Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(27): e2122050119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35763571

ABSTRACT

AIDS-defining cancers declined after combined antiretroviral therapy (cART) introduction, but lymphomas are still elevated in HIV type 1 (HIV-1)-infected patients. In particular, non-Hodgkin's lymphomas (NHLs) represent the majority of all AIDS-defining cancers and are the most frequent cause of death in these patients. We have recently demonstrated that amino acid (aa) insertions at the HIV-1 matrix protein p17 COOH-terminal region cause protein destabilization, leading to conformational changes. Misfolded p17 variants (vp17s) strongly impact clonogenic B cell growth properties that may contribute to B cell lymphomagenesis as suggested by the significantly higher frequency of detection of vp17s with COOH-terminal aa insertions in plasma of HIV-1-infected patients with NHL. Here, we expand our previous observations by assessing the prevalence of vp17s in large retrospective cohorts of patients with and without lymphoma. We confirm the significantly higher prevalence of vp17s in lymphoma patients than in HIV-1-infected individuals without lymphoma. Analysis of 3,990 sequences deposited between 1985 and 2017 allowed us to highlight a worldwide increasing prevalence of HIV-1 mutants expressing vp17s over time. Since genomic surveillance uncovered a cluster of HIV-1 expressing a B cell clonogenic vp17 dated from 2011 to 2019, we conclude that aa insertions can be fixed in HIV-1 and that mutant viruses displaying B cell clonogenic vp17s are actively spreading.


Subject(s)
B-Lymphocytes , HIV Antigens , HIV-1 , Lymphoma, AIDS-Related , gag Gene Products, Human Immunodeficiency Virus , B-Lymphocytes/virology , Genetic Variation , HIV Antigens/genetics , HIV-1/genetics , HIV-1/isolation & purification , Humans , Lymphoma, AIDS-Related/epidemiology , Lymphoma, AIDS-Related/virology , Prevalence , Retrospective Studies , gag Gene Products, Human Immunodeficiency Virus/genetics
2.
J Gen Virol ; 105(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38687324

ABSTRACT

HIV-1 matrix protein p17 variants (vp17s), characterized by amino acid insertions at the COOH-terminal region of the viral protein, have been recently identified and studied for their biological activity. Different from their wild-type counterpart (refp17), vp17s display a potent B cell growth and clonogenic activity. Recent data have highlighted the higher prevalence of vp17s in people living with HIV-1 (PLWH) with lymphoma compared with those without lymphoma, suggesting that vp17s may play a key role in lymphomagenesis. Molecular mechanisms involved in vp17 development are still unknown. Here we assessed the efficiency of HIV-1 Reverse Transcriptase (RT) in processing this genomic region and highlighted the existence of hot spots of mutation in Gag, at the end of the matrix protein and close to the matrix-capsid junction. This is possibly due to the presence of inverted repeats and palindromic sequences together with a high content of Adenine in the 322-342 nucleotide portion, which constrain HIV-1 RT to pause on the template. To define the recombinogenic properties of hot spots of mutation in the matrix gene, we developed plasmid vectors expressing Gag and a minimally modified Gag variant, and measured homologous recombination following cell co-nucleofection by next-generation sequencing. Data obtained allowed us to show that a wide range of recombination events occur in concomitance with the identified hot spots of mutation and that imperfect events may account for vp17s generation.


Subject(s)
HIV Antigens , HIV Reverse Transcriptase , HIV-1 , gag Gene Products, Human Immunodeficiency Virus , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , Humans , HIV-1/genetics , HIV Antigens/genetics , HIV Antigens/metabolism , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Mutation , HIV Infections/virology , HIV Infections/genetics , Cell Line
3.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33372148

ABSTRACT

The HIV-1 matrix protein p17 (p17) is a pleiotropic molecule impacting on different cell types. Its interaction with many cellular proteins underlines the importance of the viral protein as a major determinant of human specific adaptation. We previously showed the proangiogenic capability of p17. Here, by integrating functional analysis and receptor binding, we identify a functional epitope that displays molecular mimicry with human erythropoietin (EPO) and promotes angiogenesis through common beta chain receptor (ßCR) activation. The functional EPO-like epitope was found to be present in the matrix protein of HIV-1 ancestors SIV originated in chimpanzees (SIVcpz) and gorillas (SIVgor) but not in that of HIV-2 and its ancestor SIVsmm from sooty mangabeys. According to biological data, evolution of the EPO-like epitope showed a clear differentiation between HIV-1/SIVcpz-gor and HIV-2/SIVsmm branches, thus highlighting this epitope on p17 as a divergent signature discriminating HIV-1 and HIV-2 ancestors. P17 is known to enhance HIV-1 replication. Similarly to other ßCR ligands, p17 is capable of attracting and activating HIV-1 target cells and promoting a proinflammatory microenvironment. Thus, it is tempting to speculate that acquisition of an epitope on the matrix proteins of HIV-1 ancestors capable of triggering ßCR may have represented a critical step to enhance viral aggressiveness and early human-to-human SIVcpz/gor dissemination. The hypothesis that the p17/ßCR interaction and ßCR abnormal stimulation may also play a role in sustaining chronic activation and inflammation, thus marking the difference between HIV-1 and HIV-2 in term of pathogenicity, needs further investigation.


Subject(s)
Erythropoietin/genetics , HIV Antigens/metabolism , HIV-1/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Epitopes/immunology , Erythropoietin/metabolism , Evolution, Molecular , HIV Antigens/genetics , HIV Seropositivity , HIV-1/genetics , HIV-2 , Humans , Molecular Mimicry , Simian Immunodeficiency Virus , gag Gene Products, Human Immunodeficiency Virus/genetics
4.
New Microbiol ; 47(1): 80-87, 2024 May.
Article in English | MEDLINE | ID: mdl-38700887

ABSTRACT

The COVID-19 pandemic forced the adoption of non-pharmaceutical interventions (NPIs) which influenced the circulation of other respiratory pathogens, such as Influenza virus (FLU), Parainfluenza virus (PIV), Respiratory Syncytial virus (RSV), Rhinovirus (RV), Enterovirus (EV), Adenovirus (AdV), Human Metapneumovirus (hMPV), and Human Coronavirus (CoV). The aim of the current study was to investigate how, with the end of the pandemic, the withdrawal of the NPIs impacted on the circulation and distribution of common respiratory viruses. The analyzed samples were collected from June 2021 to March 2023 (post-pandemic period) and compared to ones from the pandemic period. Nucleic acid detection of all respiratory viruses was performed by multiplex real time Polymerase Chain Reaction (PCR) and sequencing was conducted by Next Generation Sequencing (NGS) technique. Our analysis shows that the NPIs adopted against SARS-CoV-2 were also effective in controlling the spread of other respiratory viruses. Moreover, we documented how RV/EVs were the most commonly identified species, with the more abundant strains represented by Coxsackievirus (CV)-A/B and RV-A/C. RV/EVs were also detected in some co-infection cases; in particular, the majority of co-infections concerned CV-B/RV-A, CV-B/ECHO. Given the pandemic potential of respiratory viruses, accurate molecular screening is essential for a proper surveillance and prevention strategy.


Subject(s)
COVID-19 , Respiratory Tract Infections , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/virology , Italy/epidemiology , SARS-CoV-2/genetics , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Pandemics , Viruses/genetics , Viruses/isolation & purification , Viruses/classification , Adult , Male , Child
5.
J Virol ; 96(1): e0120021, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34668776

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1)-associated neurocognitive disorder (HAND) remains an important neurological manifestation in HIV-1-infected (HIV+) patients. Furthermore, detection of the HIV-1 matrix protein p17 (p17) in the central nervous system (CNS) and its ability to form toxic assemblies in the brain have been recently confirmed. Here, we show for the first time, using both an in vitro blood-brain barrier (BBB) model and in vivo biodistribution studies in healthy mice, that p17 can cross the BBB. There is rapid brain uptake with 0.35% ± 0.19% of injected activity per gram of tissue (IA/g) 2 min after administration, followed by brain accumulation with 0.28% ± 0.09% IA/g after 1 h. The interaction of p17 with chemokine receptor 2 (CXCR2) at the surface of brain endothelial cells triggers transcytosis. The present study supports the hypothesis of a direct role of free p17 in neuronal dysfunction in HAND by demonstrating its intrinsic ability to reach the CNS. IMPORTANCE The percentage of patients affected by HIV-1-associated neurocognitive disorder (HAND) ranges from 30% to 50% of HIV-infected (HIV+) patients. The mechanisms leading to HAND development need to be elucidated, but the roles of secreted viral proteins, chemokines, and proinflammatory molecules appear to be clear. In particular, the blood-brain barrier (BBB) represents a route for entry into the central nervous system (CNS) and thus plays an important role in HAND. Several findings suggest a key role for the HIV-1 matrix protein p17 (p17) as a microenvironmental factor capable of inducing neurocognitive disorders. Here, we show the ability of the p17 to cross the BBB and to reach the CNS, thus playing a crucial role in neuronal dysfunction in HAND.


Subject(s)
Blood-Brain Barrier/metabolism , HIV Antigens/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , gag Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Autophagy , Cell Line , Cells, Cultured , Disease Susceptibility , Endosomes/metabolism , Endothelial Cells/metabolism , Endothelial Cells/virology , Humans , Mice , Protein Binding , Protein Transport , Receptors, Interleukin-8B/metabolism
6.
J Med Virol ; 95(6): e28848, 2023 06.
Article in English | MEDLINE | ID: mdl-37294038

ABSTRACT

During COVID-19 pandemic, consensus genomic sequences were used for rapidly monitor the spread of the virus worldwide. However, less attention was paid to intrahost genetic diversity. In fact, in the infected host, SARS-CoV-2 consists in an ensemble of replicating and closely related viral variants so-called quasispecies. Here we show that intrahost single nucleotide variants (iSNVs) represent a target for contact tracing analysis. Our data indicate that in the acute phase of infection, in highly likely transmission links, the number of viral particles transmitted from one host to another (bottleneck size) is large enough to propagate iSNVs among individuals. Furthermore, we demonstrate that, during SARS-CoV-2 outbreaks when the consensus sequences are identical, it is possible to reconstruct the transmission chains by genomic investigations of iSNVs. Specifically, we found that it is possible to identify transmission chains by limiting the analysis of iSNVs to only three well-conserved genes, namely nsp2, ORF3, and ORF7.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Quasispecies , Pandemics , Genome, Viral
7.
J Med Virol ; 95(8): e29012, 2023 08.
Article in English | MEDLINE | ID: mdl-37548148

ABSTRACT

This comprehensive review focuses on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its impact as the cause of the COVID-19 pandemic. Its objective is to provide a cohesive overview of the epidemic history and evolutionary aspects of the virus, with a particular emphasis on its emergence, global spread, and implications for public health. The review delves into the timelines and key milestones of SARS-CoV-2's epidemiological progression, shedding light on the challenges encountered during early containment efforts and subsequent waves of transmission. Understanding the evolutionary dynamics of the virus is crucial in monitoring its potential for adaptation and future outbreaks. Genetic characterization of SARS-CoV-2 is discussed, with a focus on the emergence of new variants and their implications for transmissibility, severity, and immune evasion. The review highlights the important role of genomic surveillance in tracking viral mutations linked to establishing public health interventions. By analyzing the origins, global spread, and genetic evolution of SARS-CoV-2, valuable insights can be gained for the development of effective control measures, improvement of pandemic preparedness, and addressing future emerging infectious diseases of international concern.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/epidemiology , Pandemics/prevention & control , Public Health , Disease Outbreaks
8.
New Microbiol ; 46(1): 60-64, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36853820

ABSTRACT

In this study we evaluated the antiviral activity of the Silver Barrier® disinfectant against SARSCoV-2. Silver Barrier® showed time- and concentration-dependent antiviral activity against SARSCoV-2. After 5 min contact time, Silver Barrier® at 0.002% showed a strong inhibitory effect (p<0.001), with a 2-fold reduction of viral genome copy numbers, and a robust suppression (94%) of SARS-CoV-2 infectivity. Considering the effects obtained in solution and within a very short time, Silver Barrier® stands as an excellent new candidate for the disinfection of work environments, especially at the healthcare level, where there are people at high risk of serious illnesses.


Subject(s)
COVID-19 , Disinfectants , Humans , SARS-CoV-2 , Disinfectants/pharmacology , COVID-19/prevention & control , Silver/pharmacology , Antiviral Agents/pharmacology
9.
J Biol Chem ; 297(2): 100934, 2021 08.
Article in English | MEDLINE | ID: mdl-34273353

ABSTRACT

HIV-1 matrix protein p17 variants (vp17s) derived from non-Hodgkin's lymphoma (NHL) tissues of HIV-1-seropositive (HIV+) patients promote B-cell growth by activating the Akt signaling pathway. It is fundamental to understand the role played by vp17s in producing a microenvironment that fosters lymphoma development and progression. Therefore, we asked whether vp17s could be secreted from infected cells in their biologically active form. In this study, we show that two B-cell growth-promoting vp17s, NHL-a101 and NHL-a102, characterized by amino acid insertions at position 117 to 118 (Ala-Ala) or 125 to 126 (Gly-Asn), respectively, are secreted from HIV-1-infected Jurkat T cells during the active phase of viral replication. Secretion of biologically active vp17s also occurred in HeLa cells nucleofected with a plasmid expressing the entire Gag gene, following proteolytic cleavage of the Gag precursor polyprotein (Pr55Gag) by cellular aspartyl proteases. Binding of Pr55Gag to phosphatidylinositol-(4,5)-bisphosphate was indispensable for allowing the unconventional secretion of both wildtype p17 and vp17s. Indeed, here we demonstrate that inhibition of Pr55Gag binding to phosphatidylinositol-(4,5)-bisphosphate by using neomycin, or its enzymatic depletion achieved by overexpression of 5ptaseIV, significantly impair the secretion of p17s. We also demonstrated that heparan sulfate proteoglycans were involved in tethering p17s at the cell surface. This finding opens up an interesting way for investigating whether tethered p17s on the surface of HIV-1 reservoirs may represent a likely target for immune-mediated killing.


Subject(s)
B-Lymphocytes , HIV Antigens , HIV-1 , gag Gene Products, Human Immunodeficiency Virus , Cell Membrane/metabolism , HeLa Cells , Humans , Protein Binding , Proteolysis , Signal Transduction , Virus Replication
10.
J Med Virol ; 94(1): 413-416, 2022 01.
Article in English | MEDLINE | ID: mdl-34515998

ABSTRACT

In December 2020, Italy experienced the first case of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) B.1.1.7 lineage. In January 2021, we identified 21 cases of this variant in Corzano, defining the first outbreak of SARS-CoV-2 B.1.1.7 lineage in Italy. The high transmissibility of the B.1.1.7 variant represented an important benefit for the virus, which became rapidly dominant on the territory. Containment measures induced the epidemic curve onto a decreasing trajectory underlining the importance of appropriate control and surveillance for restraint of virus spread. Highlights The first Italian outbreak of SARS-CoV-2 B.1.1.7 lineage occurred in Lombardy in January 2021. The outbreak originated by a single introduction of the B.1.1.7 lineage. The genomic sequencing revealed, for the first time, the presence of the V551F mutation in the B.1.1.7 lineage in Italy. Surveillance, prompt sequencing and tracing efforts were fundamental to identify and to quickly contain the outbreak.


Subject(s)
COVID-19 Nucleic Acid Testing , COVID-19/epidemiology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Adolescent , Adult , COVID-19/transmission , Child , Child, Preschool , Disease Outbreaks/statistics & numerical data , Female , Genome, Viral/genetics , High-Throughput Nucleotide Sequencing , Humans , Infection Control/methods , Italy/epidemiology , Male , Middle Aged , Phylogeny , Sequence Analysis, RNA , Whole Genome Sequencing , Young Adult
11.
J Med Virol ; 94(4): 1689-1692, 2022 04.
Article in English | MEDLINE | ID: mdl-34766651

ABSTRACT

The appearance of emerging variants of SARS-CoV-2 carrying mutations into the spike protein has recently raised concern with respect to tracking their transmission and mitigating the impact in the evolving pandemic across countries. AY.4.2, a recently detected Delta variant sublineage, is considered a new variant under investigation (VUI) as it carries specific genetic signatures present in the spike protein, called Y145H and A222V. Here, using genomic epidemiology, we provide the first preliminary insight regarding the circulation of this emerging VUI in Italy.


Subject(s)
COVID-19/epidemiology , Genome, Viral/genetics , SARS-CoV-2/genetics , Adolescent , Adult , Aged , COVID-19/virology , Child , Female , Genomics , Humans , Italy/epidemiology , Male , Middle Aged , Molecular Epidemiology , Mutation , Phylogeny , RNA, Viral/genetics , SARS-CoV-2/isolation & purification , Young Adult
12.
New Microbiol ; 45(4): 355-357, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36066212

ABSTRACT

Persistence of detectable viral RNA does not depend on the symptomatic status of the patients. Here we describe the case of a strongly immunocompromised patient living with a prolonged SARSCoV-2 Alpha variant infection without showing any symptoms. The importance of our findings is that the persistent infection with an old SARS-CoV-2 strain, in an immunocompromised host, may allow recombination events generating new viral variants whose pathogenicity cannot be predicted. Our observation calls for the urgent need for continuous monitoring of SARS-CoV-2 genomic evolution in immunocompromised patients.


Subject(s)
COVID-19 , HIV Seropositivity , HIV-1 , Humans , SARS-CoV-2/genetics , COVID-19/diagnosis , HIV-1/genetics , Immunocompromised Host
13.
Int J Mol Sci ; 23(23)2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36499592

ABSTRACT

The BQ.1 SARS-CoV-2 variant, also known as Cerberus, is one of the most recent Omicron descendant lineages. Compared to its direct progenitor BA.5, BQ.1 has some additional spike mutations in some key antigenic sites, which confer further immune escape ability over other circulating lineages. In such a context, here, we perform a genome-based survey aimed at obtaining a complete-as-possible nuance of this rapidly evolving Omicron subvariant. Genetic data suggest that BQ.1 represents an evolutionary blind background, lacking the rapid diversification that is typical of a dangerous lineage. Indeed, the evolutionary rate of BQ.1 is very similar to that of BA.5 (7.6 × 10-4 and 7 × 10-4 subs/site/year, respectively), which has been circulating for several months. The Bayesian Skyline Plot reconstruction indicates a low level of genetic variability, suggesting that the peak was reached around 3 September 2022. Concerning the affinity for ACE2, structure analyses (also performed by comparing the properties of BQ.1 and BA.5 RBD) indicate that the impact of the BQ.1 mutations may be modest. Likewise, immunoinformatic analyses showed moderate differences between the BQ.1 and BA5 potential B-cell epitopes. In conclusion, genetic and structural analyses on SARS-CoV-2 BQ.1 suggest no evidence of a particularly dangerous or high expansion capability. Genome-based monitoring must continue uninterrupted for a better understanding of its descendants and all other lineages.


Subject(s)
COVID-19 , Humans , Bayes Theorem , COVID-19/epidemiology , COVID-19/genetics , SARS-CoV-2/genetics , Biological Evolution
14.
J Infect Dis ; 223(1): 28-37, 2021 01 04.
Article in English | MEDLINE | ID: mdl-32986824

ABSTRACT

BACKGROUND: Several preclinical and clinical investigations have argued for nervous system involvement in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Some sparse case reports have described various forms of encephalitis in coronavirus disease 2019 (COVID-19) disease, but very few data have focused on clinical presentations, clinical course, response to treatment, and outcomes. METHODS: The SARS-CoV-2 related encephalopaties (ENCOVID) multicenter study included patients with encephalitis with full infectious screening, cerebrospinal fluid (CSF), electroencephalography (EEG), and magnetic resonance imaging (MRI) data and confirmed SARS-CoV-2 infection recruited from 13 centers in northern Italy. Clinical presentation and laboratory markers, severity of COVID-19 disease, response to treatment, and outcomes were recorded. RESULTS: Twenty-five cases of encephalitis positive for SARS-CoV-2 infection were included. CSF showed hyperproteinorrachia and/or pleocytosis in 68% of cases whereas SARS-CoV-2 RNA by reverse-transcription polymerase chain reaction resulted negative. Based on MRI, cases were classified as acute demyelinating encephalomyelitis (ADEM; n = 3), limbic encephalitis (LE; n = 2), encephalitis with normal imaging (n = 13), and encephalitis with MRI alterations (n = 7). ADEM and LE cases showed a delayed onset compared to the other encephalitis cases (P = .001) and were associated with previous, more severe COVID-19 respiratory involvement. Patients with MRI alterations exhibited worse response to treatment and final outcomes compared to those with other encephalitis. CONCLUSIONS: SARS-CoV-2 infection is associated with a wide spectrum of encephalitis characterized by different clinical presentation, response to treatment, and outcomes.


Subject(s)
COVID-19/complications , Encephalitis/diagnosis , Aged , Aged, 80 and over , COVID-19/therapy , Electroencephalography , Encephalitis/classification , Encephalitis/virology , Female , Humans , Italy , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged
15.
J Med Virol ; 93(3): 1780-1785, 2021 03.
Article in English | MEDLINE | ID: mdl-32926453

ABSTRACT

In early 2020 the new respiratory syndrome COVID-19 (caused by the zoonotic SARS-CoV-2 virus) spread like a pandemic, starting from Wuhan, China, causing severe economic depression. Despite some advances in drug treatments of medical complications in the later stages of the disease, the pandemic's death toll is tragic, as no vaccine or specific antiviral treatment is currently available. By using a systems approach, we identify the host-encoded pathway, which provides ribonucleotides to viral RNA synthesis, as a possible target. We show that methotrexate, an FDA-approved inhibitor of purine biosynthesis, potently inhibits viral RNA replication, viral protein synthesis, and virus release. The effective antiviral methotrexate concentrations are similar to those used for established human therapies using the same drug. Methotrexate should be most effective in patients at the earliest appearance of symptoms to effectively prevent viral replication, diffusion of the infection, and possibly fatal complications.


Subject(s)
Antiviral Agents/pharmacology , COVID-19/etiology , Methotrexate/pharmacology , SARS-CoV-2/drug effects , Virus Replication/drug effects , Animals , COVID-19/virology , Cell Line , Chlorocebus aethiops , Pandemics/prevention & control , RNA, Viral/genetics , Vero Cells
16.
J Transl Med ; 18(1): 362, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32967693

ABSTRACT

BACKGROUND: Since the first outbreak of SARS-CoV-2, the clinical characteristics of the Coronavirus Disease 2019 (COVID-19) have been progressively changed. Data reporting a viral intra-host and inter-host evolution favouring the appearance of mild SARS-CoV-2 strains are since being accumulating. To better understand the evolution of SARS-CoV-2 pathogenicity and its adaptation to the host, it is therefore crucial to investigate the genetic and phenotypic characteristics of SARS-CoV-2 strains circulating lately in the epidemic. METHODS: Nasopharyngeal swabs have been analyzed for viral load in the early (March 2020) and late (May 2020) phases of epidemic in Brescia, Italy. Isolation of SARS-CoV-2 from 2 high viral load specimens identified on March 9 (AP66) and on May 8 (GZ69) was performed on Vero E6 cells. Amount of virus released was assessed by quantitative PCR. Genotypic characterization of AP66 and GZ69 was performed by next generation sequencing followed by an in-depth in silico analysis of nucleotide mutations. RESULTS: The SARS-CoV-2 GZ69 strain, isolated in May from an asymptomatic healthcare worker, showed an unprecedented capability of replication in Vero E6 cells in the absence of any evident cytopathic effect. Vero E6 subculturing, up to passage 4, showed that SARS-CoV-2 GZ69 infection was as productive as the one sustained by the cytopathic strain AP66. Whole genome sequencing of the persistently replicating SARS-CoV-2 GZ69 has shown that this strain differs from the early AP66 variant in 9 nucleotide positions (C2939T; C3828T; G21784T; T21846C; T24631C; G28881A; G28882A; G28883C; G29810T) which lead to 6 non-synonymous substitutions spanning on ORF1ab (P892S; S1188L), S (K74N; I95T) and N (R203K, G204R) proteins. CONCLUSIONS: Identification of the peculiar SARS-CoV-2 GZ69 strain in the late Italian epidemic highlights the need to better characterize viral variants circulating among asymptomatic or paucisymptomatic individuals. The current approach could unravel the ways for future studies aimed at analyzing the selection process which favours viral mutations in the human host.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Genetic Variation , Pneumonia, Viral/virology , Amino Acid Substitution , Animals , Betacoronavirus/isolation & purification , Betacoronavirus/physiology , COVID-19 , Chlorocebus aethiops , Coronavirus Infections/epidemiology , Cytopathogenic Effect, Viral/genetics , Cytopathogenic Effect, Viral/physiology , Genome, Viral , Humans , Italy/epidemiology , Mutation , Pandemics , Phylogeny , Pneumonia, Viral/epidemiology , Polymorphism, Single Nucleotide , SARS-CoV-2 , Translational Research, Biomedical , Vero Cells , Viral Proteins/genetics , Viral Proteins/physiology , Virus Cultivation/methods , Virus Replication/genetics , Virus Replication/physiology , Whole Genome Sequencing
17.
Int J Mol Sci ; 21(6)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32188077

ABSTRACT

Although the advent of combined antiretroviral therapy has substantially improved the survival of HIV-1-infected individuals, non-AIDS-related diseases are becoming increasingly prevalent in HIV-1-infected patients. Persistent abnormalities in coagulation appear to contribute to excess risk for a broad spectrum of non-AIDS defining complications. Alterations in coagulation biology in the context of HIV infection seem to be largely a consequence of a chronically inflammatory microenvironment leading to endothelial cell (EC) dysfunction. A possible direct role of HIV-1 proteins in sustaining EC dysfunction has been postulated but not yet investigated. The HIV-1 matrix protein p17 (p17) is secreted from HIV-1-infected cells and is known to sustain inflammatory processes by activating ECs. The aim of this study was to investigate the possibility that p17-driven stimulation of human ECs is associated with increased production of critical coagulation factors. Here we show the involvement of autophagy in the p17-induced accumulation and secretion of von Willebrand factor (vWF) by ECs. In vivo experiments confirmed the capability of p17 to exert a potent pro-coagulant activity soon after its intravenous administration.


Subject(s)
Antithrombin III/metabolism , Autophagy/physiology , Endothelial Cells/metabolism , HIV Antigens/metabolism , Peptide Hydrolases/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , von Willebrand Factor/metabolism , Animals , Anti-Retroviral Agents/therapeutic use , Female , HIV Infections/complications , HIV-1/physiology , Humans , Mice
18.
Lab Invest ; 99(2): 180-190, 2019 02.
Article in English | MEDLINE | ID: mdl-30390010

ABSTRACT

HIV-associated neurocognitive disorder in HIV patients substantially reduces their quality of life. We previously showed that the HIV matrix protein, p17 could stimulate lymph-angiogenesis in vitro potentially contributing to lymphoma tumour growth and in addition is associated with vascular activation in neuro-degenerating brain tissue; here, therefore, we have investigated the detailed molecular mechanisms of this action. We performed in vitro cell culture, angiogenesis experiments, phospho-protein microarrays and Western blotting to identify cellular signalling induced by p17 within human brain endothelial cells (HbMEC), and inhibitor studies to block p17-induced vascular growth. We also characterised the effects of hippocampal CA1 injection of p17 on epidermal growth factor receptor-1 (EGFR1) expression linked to our murine model of dementia. p17 strongly induced angiogenesis of HbMEC (migration, tube formation and spheroid growth). p17 concomitantly increased phosphorylation of EGFR1 as well as down-stream intermediates ERK1/2, FAK, PLC-γ and PKC-ß whilst an inhibitor peptide of EGFR, blocked cell signalling and angiogenesis. Finally, Mice that showed reduced cognitive function and behavioural deficiencies after p17 injection, demonstrated that p17 localised in cortical microvessels and also neurones many of which stained positive for p-EGFR1 by histology/IHC. This work provides strong support that p17 may be involved in initiating and/or perpetuating vascular tissue pathophysiology associated with comorbidity in HIV patients.


Subject(s)
Brain/cytology , Endothelial Cells/drug effects , ErbB Receptors/metabolism , HIV Antigens/pharmacology , Neovascularization, Pathologic/chemically induced , gag Gene Products, Human Immunodeficiency Virus/pharmacology , Animals , Humans , Mice , Signal Transduction/drug effects
19.
Hematol Oncol ; 37(2): 176-184, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30261551

ABSTRACT

Despite antiretroviral therapy, HIV+ individuals still have increased risk to develop lymphomas, including marginal zone lymphomas, suggesting that factors other than HIV-related immunosuppression are probably acting as lymphomagenic factors in the HIV setting. The possible pathogenic involvement of HIV p17 protein variants was investigated in a particularly informative case of HIV-related splenic marginal zone lymphoma, which was negative for oncogenic virus infections, thus allowing us to assess the possible direct contribution of these HIV-encoded proteins to lymphomagenesis. The presence of p17 protein was analyzed by immunohistochemistry in lymphoma tissue. Recombinant p17 protein derived from the dominant sequence detected in plasma and lymphoma biopsy was characterized for B-cell proliferation, clonogenicity in soft agar, in vitro tube formation and wound healing. Intracellular signaling was investigated by immunoblotting. HIV p17 protein was detected in reactive lymphoid follicles but not within lymphoma cells. An identical dominant variant p17 sequence, p17-Lyrm, carrying a 117 to 118 Ala-Ala insertion was detected in both plasma and lymphoma tissue. Recombinant p17-Lyrm enhanced B-cell proliferation and clonogenicity promoted the formation of capillary-like structures and enhanced endothelial cell migration. Unlike reference p17, the p17-Lyrm variant enhanced the activation of Akt and ERK, critical kinases in lymphomagenesis. p17-Lyrm clonogenic activity was dependent on the activation of Akt but not of ERK1/2. These results indicated that HIV p17 variants with distinct molecular signatures and functional properties may accumulate in lymphoid tissues of HIV-infected individuals where they may act as a local stimulus promoting the development of lymphomas.


Subject(s)
Cell Transformation, Viral , HIV Antigens , HIV Infections , HIV-1 , Lymphoma, B-Cell, Marginal Zone , Mutagenesis, Insertional , Splenic Neoplasms , gag Gene Products, Human Immunodeficiency Virus , Female , HIV Antigens/genetics , HIV Antigens/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/genetics , HIV-1/metabolism , Humans , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, B-Cell, Marginal Zone/pathology , Lymphoma, B-Cell, Marginal Zone/virology , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Splenic Neoplasms/genetics , Splenic Neoplasms/metabolism , Splenic Neoplasms/pathology , Splenic Neoplasms/virology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
20.
Int J Mol Sci ; 20(21)2019 Oct 24.
Article in English | MEDLINE | ID: mdl-31653015

ABSTRACT

In recent years immunomodulators have gained a strong interest and represent nowadays an active expanding area of research for the control of microbial diseases and for their therapeutic potential in preventing, treating and reducing the morbidity and mortality of different diseases. Pidotimod (3-L-pyroglutamyl-L-thiaziolidine-4carboxylic acid, PDT) is a synthetic dipeptide, which possesses immunomodulatory properties and exerts a well-defined pharmacological activity against infections, but its real mechanism of action is still undefined. Here, we show that PDT is capable of activating tyrosine phosphorylation-based cell signaling in human primary monocytes and triggering rapid adhesion and chemotaxis. PDT-induced monocyte migration requires the activation of the PI3K/Akt signaling pathway and chemokine receptor CXCR3. Indeed, a mAb to CXCR3 and a specific receptor inhibitor suppressed significantly PDT-dependent chemotaxis, and CXCR3-silenced primary monocytes lost responsiveness to PDT chemoattraction. Moreover, our results highlighted that the PDT-induced migratory activity is sustained by the CXCR3A isoform, since CXCR3-transfected L1.2 cells acquired responsiveness to PDT stimulation. Finally, we show that PDT, as CXCR3 ligands, is also able to direct the migration of IL-2 activated T cells, which express the highest levels of CXCR3 among CXCR3-expressing cells. In conclusion, our study defines a chemokine-like activity for PDT through CXCR3A and points on the possible role that this synthetic dipeptide may play in leukocyte trafficking and function. Since recent studies have highlighted diverse therapeutic roles for molecules which activates CXCR3, our findings call for an exploration of using this dipeptide in different pathological processes.


Subject(s)
Monocytes/drug effects , Pyrrolidonecarboxylic Acid/analogs & derivatives , Receptors, CXCR3/metabolism , Thiazolidines/pharmacology , Antibodies, Monoclonal/immunology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemotaxis/drug effects , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Humans , Monocytes/cytology , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrrolidonecarboxylic Acid/chemical synthesis , Pyrrolidonecarboxylic Acid/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, CXCR3/antagonists & inhibitors , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thiazolidines/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL