCs) by interferon-gamma (IFN-γ). Int Immunopharmacol 2013; 17:366-72. [PMID: 23867288 DOI: 10.1016/j.intimp.2013.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/17/2013] [Accepted: 07/01/2013] [Indexed: 12/23/2022]
Abstract
IFN-γ is a cytokine that plays crucial role in innate and adaptive immunity against viral and intracellular bacterial infections and for tumor control. IFN-γ is also a key activator of macrophages [1,2]. In the present study, we studied detailed modulation of IFN-γ on phenotypic and functional maturation of murine bone marrow derived dendritic cells (BMDCs). Phenotypic and functional maturation of BMDCs was evaluated by light microscope, flow cytometry(FCM), transmission electron microscopy (TEM), cytochemistry method, acid phosphatase activity(ACP), FITC-dextran bio-assay and enzyme linked immunosorbent assay (ELISA). We elucidated that IFN-γ up-regulated the expression of MHC II, CD40, CD80, CD83 and CD86 molecules on BMDCs, down-regulated the activity of pinocytosis and phagocytosis by BMDCs, and induced higher levels of IL-12 and TNF-α secreted by BMDCs. It is therefore confirmed that IFN-γ can effectively promote the maturation of BMDCs. Our study provides more evidence and rationale on future application of IFN-γ for enhancing host immunity.
Collapse
Affiliation(s)
- Ming Xue
- Department of Endodontics, China Medical University, Shenyang, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|