Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Immunity ; 56(11): 2602-2620.e10, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37967532

ABSTRACT

Human cytomegalovirus (HCMV) can cause severe diseases in fetuses, newborns, and immunocompromised individuals. Currently, no vaccines are approved, and treatment options are limited. Here, we analyzed the human B cell response of four HCMV top neutralizers from a cohort of 9,000 individuals. By single-cell analyses of memory B cells targeting the pentameric and trimeric HCMV surface complexes, we identified vulnerable sites on the shared gH/gL subunits as well as complex-specific subunits UL128/130/131A and gO. Using high-resolution cryogenic electron microscopy, we revealed the structural basis of the neutralization mechanisms of antibodies targeting various binding sites. Moreover, we identified highly potent antibodies that neutralized a broad spectrum of HCMV strains, including primary clinical isolates, that outperform known antibodies used in clinical trials. Our study provides a deep understanding of the mechanisms of HCMV neutralization and identifies promising antibody candidates to prevent and treat HCMV infection.


Subject(s)
Cytomegalovirus , Viral Envelope Proteins , Infant, Newborn , Humans , Membrane Glycoproteins , Antibodies, Neutralizing , Memory B Cells , Antibodies, Viral , Single-Cell Analysis
2.
J Infect Dis ; 226(9): 1667-1677, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35970817

ABSTRACT

BACKGROUND: Human cytomegalovirus (HCMV) is the most common infectious complication of organ transplantation and cause of birth defects worldwide. There are limited therapeutic options and no licensed vaccine to prevent HCMV infection or disease. To inform development of HCMV antibody-based interventions, a previous study identified individuals with potent and broad plasma HCMV-neutralizing activity, termed elite neutralizers (ENs), from a cohort of HCMV-seropositive (SP) blood donors. However, the specificities and functions of plasma antibodies associated with EN status remained undefined. METHODS: We sought to determine the plasma antibody specificities, breadth, and Fc-mediated antibody effector functions associated with the most potent HCMV-neutralizing responses in plasma from ENs (n = 25) relative to that from SP donors (n = 19). We measured antibody binding against various HCMV strains and glycoprotein targets and evaluated Fc-mediated effector functions, antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP). RESULTS: We demonstrate that ENs have elevated immunoglobulin G binding responses against multiple viral glycoproteins, relative to SP donors. Our study also revealed potent HCMV-specific antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis activity of plasma from ENs. CONCLUSIONS: We conclude that antibody responses against multiple glycoprotein specificities may be needed to achieve potent plasma neutralization and that potently HCMV elite-neutralizing plasma antibodies can also mediate polyfunctional responses.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Humans , Immunoglobulin G , Antibodies, Neutralizing , Antibody Formation , Antibodies, Viral , Viral Envelope Proteins
3.
Viruses ; 13(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34578361

ABSTRACT

Cell-free human cytomegalovirus (HCMV) can be inhibited by a soluble form of the cellular HCMV-receptor PDGFRα, resembling neutralization by antibodies. The cell-associated growth of recent HCMV isolates, however, is resistant against antibodies. We investigated whether PDGFRα-derivatives can inhibit this transmission mode. A protein containing the extracellular PDGFRα-domain and 40-mer peptides derived therefrom were tested regarding the inhibition of the cell-associated HCMV strain Merlin-pAL1502, hits were validated with recent isolates, and the most effective peptide was modified to increase its potency. The modified peptide was further analyzed regarding its mode of action on the virion level. While full-length PDGFRα failed to inhibit HCMV isolates, three peptides significantly reduced virus growth. A 30-mer version of the lead peptide (GD30) proved even more effective against the cell-free virus, and this effect was HCMV-specific and depended on the viral glycoprotein O. In cell-associated spread, GD30 reduced both the number of transferred particles and their penetration. This effect was reversible after peptide removal, which allowed the synchronized analysis of particle transfer, showing that two virions per hour were transferred to neighboring cells and one virion was sufficient for infection. In conclusion, PDGFRα-derived peptides are novel inhibitors of the cell-associated spread of HCMV and facilitate the investigation of this transmission mode.


Subject(s)
Cytomegalovirus/drug effects , Peptides/chemistry , Peptides/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/chemistry , Receptor, Platelet-Derived Growth Factor alpha/pharmacology , Cytomegalovirus Infections/virology , Humans , Membrane Glycoproteins/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Virion/metabolism , Virus Internalization/drug effects
4.
Viruses ; 13(6)2021 06 08.
Article in English | MEDLINE | ID: mdl-34201364

ABSTRACT

The human cytomegalovirus (HCMV) infects fibroblasts via an interaction of its envelope glycoprotein gO with the cellular platelet-derived growth factor receptor alpha (PDGFRα), and soluble derivatives of this receptor can inhibit viral entry. We aimed to select mutants with resistance against PDGFRα-Fc and the PDGFRα-derived peptides GT40 and IK40 to gain insight into the underlying mechanisms and determine the genetic barrier to resistance. An error-prone variant of strain AD169 was propagated in the presence of inhibitors, cell cultures were monitored weekly for signs of increased viral growth, and selected viruses were tested regarding their sensitivity to the inhibitor. Resistant virus was analyzed by DNA sequencing, candidate mutations were transferred into AD169 clone pHB5 by seamless mutagenesis, and reconstituted virus was again tested for loss of sensitivity by dose-response analyses. An S48Y mutation in gO was identified that conferred a three-fold loss of sensitivity against PDGFRα-Fc, a combination of mutations in gO, gH, gB and gN reduced sensitivity to GT40 by factor 4, and no loss of sensitivity occurred with IK40. The resistance-conferring mutations support the notion that PDGFRα-Fc and GT40 perturb the interaction of gO with its receptor, but the relatively weak effect indicates a high genetic barrier to resistance.


Subject(s)
Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Drug Resistance, Viral/genetics , Mutation , Receptor, Platelet-Derived Growth Factor alpha/pharmacology , Virus Internalization/drug effects , Cell Line , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections , Epithelial Cells/drug effects , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/virology , Humans
5.
Viruses ; 13(4)2021 04 02.
Article in English | MEDLINE | ID: mdl-33918406

ABSTRACT

The role of viral envelope glycoproteins, particularly the accessory proteins of trimeric and pentameric gH/gL-complexes, in cell-associated spread of human cytomegalovirus (HCMV) is unclear. We aimed to investigate their contribution in the context of HCMV variants that grow in a strictly cell-associated manner. In the genome of Merlin pAL1502, the glycoproteins gB, gH, gL, gM, and gN were deleted by introducing stop codons, and the mutants were analyzed for viral growth. Merlin and recent HCMV isolates were compared by quantitative immunoblotting for expression of accessory proteins of the trimeric and pentameric gH/gL-complexes, gO and pUL128. Isolates were treated with siRNAs against gO and pUL128 and analyzed regarding focal growth and release of infectious virus. All five tested glycoproteins were essential for growth of Merlin pAL1502. Compared with this model virus, higher gO levels were measured in recent isolates of HCMV, and its knockdown decreased viral growth. Knockdown of pUL128 abrogated the strict cell-association and led to release of infectivity, which allowed cell-free transfer to epithelial cells where the virus grew again strictly cell-associated. We conclude that both trimer and pentamer contribute to cell-associated spread of recent clinical HCMV isolates and downregulation of pentamer can release infectious virus into the supernatant.


Subject(s)
Cytomegalovirus/growth & development , Cytomegalovirus/genetics , Epithelial Cells/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Cytomegalovirus/chemistry , Cytomegalovirus Infections/virology , Humans , Membrane Glycoproteins/genetics , Mutation , RNA, Small Interfering , Virus Internalization
6.
PLoS Pathog ; 17(3): e1009471, 2021 03.
Article in English | MEDLINE | ID: mdl-33780515

ABSTRACT

Platelet-derived growth factor receptor alpha (PDGFRα) serves as an entry receptor for the human cytomegalovirus (HCMV), and soluble PDGFRα-Fc can neutralize HCMV at a half-maximal effective concentration (EC50) of about 10 ng/ml. While this indicates a potential for usage as an HCMV entry inhibitor PDGFRα-Fc can also bind the physiological ligands of PDGFRα (PDGFs), which likely interferes with the respective signaling pathways and represents a potential source of side effects. Therefore, we tested the hypothesis that interference with PDGF signaling can be prevented by mutations in PDGFRα-Fc or combinations thereof, without losing the inhibitory potential for HCMV. To this aim, a targeted mutagenesis approach was chosen. The mutations were quantitatively tested in biological assays for interference with PDGF-dependent signaling as well as inhibition of HCMV infection and biochemically for reduced affinity to PDGF-BB, facilitating quantification of PDGFRα-Fc selectivity for HCMV inhibition. Mutation of Ile 139 to Glu and Tyr 206 to Ser strongly reduced the affinity for PDGF-BB and hence interference with PDGF-dependent signaling. Inhibition of HCMV infection was less affected, thus increasing the selectivity by factor 4 and 8, respectively. Surprisingly, the combination of these mutations had an additive effect on binding of PDGF-BB but not on inhibition of HCMV, resulting in a synergistic 260fold increase of selectivity. In addition, a recently reported mutation, Val 242 to Lys, was included in the analysis. PDGFRα-Fc with this mutation was fully effective at blocking HCMV entry and had a drastically reduced affinity for PDGF-BB. Combining Val 242 to Lys with Ile 139 to Glu and/or Tyr 206 to Ser further reduced PDGF ligand binding beyond detection. In conclusion, this targeted mutagenesis approach identified combinations of mutations in PDGFRα-Fc that prevent interference with PDGF-BB but maintain inhibition of HCMV, which qualifies such mutants as candidates for the development of HCMV entry inhibitors.


Subject(s)
Cytomegalovirus Infections , Immunoglobulin Fc Fragments , Receptor, Platelet-Derived Growth Factor alpha , Becaplermin/drug effects , Becaplermin/metabolism , Cytomegalovirus , Fibroblasts , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Mutagenesis, Site-Directed , Receptor, Platelet-Derived Growth Factor alpha/chemistry , Receptor, Platelet-Derived Growth Factor alpha/pharmacology
7.
J Infect Dis ; 218(6): 876-885, 2018 08 14.
Article in English | MEDLINE | ID: mdl-29684155

ABSTRACT

To improve the potency of anti-human cytomegalovirus (HCMV) immunoglobulin preparations, we intended to find elite neutralizers among 9000 HCMV-seropositive blood donors. We identified the top 2.6% neutralizers by use of high-throughput screening and further analyzed the 80 neutralizers with the most effective plasma for strain-independent activity. Of those, 58 had broad neutralizing activity against various HCMV strains and hence were regarded as elite neutralizers. All elite neutralizers were then analyzed to determine their effect on individual virus particles during entry. Most had plasma specimens that preferentially inhibited viral penetration, whereas 2 had exceptional plasma specimens that prevented adsorption of virus to cells. Furthermore, the neutralizing capacity of plasma samples from 3 randomly chosen elite neutralizers was up to 10-fold higher than that for commercial immunoglobulins. In a retrospective analysis of 6 selected donors, anti-HCMV neutralization titers in repeated donations were constantly high over 5 years. In conclusion, plasma samples from elite-neutralizing donors can be considered to improve antibody-based treatment of HCMV infections.


Subject(s)
Antibodies, Neutralizing/blood , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Adsorption , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/blood , Antibodies, Viral/pharmacology , Cells, Cultured , Cytomegalovirus/classification , Cytomegalovirus/physiology , Cytomegalovirus Infections/blood , High-Throughput Screening Assays , Humans , Retrospective Studies , Virus Internalization/drug effects
8.
PLoS Pathog ; 13(4): e1006273, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28403220

ABSTRACT

Human cytomegalovirus (HCMV) is a widely distributed herpesvirus that causes significant morbidity in immunocompromised hosts. Inhibitors of viral DNA replication are available, but adverse effects limit their use. Alternative antiviral strategies may include inhibition of entry. We show that soluble derivatives of the platelet-derived growth factor receptor alpha (PDGFR-alpha), a putative receptor of HCMV, can inhibit HCMV infection of various cell types. A PDGFR-alpha-Fc fusion protein binds to and neutralizes cell-free virus particles at an EC50 of 10-30 ng/ml. Treatment of particles reduced both attachment to and fusion with cells. In line with the latter, PDGFR-alpha-Fc was also effective when applied postattachment. A peptide scan of the extracellular domain of PDGFR-alpha identified a 40mer peptide that inhibits infection at an EC50 of 1-2 nmol/ml. Both, peptide and fusion protein, were effective against various HCMV strains and are hence promising candidates for the development of novel anti-HCMV therapies.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/therapy , Cytomegalovirus/drug effects , Peptides/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Virus Internalization/drug effects , Antiviral Agents/isolation & purification , Cell Line , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Endothelial Cells/virology , Fibroblasts/virology , Humans , Peptides/isolation & purification , Receptor, Platelet-Derived Growth Factor alpha/genetics , Recombinant Fusion Proteins , Virion
9.
Transfusion ; 57(2): 412-422, 2017 02.
Article in English | MEDLINE | ID: mdl-27861998

ABSTRACT

BACKGROUND: Hyperimmunoglobulins are frequently applied for prophylaxis and treatment of human cytomegalovirus (HCMV) infections but were only marginally effective in meta-analyses of clinical studies. This might be partially due to selection of donors rather for total anti-HCMV titers than for neutralizing capacities. To improve efficacy against HCMV infection, we aimed at developing a high-throughput screening method for identification of blood donors with highly and broadly neutralizing capacities. STUDY DESIGN AND METHODS: Using a Gaussia luciferase-expressing reporter virus, 1000 HCMV immunoglobulin (Ig)G-positive plasma samples with known anti-HCMV immunoglobulin titers were analyzed regarding their neutralization titers against fibroblast and endothelial cell infection. Based on these results, a high-throughput screening was designed. Highly neutralizing plasma samples were further tested 1) by an enzyme-linked immunosorbent assay-based neutralization assay regarding efficiency against different HCMV strains and 2) for their efficiency compared to commercially available hyperimmunoglobulins. RESULTS: Total anti-HCMV immunoglobulin titers did not correlate with neutralization. Mean neutralization capacities were 15-fold higher in endothelial cells compared to fibroblasts. All plasma samples neutralizing fibroblast infection were at least equally effective against infection of endothelial cells, providing the possibility to simplify our screening method by testing only fibroblasts as target cells with a plasma dilution of 1 in 400. Of the nine tested top HCMV neutralizers, four were broadly effective against different HCMV strains. All nine were significantly superior to hyperimmunoglobulins. CONCLUSION: Donors with highly and broadly neutralizing capacities can be identified by a two-step high-throughput screening approach. This may provide a basis for improved antibody-based treatment or prophylaxis of HCMV infections.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Blood Donors , Cytomegalovirus Infections , Cytomegalovirus , Donor Selection/methods , Immunoglobulin G , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Cell Line, Transformed , Cytomegalovirus/immunology , Cytomegalovirus/metabolism , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Male
10.
Leuk Lymphoma ; 45(5): 1009-15, 2004 May.
Article in English | MEDLINE | ID: mdl-15291361

ABSTRACT

Expression of the cytokine receptor CD30 is a characteristic feature of anaplastic large cell lymphoma (ALCL). Reports regarding CD30-mediated signaling in ALCL cells are highly controversial, especially with respect to the regulation of cell survival. In this study, we stimulated 6 ALCL-derived cell lines with immobilized anti-CD30 antibody. CD30-induced cell death was investigated by Western blot and FACS analysis. CD30-dependent cell proliferation and activation was analyzed by applying the trypan blue exclusion method and a luciferase-based ATP assay. The expression of cell cycle relevant proteins and the activation of mitogen-activated protein (MAP) kinases were also examined. We demonstrated that activation of CD30 did not lead to the cleavage of pro-caspase-3. FACS analysis confirmed that in all examined cells cell death was not mediated by CD30. Cell growth was strongly inhibited in 2 of the 6 cell lines and restrained cell growth was accompanied by expression of the cell cycle inhibitor p21(WAF1/CIP1). Furthermore, stimulation of CD30 led to the activation of the p38 MAP kinase but not of the extracellular signal-regulated kinase (ERK) or the jun N-terminal kinase (JNK). Interestingly, activation of CD30 induced a strong synergistic reduction of cell activity, if the p38 MAP kinase activity was blocked by SB203580. The aim of the study was to elucidate CD30-induced signaling in different ALCL-cells. Our results suggest that CD30-mediated apoptosis is not a common feature in this cell type and that p38 MAP kinase is involved in CD30-mediated singal transduction.


Subject(s)
Ki-1 Antigen/physiology , Lymphoma, Large-Cell, Anaplastic/pathology , Signal Transduction , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Gene Expression Regulation/drug effects , Humans , Ki-1 Antigen/immunology , Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Exp Hematol ; 32(4): 382-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15050749

ABSTRACT

OBJECTIVE: Expression of the cytokine receptor CD30 is a typical feature of anaplastic large cell lymphomas (ALCL). CD30-induced effects have a great impact on cell activation and viability. MATERIALS AND METHODS: Using Karpas 299 cells, we performed differential display reverse transcriptase polymerase chain reaction (DDRT-PCR) to identify novel genes involved in CD30 signaling in ALCL. Activation of CD30 was induced by treatment with immobilized anti-CD30 antibody. RNA and protein expression were confirmed in different cell lines by Northern and Western blot analysis. Fluorescence-activated cell sorting (FACS) analysis was applied to examine cell viability. Nuclear factor kappaB (NFkappaB) pathways were blocked using a specific inhibitor. RESULTS: We found strongly enhanced expression of the cellular inhibitor of apoptosis cIAP1 and cIAP2 in Karpas 299 cells stimulated with anti-CD30. Furthermore, we showed that CD30-regulated expression of cIAP1 and cIAP2 was mediated by NFkappaB. Induction of NFkappaB, cIAP1, and cIAP2 correlated with partial protection from apoptotic cell death caused by etoposide. Correspondingly, inhibition of the NFkappaB pathway not only prevented the prevalent antiapoptotic effects mediated by CD30, but even led to CD30-induced apoptosis. Finally, we found enhanced expression of cIAP1 and cIAP2 in several other ALCL cell lines and the HD-derived cell line HDLM-2 upon CD30 stimulation. CONCLUSIONS: Our results indicate that CD30-mediated protection from apoptosis is a common feature of CD30(+) cells. Therefore, CD30-induced signaling may have a significant impact on the clinical outcome of patients with ALCL.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Neoplastic/physiology , Ki-1 Antigen/physiology , Lymphoma, Large B-Cell, Diffuse/pathology , NF-kappa B/physiology , Neoplasm Proteins/physiology , Protein Biosynthesis , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Etoposide/pharmacology , Humans , Inhibitor of Apoptosis Proteins , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...