Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4841, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844783

ABSTRACT

Kaposi sarcoma associated herpesvirus (KSHV) is associated with around 1% of all human tumors, including the B cell malignancy primary effusion lymphoma (PEL), in which co-infection with the Epstein Barr virus (EBV) can almost always be found in malignant cells. Here, we demonstrate that KSHV/EBV co-infection of mice with reconstituted human immune systems (humanized mice) leads to IgM responses against both latent and lytic KSHV antigens, and expansion of central and effector memory CD4+ and CD8+ T cells. Among these, KSHV/EBV dual-infection allows for the priming of CD8+ T cells that are specific for the lytic KSHV antigen K6 and able to kill KSHV/EBV infected B cells. This suggests that K6 may represent a vaccine antigen for the control of KSHV and its associated pathologies in high seroprevalence regions, such as Sub-Saharan Africa.


Subject(s)
B-Lymphocytes , CD8-Positive T-Lymphocytes , Herpesvirus 8, Human , Animals , Herpesvirus 8, Human/immunology , Humans , B-Lymphocytes/immunology , Mice , CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Coinfection/immunology , Coinfection/virology , CD4-Positive T-Lymphocytes/immunology , Herpesvirus 4, Human/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immunoglobulin M/immunology , Antigens, Viral/immunology , Mice, SCID , Lymphoma, Primary Effusion/immunology , Lymphoma, Primary Effusion/virology , Antibodies, Viral/immunology
2.
Angew Chem Int Ed Engl ; 62(15): e202300785, 2023 Apr 03.
Article in English | MEDLINE | ID: mdl-36779363

ABSTRACT

The incorporation of heteroatoms into hydrocarbon compounds greatly expands the chemical space of molecular materials. In this context, B-N doping takes a center stage due to its isosterism with a C=C-bond. Herein, we present a new and modular synthetic concept to access novel diazadiborabenzo[b]triphenylenes 7 a-h using the B-N doped biradical 16 as intermediate. Characterization of the photophysical properties revealed the emission spectra of the diazadibora benzo[b]triphenylenes 7 a-h can conveniently be tuned by small changes of the substitution on the boron-atom. All of the diazadibora compounds show a short life-time phosphorescence. Additionally, we were able to rationalize the excited-state relaxation of the diazadiboraacene 7 a via intersystem crossing by quantum chemical calculations. The new synthetic strategy provides an elegant route to various novel B-N doped acenes with great potential for applications in molecular materials.

3.
Eur J Immunol ; 52(11): 1829-1841, 2022 11.
Article in English | MEDLINE | ID: mdl-34932821

ABSTRACT

Human skin is a preferred vaccination site as it harbors multiple dendritic cell (DC) subsets, which display distinct C-type lectin receptors (CLR) that recognize pathogens. Antigens can be delivered to CLR by antibodies or ligands to boost antigen-specific immune responses. This concept has been established in mouse models but detailed insights into the functional consequences of antigen delivery to human skin DC in situ are sparse. In this study, we cloned and produced an anti-human Langerin antibody conjugated to the EBV nuclear antigen 1 (EBNA1). We confirmed specific binding of anti-Langerin-EBNA1 to Langerhans cells (LC). This novel LC-based vaccine was then compared to an existing anti-DEC-205-EBNA1 fusion protein by loading LC in epidermal cell suspensions before coculturing them with autologous T cells. After restimulation with EBNA1-peptides, we detected elevated levels of IFN-γ- and TNF-α-positive CD4+ T cells with both vaccines. When we injected the fusion proteins intradermally into human skin explants, emigrated skin DC targeted via DEC-205-induced cytokine production by T cells, whereas the Langerin-based vaccine failed to do so. In summary, we demonstrate that antibody-targeting approaches via the skin are promising vaccination strategies, however, further optimizations of vaccines are required to induce potent immune responses.


Subject(s)
Dendritic Cells , Langerhans Cells , Lectins, C-Type , Vaccines , Animals , Humans , Mice , Antigens/metabolism , Langerhans Cells/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins , Skin
5.
Org Lett ; 23(6): 2089-2093, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33629862

ABSTRACT

The combination of a Lewis acid-catalyzed inverse electron-demand Diels-Alder (IEDDA) reaction with a photoinduced ring-opening (PIRO) reaction in a domino process has been established as an efficient synthetic method to access medium-sized carbocycles. From readily available electron-rich and electron-poor phthalazines and enamines, respectively, as starting materials, various 9- and 11-membered carbocycles were prepared. This versatile transition-metal-free tool will be valuable for broadening the structural space in biologically active compounds and functional materials.

6.
Eur J Immunol ; 51(1): 64-75, 2021 01.
Article in English | MEDLINE | ID: mdl-32949466

ABSTRACT

Immune responses to Epstein-Barr virus (EBV) infection synergize with the main genetic risk factor HLA-DRB1*15:01 (HLA-DR15) to increase the likelihood to develop the autoimmune disease multiple sclerosis (MS) at least sevenfold. In order to gain insights into this synergy, we investigated HLA-DR15 positive human immune compartments after reconstitution in immune-compromised mice (humanized mice) with and without EBV infection. We detected elevated activation of both CD4+ and CD8+ T cells in HLA-DR15 donor-reconstituted humanized mice at steady state, even when compared to immune compartments carrying HLA-DRB1*04:01 (HLA-DR4), which is associated with other autoimmune diseases. Increased CD8+ T cell expansion and activation was also observed in HLA-DR15 donor-reconstituted humanized mice after EBV infection. Despite this higher immune activation, EBV viral loads were less well controlled in the context of HLA-DR15. Indeed, HLA-DR15-restricted CD4+ T cell clones recognized EBV-transformed B cell lines less efficiently and demonstrated cross-reactivity toward allogeneic target cells and one MS autoantigen. These findings suggest that EBV as one of the main environmental risk factors and HLA-DR15 as the main genetic risk factor for MS synergize by priming hyperreactive T-cell compartments, which then control the viral infection less efficiently and contain cross-reactive CD4+ T cell clones.


Subject(s)
Epstein-Barr Virus Infections/immunology , HLA-DR Serological Subtypes/immunology , Multiple Sclerosis/immunology , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Reactions , Disease Models, Animal , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Genetic Predisposition to Disease , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-DR Serological Subtypes/genetics , Herpesvirus 4, Human/immunology , Humans , Isoantigens , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Multiple Sclerosis/etiology , Multiple Sclerosis/genetics , Myelin Sheath/immunology , Risk Factors
7.
Life Sci Alliance ; 3(8)2020 08.
Article in English | MEDLINE | ID: mdl-32576602

ABSTRACT

HIV and EBV are human pathogens that cause a considerable burden to worldwide health. In combination, these viruses are linked to AIDS-associated lymphomas. We found that EBV, which transforms B cells, renders them susceptible to HIV-1 infection in a CXCR4 and CD4-dependent manner in vitro and that CXCR4-tropic HIV-1 integrates into the genome of these B cells with the same molecular profile as in autologous CD4+ T cells. In addition, we established a humanized mouse model to investigate the in vivo interactions of EBV and HIV-1 upon coinfection. The respective mice that reconstitute human immune system components upon transplantation with CD34+ human hematopoietic progenitor cells could recapitulate aspects of EBV and HIV immunobiology observed in dual-infected patients. Upon coinfection of humanized mice, EBV/HIV dual-infected B cells could be detected, but were susceptible to CD8+ T-cell-mediated immune control.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , Herpesvirus 4, Human/pathogenicity , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , CD4 Antigens/immunology , CD4 Antigens/metabolism , Coinfection , Disease Models, Animal , Disease Susceptibility/metabolism , Disease Susceptibility/virology , Epstein-Barr Virus Infections/immunology , HIV Infections/genetics , HIV Seropositivity , HIV-1/metabolism , HIV-1/pathogenicity , Hematopoietic Stem Cells/pathology , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Receptors, CXCR4/metabolism , Receptors, CXCR4/physiology , T-Lymphocytes/immunology
8.
Cell Mol Life Sci ; 77(21): 4315-4324, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32367191

ABSTRACT

Epstein-Barr virus (EBV) was the first human tumor virus being discovered and remains to date the only human pathogen that can transform cells in vitro. 55 years of EBV research have now brought us to the brink of an EBV vaccine. For this purpose, recombinant viral vectors and their heterologous prime-boost vaccinations, EBV-derived virus-like particles and viral envelope glycoprotein formulations are explored and are discussed in this review. Even so, cell-mediated immune control by cytotoxic lymphocytes protects healthy virus carriers from EBV-associated malignancies, antibodies might be able to prevent symptomatic primary infection, the most likely EBV-associated pathology against which EBV vaccines will be initially tested. Thus, the variety of EBV vaccines reflects the sophisticated life cycle of this human tumor virus and only vaccination in humans will finally be able to reveal the efficacy of these candidates. Nevertheless, the recently renewed efforts to develop an EBV vaccine and the long history of safe adoptive T cell transfer to treat EBV-associated malignancies suggest that this oncogenic γ-herpesvirus can be targeted by immunotherapies. Such vaccination should ideally implement the very same immune control that protects healthy EBV carriers.


Subject(s)
Epstein-Barr Virus Infections/prevention & control , Herpesvirus 4, Human/immunology , Herpesvirus Vaccines/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus Vaccines/immunology , Humans , Vaccination , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/therapeutic use , Viral Envelope Proteins/immunology , Viral Envelope Proteins/therapeutic use
10.
J Clin Invest ; 129(5): 2071-2087, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31042161

ABSTRACT

The Epstein-Barr virus (EBV) is one of the predominant tumor viruses in humans, but so far no therapeutic or prophylactic vaccination against this transforming pathogen is available. We demonstrated that heterologous prime-boost vaccination with the nuclear antigen 1 of EBV (EBNA1), either targeted to the DEC205 receptor on DCs or expressed from a recombinant modified vaccinia virus Ankara (MVA) vector, improved priming of antigen-specific CD4+ T cell help. This help supported the expansion and maintenance of EBNA1-specific CD8+ T cells that are most efficiently primed by recombinant adenoviruses that encode EBNA1. These combined CD4+ and CD8+ T cell responses protected against EBNA1-expressing T and B cell lymphomas, including lymphoproliferations that emerged spontaneously after EBNA1 expression. In particular, the heterologous EBNA1-expressing adenovirus, boosted by EBNA1-encoding MVA vaccination, demonstrated protection as a prophylactic and therapeutic treatment for the respective lymphoma challenges. Our study shows that such heterologous prime-boost vaccinations against EBV-associated malignancies as well as symptomatic primary EBV infection should be further explored for clinical development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Nuclear Antigens/immunology , Lymphoma/therapy , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Epstein-Barr Virus Infections/complications , Genetic Vectors , HEK293 Cells , Herpesvirus 4, Human , Humans , Immunoglobulin G/chemistry , Interferon-gamma/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphoma/immunology , Lymphoma/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Treatment Outcome , Vaccination , Vaccinia virus/immunology
11.
Org Lett ; 21(11): 3927-3930, 2019 Jun 07.
Article in English | MEDLINE | ID: mdl-31079462

ABSTRACT

A bidentate Lewis acid catalyzed domino inverse-electron-demand Diels-Alder reaction combined with a photoinduced ring opening formally inserts o-xylene moieties into enamine double bonds. After reduction, phenethylamines were obtained in good yields. The scope of the reaction was determined by variation of all three starting compounds: phthalazines, aldehydes, and amines.

12.
mBio ; 10(1)2019 01 15.
Article in English | MEDLINE | ID: mdl-30647153

ABSTRACT

The human persistent and oncogenic Epstein-Barr virus (EBV) was one of the first viruses that were described to express viral microRNAs (miRNAs). These have been proposed to modulate many host and viral functions, but their predominant role in vivo has remained unclear. We compared recombinant EBVs expressing or lacking miRNAs during in vivo infection of mice with reconstituted human immune system components and found that miRNA-deficient EBV replicates to lower viral titers with decreased frequencies of proliferating EBV-infected B cells. In response, activated cytotoxic EBV-specific T cells expand to lower frequencies than during infection with miRNA-expressing EBV. However, when we depleted CD8+ T cells the miRNA-deficient virus reached similar viral loads as wild-type EBV, increasing by more than 200-fold in the spleens of infected animals. Furthermore, CD8+ T cell depletion resulted in lymphoma formation in the majority of animals after miRNA-deficient EBV infection, while no tumors emerged when CD8+ T cells were present. Thus, miRNAs mainly serve the purpose of immune evasion from T cells in vivo and could become a therapeutic target to render EBV-associated malignancies more immunogenic.IMPORTANCE Epstein-Barr virus (EBV) infects the majority of the human population and usually persists asymptomatically within its host. Nevertheless, EBV is the causative agent for infectious mononucleosis (IM) and for lymphoproliferative disorders, including Burkitt and Hodgkin lymphomas. The immune system of the infected host is thought to prevent tumor formation in healthy virus carriers. EBV was one of the first viruses described to express miRNAs, and many host and viral targets were identified for these in vitro However, their role during EBV infection in vivo remained unclear. This work is the first to describe that EBV miRNAs mainly increase viremia and virus-associated lymphomas through dampening antigen recognition by adaptive immune responses in mice with reconstituted immune responses. Currently, there is no prophylactic or therapeutic treatment to restrict IM or EBV-associated malignancies; thus, targeting EBV miRNAs could promote immune responses and limit EBV-associated pathologies.


Subject(s)
Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Immune Evasion , MicroRNAs/metabolism , RNA, Viral/metabolism , T-Lymphocytes/immunology , Animals , B-Lymphocytes/virology , Disease Models, Animal , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Gene Deletion , Humans , Mice , Mice, Transgenic , MicroRNAs/genetics , RNA, Viral/genetics , Viral Load
13.
Angew Chem Int Ed Engl ; 58(13): 4259-4263, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30657239

ABSTRACT

We present a new concept to control the conformations of molecules in the excited state through harvesting negative hyperconjugation. The strategy was realized with the 2,3,1,4-benzodiazadiborinane scaffold, which was prepared by a new synthetic procedure. Photochemical studies identified dual light emission, which was assigned to well-defined conformers. The emission at longer wavelength can be switched off by restricting the rotational degrees of freedom in the solid state as well as by controlling the energy levels of the excited states through adjusting the solvent polarity.

14.
Clin Cancer Res ; 24(16): 3981-3993, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29748183

ABSTRACT

Purpose: Combination therapy of adoptively transferred redirected T cells and checkpoint inhibitors aims for higher response rates in tumors poorly responsive to immunotherapy like malignant pleural mesothelioma (MPM). Only most recently the issue of an optimally active chimeric antigen receptor (CAR) and the combination with checkpoint inhibitors is starting to be addressed.Experimental Design: Fibroblast activation protein (FAP)-specific CARs with different costimulatory domains, including CD28, Δ-CD28 (lacking lck binding moiety), or 4-1BB were established. CAR-T cells were characterized in vitro and antitumor efficacy was tested in vivo in a humanized mouse model in combination with PD-1 blockade. Finally, the Δ-CD28 CAR was tested clinically in a patient with MPM.Results: All the three CARs demonstrated FAP-specific functionality in vitro Gene expression data indicated a distinct activity profile for the Δ-CD28 CAR, including higher expression of genes involved in cell division, glycolysis, fatty acid oxidation, and oxidative phosphorylation. In vivo, only T cells expressing the Δ-CD28 CAR in combination with PD-1 blockade controlled tumor growth. When injected into the pleural effusion of a patient with MPM, the Δ-CD28 CAR could be detected for up to 21 days and showed functionality.Conclusions: Overall, anti-FAP-Δ-CD28/CD3ζ CAR T cells revealed superior in vitro functionality, better tumor control in combination with PD-1 blockade in humanized mice, and persistence up to 21 days in a patient with MPM. Therefore, further clinical investigation of this optimized CAR is warranted. Clin Cancer Res; 24(16); 3981-93. ©2018 AACR.


Subject(s)
Gelatinases/genetics , Lung Neoplasms/therapy , Membrane Proteins/genetics , Mesothelioma/therapy , Pleural Neoplasms/therapy , Programmed Cell Death 1 Receptor/genetics , Serine Endopeptidases/genetics , Adult , Aged , Animals , CD28 Antigens/immunology , CD28 Antigens/therapeutic use , Endopeptidases , Female , Gelatinases/immunology , Humans , Immunotherapy, Adoptive , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Male , Membrane Proteins/immunology , Mesothelioma/genetics , Mesothelioma/immunology , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Middle Aged , Oxidative Phosphorylation , Pleural Neoplasms/genetics , Pleural Neoplasms/immunology , Pleural Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Serine Endopeptidases/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...