Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (38)

Search Parameters:
Keywords = HIV blocking antibodies

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 1069 KiB  
Article
Evaluation of Torquetenovirus (TTV) Particle Integrity Utilizing PMAxx™
by Giuseppe Sberna, Claudia Minosse, Cosmina Mija, Eliana Specchiarello, Pietro Giorgio Spezia, Sara Belladonna, Giulia Berno, Lavinia Fabeni, Giulia Matusali, Silvia Meschi, Daniele Focosi and Fabrizio Maggi
Int. J. Mol. Sci. 2025, 26(13), 6542; https://doi.org/10.3390/ijms26136542 - 7 Jul 2025
Viewed by 453
Abstract
Torquetenovirus (TTV) is a ubiquitous, non-pathogenic DNA virus that has been suggested as a biomarker of immune competence, with the viral load correlating with the level of immunosuppression. However, by detecting non-intact viral particles, standard PCR-based quantification may overestimate the TTV viremia. To [...] Read more.
Torquetenovirus (TTV) is a ubiquitous, non-pathogenic DNA virus that has been suggested as a biomarker of immune competence, with the viral load correlating with the level of immunosuppression. However, by detecting non-intact viral particles, standard PCR-based quantification may overestimate the TTV viremia. To improve the clinical relevance of TTV quantification, in this study, we investigated the use of PMAxx™, a virion viability dye that selectively blocks the amplification of compromised virions. Serum samples from 10 Hepatitis C Virus-positive (HCV+) individuals, 81 liver transplant recipients (LTRs), and 40 people with HIV (PWH) were treated with PMAxx™ and analyzed for TTV DNA loads by digital droplet PCR (ddPCR). Furthermore, anti-SARS-CoV-2 IgG levels and neutralizing antibody (nAbs) titers were measured post-COVID-19 vaccination. Using ddPCR, the PMAxx™ treatment significantly reduced the TTV DNA levels in all the groups (mean reduction: 0.66 Log copies/mL), indicating the abundant presence of non-intact, circulating viral genomes. However, correlations between TTV DNA and SARS-CoV-2 IgG or nAbs were weak or absent in both PMAxx™-treated and untreated samples. These findings suggest that while PMAxx™ enhanced the specificity of TTV quantification, it did not improve the predictive value of TTV viremia at assessing vaccine-induced humoral responses. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

22 pages, 3349 KiB  
Hypothesis
Does SARS-CoV-2 Possess “Allergen-Like” Epitopes?
by Alberto Rubio-Casillas, David Cowley, Vladimir N. Uversky, Elrashdy M. Redwan, Carlo Brogna and Marina Piscopo
COVID 2025, 5(4), 55; https://doi.org/10.3390/covid5040055 - 16 Apr 2025
Viewed by 1694
Abstract
An increase in immunoglobulin G4 (IgG4) levels is typically associated with immunological tolerance states and develops after prolonged exposure to antigens. Accordingly, IgG4 is considered an anti-inflammatory antibody with a limited ability to trigger efficient immune responses. Additionally, IgG4 reduces allergic reactions by [...] Read more.
An increase in immunoglobulin G4 (IgG4) levels is typically associated with immunological tolerance states and develops after prolonged exposure to antigens. Accordingly, IgG4 is considered an anti-inflammatory antibody with a limited ability to trigger efficient immune responses. Additionally, IgG4 reduces allergic reactions by blocking immunoglobulin E (IgE) activity. In the case of COVID-19, it has been reported that the repeated administration of some vaccines induces high IgG4 levels. The latest research data have revealed a surprising IgE anti-receptor binding domain response after both natural infection and several SARS-CoV-2 vaccines. The presence of IgG4 and IgE in COVID-19 disease suggests that the virus may induce an “allergic-like” response to evade immune surveillance, leading to a shift from T helper 1 (Th1) to T helper 2 (Th2) cells, which promotes tolerance to the virus and potentially contributes to chronic infection. The spike protein from vaccines could also induce such a response. Interestingly, “allergen-like” epitopes and IgE responses have been reported for other viruses, such as influenza, human immunodeficiency virus (HIV), and respiratory syncytial virus (RSV). The impact of this viral-induced tolerance will be discussed, concerning long COVID and the protective efficacy of vaccines. Full article
(This article belongs to the Section Human or Animal Coronaviruses)
Show Figures

Figure 1

18 pages, 6126 KiB  
Article
Development and Optimization of a Cost-Effective Electrochemical Immunosensor for Rapid COVID-19 Diagnosis
by Thaís Machado Lima, Daiane Martins Leal, Zirlane Coelho Ferreira, Fernando de Jesus Souza, Danilo Bretas de Oliveira, Etel Rocha-Vieira, Helen Rodrigues Martins, Arnaldo César Pereira and Lucas Franco Ferreira
Biosensors 2025, 15(2), 67; https://doi.org/10.3390/bios15020067 - 22 Jan 2025
Cited by 2 | Viewed by 1949
Abstract
The coronavirus disease (COVID-19) pandemic has created an urgent need for rapid, accurate, and cost-effective diagnostic tools. In this study, an economical electrochemical immunosensor for the rapid diagnosis of COVID-19 was developed and optimized based on charge transfer resistance (Rct) values obtained by [...] Read more.
The coronavirus disease (COVID-19) pandemic has created an urgent need for rapid, accurate, and cost-effective diagnostic tools. In this study, an economical electrochemical immunosensor for the rapid diagnosis of COVID-19 was developed and optimized based on charge transfer resistance (Rct) values obtained by electrochemical impedance spectroscopy (EIS) from the interaction between antibodies (anti-SARS-CoV-2) immobilized as a bioreceptor and the virus (SARS-CoV-2). The sensor uses modified pencil graphite electrodes (PGE) coated with poly(4-hydroxybenzoic acid), anti-SARS-CoV-2, and silver nanoparticles. The immobilization of anti-SARS-CoV-2 antibodies was optimized at a concentration of 1:250 for 30 min, followed by blocking the surface with 0.01% bovine serum albumin for 10 min. The optimal conditions for virus detection in clinical samples were a 1:10 dilution with a response time of 20 min. The immunosensor responded linearly in the range of 0.2–2.5 × 106 particles/μL. From the relationship between the obtained signal and the concentration of the analyzed sample, the limit of detection (LOD) and limit of quantification (LOQ) obtained were 1.21 × 106 and 4.04 × 106 particles/μL, respectively. The device did not cross-react with other viruses, including Influenza A and B, HIV, and Vaccinia virus. The relative standard deviation (RSD) of the six immunosensors prepared using the shared-pool sample was 3.87. Decreases of 22.3% and 12.4% were observed in the response values of the ten immunosensors stored at 25 °C and 4.0 °C, respectively. The sensor provides timely and accurate results with high sensitivity and specificity, offering a cost-effective alternative to the existing diagnostic methods. Full article
(This article belongs to the Special Issue Nanomaterial-Enhanced Biosensing for Point-of-Care Diagnostics)
Show Figures

Figure 1

23 pages, 1732 KiB  
Review
The Proviral Reservoirs of Human Immunodeficiency Virus (HIV) Infection
by Andrey I. Murzin, Kirill A. Elfimov and Natalia M. Gashnikova
Pathogens 2025, 14(1), 15; https://doi.org/10.3390/pathogens14010015 - 30 Dec 2024
Viewed by 2128
Abstract
Human Immunodeficiency Virus (HIV) proviral reservoirs are cells that harbor integrated HIV proviral DNA within their nuclear genomes. These cells form a heterogeneous group, represented by peripheral blood mononuclear cells (PBMCs), tissue-resident lymphoid and monocytic cells, and glial cells of the central nervous [...] Read more.
Human Immunodeficiency Virus (HIV) proviral reservoirs are cells that harbor integrated HIV proviral DNA within their nuclear genomes. These cells form a heterogeneous group, represented by peripheral blood mononuclear cells (PBMCs), tissue-resident lymphoid and monocytic cells, and glial cells of the central nervous system. The importance of studying the properties of proviral reservoirs is connected with the inaccessibility of integrated HIV proviral DNA for modern anti-retroviral therapies (ARTs) that block virus reproduction. If treatment is not effective enough or is interrupted, the proviral reservoir can reactivate. Early initiation of ART improves the prognosis of the course of HIV infection, which is explained by the reduction in the proviral reservoir pool observed in the early stages of the disease. Different HIV subtypes present differences in the number of latent reservoirs, as determined by structural and functional differences. Unique signatures of patients with HIV, such as elite controllers, have control over viral replication and can be said to have achieved a functional cure for HIV infection. Uncovering the causes of this phenomenon will bring humanity closer to curing HIV infection, potential approaches to which include stem cell transplantation, clustered regularly interspaced short palindromic repeats (CRISPR)/cas9, “Shock and kill”, “Block and lock”, and the application of broad-spectrum neutralizing antibodies (bNAbs). Full article
(This article belongs to the Special Issue Retroviruses: Molecular Biology, Immunology and Pathogenesis)
Show Figures

Figure 1

14 pages, 2047 KiB  
Article
Enhancement of Human Immunodeficiency Virus-Specific CD8+ T Cell Responses with TIGIT Blockade Involves Trogocytosis
by Nazanin Ghasemi, Kayla A. Holder, Danielle P. Ings and Michael D. Grant
Pathogens 2024, 13(12), 1137; https://doi.org/10.3390/pathogens13121137 - 23 Dec 2024
Viewed by 1231
Abstract
Natural killer (NK) and CD8+ T cell function is compromised in human immunodeficiency virus type 1 (HIV-1) infection by increased expression of inhibitory receptors such as TIGIT (T cell immunoreceptor with Ig and ITIM domains). Blocking inhibitory receptors or their ligands with [...] Read more.
Natural killer (NK) and CD8+ T cell function is compromised in human immunodeficiency virus type 1 (HIV-1) infection by increased expression of inhibitory receptors such as TIGIT (T cell immunoreceptor with Ig and ITIM domains). Blocking inhibitory receptors or their ligands with monoclonal antibodies (mAb) has potential to improve antiviral immunity in general and facilitate HIV eradication strategies. We assessed the impact of TIGIT engagement and blockade on cytotoxicity, degranulation, and interferon-gamma (IFN-γ) production by CD8+ T cells from persons living with HIV (PLWH). The effect of TIGIT engagement on non-specific anti-CD3-redirected cytotoxicity was assessed in redirected cytotoxicity assays, and the effect of TIGIT blockade on HIV-specific CD8+ T cell responses was assessed by flow cytometry. In 14/19 cases where peripheral blood mononuclear cells (PBMC) mediated >10% redirected cytotoxicity, TIGIT engagement reduced the level of cytotoxicity to <90% of control values. We selected PLWH with >1000 HIV Gag or Nef-specific IFN-γ spot forming cells per million PBMC to quantify the effects of TIGIT blockade on HIV-specific CD8+ T cell responses by flow cytometry. Cell surface TIGIT expression decreased on CD8+ T cells from 23/40 PLWH following TIGIT blockade and this loss was associated with increased anti-TIGIT mAb fluorescence on monocytes. In total, 6 of these 23 PLWH had enhanced HIV-specific CD8+ T cell degranulation and IFN-γ production with TIGIT blockade, compared to 0/17 with no decrease in cell surface TIGIT expression. Reduced CD8+ T cell TIGIT expression with TIGIT blockade involved trogocytosis by circulating monocytes, suggesting that an effector monocyte population and intact fragment crystallizable (Fc) functions are required for mAb-based TIGIT blockade to effectively enhance HIV-specific CD8+ T cell responses. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

16 pages, 4757 KiB  
Article
A hTfR1 Receptor-Specific VHH Antibody Neutralizes Pseudoviruses Expressing Glycoproteins from Junín and Machupo Viruses
by Qinglin Kang, Gege Li, Yan Wu, Shaoyan Wang, Zhengshan Chen, Xiaodong Zai, Xiaoyan Pan, Rong Wang, Jiansheng Lu, Peng Du, Zhixin Yang, Xiangyang Chi, Gengfu Xiao and Junjie Xu
Viruses 2024, 16(12), 1951; https://doi.org/10.3390/v16121951 - 20 Dec 2024
Viewed by 1892
Abstract
The Junín virus (JUNV) is one of the New World arenaviruses that cause severe hemorrhagic fever. Human transferrin receptor 1 (hTfR1) has been identified as the main receptor for JUNV for virus entry into host cells. To date, no treatment has been approved [...] Read more.
The Junín virus (JUNV) is one of the New World arenaviruses that cause severe hemorrhagic fever. Human transferrin receptor 1 (hTfR1) has been identified as the main receptor for JUNV for virus entry into host cells. To date, no treatment has been approved for JUNV. Herein, we investigated 12 anti-hTfR1 VHH (variable domain of the heavy chain of heavy-chain antibody) antibodies and confirmed their interaction with hTfR1. Most of them could bind to the hTfR1 apical domain, which is the glycoprotein 1 (GP1) binding domain of JUNV. Among them, 18N18 exhibited neutralizing activity against both the human immunodeficiency virus (HIV)-vectored lentiviral Junín pseudoviruses and the recombinant vesicular stomatitis virus (VSV)-vectored Junín pseudoviruses. We also verified that 18N18 blocked the interaction between hTfR1 and JUNV GP1. In addition, 18N18 could neutralize another New World arenavirus, the Machupo virus. Using AlphaFold 3-based simulation of 18N18–hTfR1 docking, we determined that 18N18’s binding epitope was located at the JUNV GP1 binding epitope. 18N18 represents a candidate for JUNV treatment and provides a potential approach that could be applied to New World arenaviruses. Full article
(This article belongs to the Special Issue B Cell-Mediated Immunity to Viruses)
Show Figures

Figure 1

26 pages, 2734 KiB  
Review
Interventions during Early Infection: Opening a Window for an HIV Cure?
by Christopher R. Hiner, April L. Mueller, Hang Su and Harris Goldstein
Viruses 2024, 16(10), 1588; https://doi.org/10.3390/v16101588 - 9 Oct 2024
Cited by 3 | Viewed by 3252
Abstract
Although combination antiretroviral therapy (ART) has been a landmark achievement for the treatment of human immunodeficiency virus (HIV), an HIV cure has remained elusive. Elimination of latent HIV reservoirs that persist throughout HIV infection is the most challenging barrier to an HIV cure. [...] Read more.
Although combination antiretroviral therapy (ART) has been a landmark achievement for the treatment of human immunodeficiency virus (HIV), an HIV cure has remained elusive. Elimination of latent HIV reservoirs that persist throughout HIV infection is the most challenging barrier to an HIV cure. The progressive HIV infection is marked by the increasing size and diversity of latent HIV reservoirs until an effective immune response is mobilized, which can control but not eliminate HIV infection. The stalemate between HIV replication and the immune response is manifested by the establishment of a viral set point. ART initiation during the early stage limits HIV reservoir development, preserves immune function, improves the quality of life, and may lead to ART-free viral remission in a few people living with HIV (PLWH). However, for the overwhelming majority of PLWH, early ART initiation alone does not cure HIV, and lifelong ART is needed to sustain viral suppression. A critical area of research is focused on determining whether HIV could be functionally cured if additional treatments are provided alongside early ART. Several HIV interventions including Block and Lock, Shock and Kill, broadly neutralizing antibody (bNAb) therapy, adoptive CD8+ T cell therapy, and gene therapy have demonstrated delayed viral rebound and/or viral remission in animal models and/or some PLWH. Whether or not their application during early infection can improve the success of HIV remission is less studied. Herein, we review the current state of clinical and investigative HIV interventions and discuss their potential to improve the likelihood of post-treatment remission if initiated during early infection. Full article
(This article belongs to the Special Issue Acute HIV Infections)
Show Figures

Graphical abstract

18 pages, 6477 KiB  
Article
A New Chimeric Antibody against the HIV-1 Fusion Inhibitory Peptide MT-C34 with a High Affinity and Fc-Mediated Cellular Cytotoxicity
by Svetlana V. Kalinichenko, Lama Ramadan, Natalia A. Kruglova, Konstantin I. Balagurov, Marina I. Lukashina, Dmitriy V. Mazurov and Mikhail V. Shepelev
Biology 2024, 13(9), 675; https://doi.org/10.3390/biology13090675 - 29 Aug 2024
Cited by 2 | Viewed by 1679
Abstract
Peptides from heptad repeat (HR1 and HR2) regions of gp41 are effective inhibitors of HIV-1 entry that block the fusion of viral and cellular membranes, but the generation of antibodies highly specific for these peptides is challenging. We have previously described a mouse [...] Read more.
Peptides from heptad repeat (HR1 and HR2) regions of gp41 are effective inhibitors of HIV-1 entry that block the fusion of viral and cellular membranes, but the generation of antibodies highly specific for these peptides is challenging. We have previously described a mouse hybridoma that recognizes MT-C34-related peptides derived from HR2. It was used for the selection of HIV-1-resistant CD4 lymphocytes engineered to express the MT-C34 peptide via a CRISPR/Cas9-mediated knock-in into the CXCR4 locus. In this study, we cloned variable domains of this antibody and generated a recombinant chimeric antibody (chAb) by combining it with the constant regions of the humanized antibody Trastuzumab. The new chAb displayed a high specificity and two-fold higher level of affinity than the parental mouse monoclonal antibody. In addition, chAb mediated up to 27–43% of the antibody-dependent cellular cytotoxicity towards cells expressing MT-C34 on their surface. The anti-MT-C34 chAb can be easily generated using plasmids available for the research community and can serve as a valuable tool for the detection, purification, and even subsequent elimination of HIV-1-resistant CD4 cells or CAR cells engineered to fight HIV-1 infection. Full article
(This article belongs to the Special Issue B and T Cells in HIV and Other Viral Infections)
Show Figures

Graphical abstract

23 pages, 1442 KiB  
Review
Role of HIV-1 Tat Protein Interactions with Host Receptors in HIV Infection and Pathogenesis
by Aurelio Cafaro, Ivan Schietroma, Leonardo Sernicola, Roberto Belli, Massimo Campagna, Flavia Mancini, Stefania Farcomeni, Maria Rosaria Pavone-Cossut, Alessandra Borsetti, Paolo Monini and Barbara Ensoli
Int. J. Mol. Sci. 2024, 25(3), 1704; https://doi.org/10.3390/ijms25031704 - 30 Jan 2024
Cited by 14 | Viewed by 5675
Abstract
Each time the virus starts a new round of expression/replication, even under effective antiretroviral therapy (ART), the transactivator of viral transcription Tat is one of the first HIV-1 protein to be produced, as it is strictly required for HIV replication and spreading. At [...] Read more.
Each time the virus starts a new round of expression/replication, even under effective antiretroviral therapy (ART), the transactivator of viral transcription Tat is one of the first HIV-1 protein to be produced, as it is strictly required for HIV replication and spreading. At this stage, most of the Tat protein exits infected cells, accumulates in the extracellular matrix and exerts profound effects on both the virus and neighbor cells, mostly of the innate and adaptive immune systems. Through these effects, extracellular Tat contributes to the acquisition of infection, spreading and progression to AIDS in untreated patients, or to non-AIDS co-morbidities in ART-treated individuals, who experience inflammation and immune activation despite virus suppression. Here, we review the role of extracellular Tat in both the virus life cycle and on cells of the innate and adaptive immune system, and we provide epidemiological and experimental evidence of the importance of targeting Tat to block residual HIV expression and replication. Finally, we briefly review vaccine studies showing that a therapeutic Tat vaccine intensifies ART, while its inclusion in a preventative vaccine may blunt escape from neutralizing antibodies and block early events in HIV acquisition. Full article
Show Figures

Figure 1

11 pages, 1512 KiB  
Article
A dePEGylated Lipopeptide-Based Pan-Coronavirus Fusion Inhibitor Exhibits Potent and Broad-Spectrum Anti-HIV-1 Activity without Eliciting Anti-PEG Antibodies
by Ling Xu, Chao Wang, Wei Xu, Lixiao Xing, Jie Zhou, Jing Pu, Mingming Fu, Lu Lu, Shibo Jiang and Qian Wang
Int. J. Mol. Sci. 2023, 24(11), 9779; https://doi.org/10.3390/ijms24119779 - 5 Jun 2023
Cited by 2 | Viewed by 2174
Abstract
We previously identified a lipopeptide, EK1C4, by linking cholesterol to EK1, a pan-CoV fusion inhibitory peptide via a polyethylene glycol (PEG) linker, which showed potent pan-CoV fusion inhibitory activity. However, PEG can elicit antibodies to PEG in vivo, which will attenuate its antiviral [...] Read more.
We previously identified a lipopeptide, EK1C4, by linking cholesterol to EK1, a pan-CoV fusion inhibitory peptide via a polyethylene glycol (PEG) linker, which showed potent pan-CoV fusion inhibitory activity. However, PEG can elicit antibodies to PEG in vivo, which will attenuate its antiviral activity. Therefore, we designed and synthesized a dePEGylated lipopeptide, EKL1C, by replacing the PEG linker in EK1C4 with a short peptide. Similar to EK1C4, EKL1C displayed potent inhibitory activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other coronaviruses. In this study, we found that EKL1C also exhibited broad-spectrum fusion inhibitory activity against human immunodeficiency virus type 1 (HIV-1) infection by interacting with the N-terminal heptad repeat 1 (HR1) of viral gp41 to block six-helix bundle (6-HB) formation. These results suggest that HR1 is a common target for the development of broad-spectrum viral fusion inhibitors and EKL1C has potential clinical application as a candidate therapeutic or preventive agent against infection by coronavirus, HIV-1, and possibly other class I enveloped viruses. Full article
(This article belongs to the Collection Feature Papers in Molecular Immunology)
Show Figures

Figure 1

14 pages, 2587 KiB  
Article
Unveiling CD59-Antibody Interactions to Design Paratope-Mimicking Peptides for Complement Modulation
by Annamaria Sandomenico, Alessia Ruggiero, Emanuela Iaccarino, Angela Oliver, Flavia Squeglia, Miguel Moreira, Luciana Esposito, Menotti Ruvo and Rita Berisio
Int. J. Mol. Sci. 2023, 24(10), 8561; https://doi.org/10.3390/ijms24108561 - 10 May 2023
Cited by 1 | Viewed by 2450
Abstract
CD59 is an abundant immuno-regulatory human protein that protects cells from damage by inhibiting the complement system. CD59 inhibits the assembly of the Membrane Attack Complex (MAC), the bactericidal pore-forming toxin of the innate immune system. In addition, several pathogenic viruses, including HIV-1, [...] Read more.
CD59 is an abundant immuno-regulatory human protein that protects cells from damage by inhibiting the complement system. CD59 inhibits the assembly of the Membrane Attack Complex (MAC), the bactericidal pore-forming toxin of the innate immune system. In addition, several pathogenic viruses, including HIV-1, escape complement-mediated virolysis by incorporating this complement inhibitor in their own viral envelope. This makes human pathogenic viruses, such as HIV-1, not neutralised by the complement in human fluids. CD59 is also overexpressed in several cancer cells to resist the complement attack. Consistent with its importance as a therapeutical target, CD59-targeting antibodies have been proven to be successful in hindering HIV-1 growth and counteracting the effect of complement inhibition by specific cancer cells. In this work, we make use of bioinformatics and computational tools to identify CD59 interactions with blocking antibodies and to describe molecular details of the paratope–epitope interface. Based on this information, we design and produce paratope-mimicking bicyclic peptides able to target CD59. Our results set the basis for the development of antibody-mimicking small molecules targeting CD59 with potential therapeutic interest as complement activators. Full article
(This article belongs to the Collection Immunopathology and Immunosenescence)
Show Figures

Figure 1

10 pages, 1504 KiB  
Article
Infection of the Ex Vivo Tonsil Model by HTLV-1 Envelope-Pseudotyped Viruses
by Mélanie Langlois, Salim Bounou, Michel J. Tremblay and Benoit Barbeau
Pathogens 2023, 12(2), 182; https://doi.org/10.3390/pathogens12020182 - 24 Jan 2023
Cited by 3 | Viewed by 2280
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the causal agent of adult T-cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. Its tropism is known to be broad in cultured cell lines, while in vivo data support a more selective transmission toward CD4+ T [...] Read more.
Human T-cell leukemia virus type 1 (HTLV-1) is the causal agent of adult T-cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. Its tropism is known to be broad in cultured cell lines, while in vivo data support a more selective transmission toward CD4+ T cells and the limited targeting of other hematopoietic cell types. An essential condition for HTLV-1 infection is cell-to-cell contact, to which both virological synapse and viral biofilm have been suggested to strongly contribute. As cell lines and animal models each present their own limitations in studying HTLV-1 replication, we have explored the use of an ex vivo model based on the secondary lymphoid tonsillar tissue. HIV-1 luciferase-expressing pseudotyped viruses bearing the HTLV-1 envelope protein at their surface were first shown to recapitulate the wide spectrum of infectivity of HTLV-1 toward various cell lines. Tonsil fragments were next exposed to pseudotyped viruses and shown to be reproducibly infected. Infection by HTLV-1 Env-pseudotyped viruses was blocked by different anti-gp46 antibodies, unlike infection by HIV-1 virions. The dose-dependent infection revealed a gradual increase in luciferase activity, which was again sensitive to anti-gp46 antibodies. Overall, these results suggest that the ex vivo tonsil model represents a reliable alternative for studying HTLV-1 replication and potentially viral latency, as well as early clonal formation. Full article
(This article belongs to the Special Issue Emerging Infectious Diseases)
Show Figures

Figure 1

11 pages, 1318 KiB  
Article
Ex Vivo Blockade of the PD-1 Pathway Improves Recall IFNγ Responses of HIV-Infected Persons on Antiretroviral Therapy
by Natalie Fischhaber, Moritz Schmiedeberg, Sabrina Kübel, Ellen G. Harrer, Thomas Harrer and Krystelle Nganou-Makamdop
Vaccines 2023, 11(2), 211; https://doi.org/10.3390/vaccines11020211 - 18 Jan 2023
Cited by 1 | Viewed by 2383
Abstract
Despite antiretroviral therapy (ART), immune exhaustion persists in HIV infection and limits T cell responses to HIV or other pathogens. Moreover, HIV infection results in the loss of pre-existing immunity. Here, we investigated the effect of blocking the PD-1 pathway on recall IFNγ [...] Read more.
Despite antiretroviral therapy (ART), immune exhaustion persists in HIV infection and limits T cell responses to HIV or other pathogens. Moreover, HIV infection results in the loss of pre-existing immunity. Here, we investigated the effect of blocking the PD-1 pathway on recall IFNγ responses to tetanus toxoid (TT) and measles virus (MV) antigens in HIV-infected persons on ART with prior TT and MV immunity. The ex vivo treatment of lymphocytes with anti-PD-1 and anti-PD-L1 antibodies significantly increased TT- and MV-specific IFNγ responses. The responses to TT and MV antigens alone or in combination with antibodies blocking the PD-1 pathway positively correlated with CD4 T cell levels. Furthermore, T cell PD-1 expression levels inversely correlated with recall IFNγ responses in combination with antibodies blocking the PD-1 pathway but not with IFNγ responses to antigens only. Our study suggested that targeting the PD-1 pathway may boost vaccine-induced pre-existing immunity in HIV-infected persons on ART depending on the degree of immune exhaustion. Full article
(This article belongs to the Section Vaccines, Clinical Advancement, and Associated Immunology)
Show Figures

Figure 1

14 pages, 1782 KiB  
Article
Fusion Proteins CLD and CLDmut Demonstrate Potent and Broad Neutralizing Activity against HIV-1
by Ming Fu, Yingying Xiao, Tao Du, Huimin Hu, Fengfeng Ni, Kai Hu and Qinxue Hu
Viruses 2022, 14(7), 1365; https://doi.org/10.3390/v14071365 - 23 Jun 2022
Cited by 2 | Viewed by 2495
Abstract
HIV-1 envelope glycoprotein (Env) interacts with cellular receptors and mediates virus entry into target cells. Blocking Env-receptor interactions represents an effective interventional strategy for developing HIV-1 entry inhibitors. We previously designed a panel of CD4-linker-DC-SIGN (CLD) constructs by fusing the extracellular CD4 and [...] Read more.
HIV-1 envelope glycoprotein (Env) interacts with cellular receptors and mediates virus entry into target cells. Blocking Env-receptor interactions represents an effective interventional strategy for developing HIV-1 entry inhibitors. We previously designed a panel of CD4-linker-DC-SIGN (CLD) constructs by fusing the extracellular CD4 and DC-SIGN domains with various linkers. Such CLDs produced by the prokaryotic system efficiently inhibited HIV-1 infection and dissemination in vitro and ex vivo. In this study, following the construction and identification of the most promising candidate with a linker of 8 Gly4Ser repeats (named CLD), we further designed an improved form (named CLDmut) by back mutating Cys to Ser at amino acid 60 of CD4. Both CLD and CLDmut were produced in mammalian (293F) cells for better protein translation and modification. The anti-HIV-1 activity of CLD and CLDmut was assessed against the infection of a range of HIV-1 isolates, including transmitted and founder (T/F) viruses. While both CLD and CLDmut efficiently neutralized the tested HIV-1 isolates, CLDmut demonstrated much higher neutralizing activity than CLD, with an IC50 up to one log lower. The neutralizing activity of CLDmut was close to or more potent than those of the highly effective HIV-1 broadly neutralizing antibodies (bNAbs) reported to date. Findings in this study indicate that mammalian cell-expressed CLDmut may have the potential to be used as prophylaxis or/and therapeutics against HIV-1 infection. Full article
(This article belongs to the Special Issue Viral Entry Inhibitors 2022)
Show Figures

Figure 1

23 pages, 2360 KiB  
Article
Targeted Immuno-Antiretroviral to Promote Dual Protection against HIV: A Proof-of-Concept Study
by Subhra Mandal, Shawnalyn W. Sunagawa, Pavan Kumar Prathipati, Michael Belshan, Annemarie Shibata and Christopher J. Destache
Nanomaterials 2022, 12(11), 1942; https://doi.org/10.3390/nano12111942 - 6 Jun 2022
Cited by 2 | Viewed by 2912
Abstract
The C–C motif chemokine receptor-5 (CCR5) expression on the T-cell surface is the prime barrier to HIV/AIDS eradication, as it promotes both active human immunodeficiency virus (HIV)-infection and latency; however, antiretrovirals (ARVs) suppress plasma viral loads to non-detectable levels. Keeping this in mind, [...] Read more.
The C–C motif chemokine receptor-5 (CCR5) expression on the T-cell surface is the prime barrier to HIV/AIDS eradication, as it promotes both active human immunodeficiency virus (HIV)-infection and latency; however, antiretrovirals (ARVs) suppress plasma viral loads to non-detectable levels. Keeping this in mind, we strategically designed a targeted ARVs-loaded nanoformulation that targets CCR5 expressing T-cells (e.g., CD4+ cells). Conceptually, CCR5-blocking and targeted ARV delivery would be a dual protection strategy to prevent HIV infection. For targeting CCR5+ T-cells, the nanoformulation was surface conjugated with anti-CCR5 monoclonal antibodies (CCR5 mAb) and loaded with dolutegravir+tenofovir alafenamide (D+T) ARVs to block HIV replication. The result demonstrated that the targeted-ARV nanoparticle’s multimeric CCR5 binding property improved its antigen-binding affinity, prolonged receptor binding, and ARV intracellular retention. Further, nanoformulation demonstrated high binding affinity to CCR5 expressing CD4+ cells, monocytes, and other CCR5+ T-cells. Finally, the short-term pre-exposure prophylaxis study demonstrated that prolonged CCR5 blockage and ARV presence further induced a “protective immune phenotype” with a boosted T-helper (Th), temporary memory (TM), and effector (E) sub-population. The proof-of-concept study that the targeted-ARV nanoformulation dual-action mechanism could provide a multifactorial solution toward achieving HIV “functional cure.” Full article
Show Figures

Graphical abstract

Back to TopTop