[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (6)

Search Parameters:
Keywords = iciHHV-6

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 926 KiB  
Article
Human Herpesvirus 6—A Rare Aetiologic Agent for CNS Infections in Immunocompetent Individuals or an Underestimation?
by Oana Alexandra Ganea, Cătălin Tilișcan, Anca Streinu-Cercel, Daniela Pițigoi, Anca Cristina Drăgănescu, Mihai Lazar, Nicoleta Mihai, Dragoș Florea, Sorin Ștefan Aramă and Victoria Aramă
J. Clin. Med. 2024, 13(16), 4660; https://doi.org/10.3390/jcm13164660 - 8 Aug 2024
Viewed by 371
Abstract
Background: Human herpesvirus 6 (HHV-6) is considered a ubiquitous virus, with many countries reporting a seroprevalence of more than 80–90% among the general population. However, this virus is unique among herpesviruses in its ability to integrate into the genetic material of the [...] Read more.
Background: Human herpesvirus 6 (HHV-6) is considered a ubiquitous virus, with many countries reporting a seroprevalence of more than 80–90% among the general population. However, this virus is unique among herpesviruses in its ability to integrate into the genetic material of the host’s cells. Thus, there are three ways by which HHV-6 can cause an active infection–primary infection, reactivation of a latent acquired infection, or activation of iciHHV-6 (inherited chromosomally integrated HHV-6). Whole blood quantitative polymerase chain reaction (qPCR) is very useful in distinguishing between iciHHV-6 and primary infection/reactivation. Our aim is to assess the role of HHV-6 in the aetiology of central nervous system (CNS) infections in adults and children, to describe all HHV-6-positive cases in an attempt to determine the susceptible population and to identify potential risk factors that can be linked to HHV-6 meningoencephalitis. Methods: We performed a retrospective study involving patients that were admitted to Prof. Dr. Matei Bals National Institute of Infectious Diseases, Bucharest, Romania, with a diagnosis of meningitis or encephalitis. We only selected the clinical records of patients that had a multiplex PCR Biofire® FilmArray® meningitis/encephalitis panel. Results: We report a 5% HHV-6 positivity in the cerebrospinal fluid (CSF) of patients with CNS infections tested with a commercial multiplex PCR M/E (meningitis/encephalitis) panel. Additionally, 2% to 4% of the total study population (n = 100) had active HHV-6 infections, which denotes 40 to 80% of the HHV-6-positive samples. We did not observe any statistically significant correlation between HHV-6 positivity in the CSF and variables such as age, sex, or comorbidities, including obesity, diabetes, hypertension, immunosuppression, or oncologic disease. Therefore, no risk factors could be linked with HHV-6 positivity in the CSF. Conclusions: although multiplex qualitative PCR is highly useful for providing rapid results and identifying nearly every pathogen that can cause meningitis/encephalitis, we have to be aware of this type of test’s limitations. All patients with HHV-6 detectable in their CSF via a multiplex PCR test should also undergo qPCR testing from both CSF and blood to prevent over-diagnosing HHV-6 CNS infections, to avoid unnecessary antiviral treatments, and ensure the accurate identification of the true diagnosis. Full article
(This article belongs to the Section Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Study population description. * M/E = meningitis/encephalitis/meningoencephalitis.</p>
Full article ">Figure 2
<p>Aetiologic agents identified in our cohort.</p>
Full article ">
15 pages, 536 KiB  
Article
Inherited Chromosomally Integrated Human Herpesvirus 6: Laboratory and Clinical Features
by Liliana Gabrielli, Alice Balboni, Eva Caterina Borgatti, Giulio Virgili, Evangelia Petrisli, Alessia Cantiani, Matteo Pavoni, Federico Baiesi Pillastrini, Simona Venturoli, Giulia Piccirilli and Tiziana Lazzarotto
Microorganisms 2023, 11(3), 548; https://doi.org/10.3390/microorganisms11030548 - 21 Feb 2023
Cited by 3 | Viewed by 2792
Abstract
Inherited chromosomally integrated human herpesvirus 6 (iciHHV-6) is a condition in which the complete HHV-6 genome is integrated into the chromosomes of the host germ cell and is vertically transmitted. The aims of this study were to identify iciHHV-6 prevalence in hospitalized patients [...] Read more.
Inherited chromosomally integrated human herpesvirus 6 (iciHHV-6) is a condition in which the complete HHV-6 genome is integrated into the chromosomes of the host germ cell and is vertically transmitted. The aims of this study were to identify iciHHV-6 prevalence in hospitalized patients and clinical features in individuals carrying this integration. HHV-6 PCR on hair follicles was used to confirm iciHHV-6 status when the blood viral load was more than 5 Log10 copies/mL. From January 2012 to June 2022, HHV-6 DNAemia was investigated in 2019 patients. In particular, 49 had a viral load higher than 6 Log10 copies/mL and HHV-6 DNA in hair follicles was positive. A viral load between 5.0 and 5.9 Log10 copies/mL was observed in 10 patients: 6 infants with acute HHV-6 infection and 4 patients with leukopenia and HHV-6 integration. Therefore, the iciHHV-6 prevalence in our population was 2.6% (53/2019). Adult patients with integration presented hematological (24%), autoimmune (11%), autoimmune neurological (19%), not-autoimmune neurological (22%), and other diseases (19%), whereas 5% had no clinically relevant disease. Although in our study population a high percentage of iciHHV-6 adult hospitalized patients presented a specific pathology, it is still unknown whether the integration is responsible for, or contributes to, the disease development. Full article
Show Figures

Figure 1

Figure 1
<p>Monitoring of blood HHV-6 DNA post-hematopoietic stem cell transplantation (HSCT) in a patient with iciHHV-6 who received a graft from a donor without iciHHV-6. HHV-6 DNA was related to white blood cell (WBC) count in the early period after transplantation.</p>
Full article ">
10 pages, 800 KiB  
Case Report
Investigation of Inherited Chromosomally Integrated Human Herpesvirus-6A+ and -6B+ in a Patient with Ulipristal Acetate-Induced Fulminant Hepatic Failure
by Laure Izquierdo, Clémence M. Canivet, Eleonora De Martin, Teresa M. Antonini, Anne-Marie Roque-Afonso, Audrey Coilly and Claire Deback
Viruses 2022, 14(1), 62; https://doi.org/10.3390/v14010062 - 30 Dec 2021
Cited by 2 | Viewed by 2135
Abstract
Inherited chromosomally integrated (ici) human herpes virus 6 (HHV-6) is estimated to occur in 0.6–2.7% of people worldwide. HHV-6 comprises two distinct species: HHV-6A and HHV-6B. Both HHV-6A and HHV-6B integration have been reported. Several drugs are capable of activating iciHHV-6 in tissues, [...] Read more.
Inherited chromosomally integrated (ici) human herpes virus 6 (HHV-6) is estimated to occur in 0.6–2.7% of people worldwide. HHV-6 comprises two distinct species: HHV-6A and HHV-6B. Both HHV-6A and HHV-6B integration have been reported. Several drugs are capable of activating iciHHV-6 in tissues, the consequences of which are poorly understood. We report herein a case of a woman with iciHHV-6A+ and iciHHV-6B+, who developed ulipristal acetate (a selective progesterone receptor modulator)-induced fulminant hepatic failure that required liver transplantation. We confirmed the presence of ~one copy per cell of both HHV-6A and HHV-6B DNA in her hair follicles using multiplex HHV-6A/B real-time PCR and demonstrated the Mendelian inheritance of both iciHHV-6A and iciHHV-6B in her family members over three generations. Because of the rarity of this presentation, we discuss herein the possible links between reactivated HHV-6 from iciHHV-6A and/or iciHHV-6B and adverse drug reactions, suggesting that iciHHV-6 could be screened before the introduction of any hepatotoxic drugs to exclude HHV-6 active disease or combined idiosyncratic drug-induced liver injury in these patients. Full article
(This article belongs to the Topic Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Kinetics of biomarkers and samples. (<b>A</b>) Loads of HHV-6A and -6B, expressed as log<sub>10</sub> copies/mL of whole blood, before and after liver transplantation. The gray zone corresponds to two weeks of ganciclovir treatment. Hair follicle analysis for iciHHV-6A and/or iciHHV-6B (✯) was performed on the seventh day of the treatment; (<b>B</b>) ALT (IU/L), PR (%) and factor V (%) kinetics during the follow-up period. The gray zone corresponds to 2 weeks of ganciclovir treatment. ALT, alanine transaminase; PR, prothrombin ratio.</p>
Full article ">Figure 2
<p>(<b>A</b>) Genealogical tree of the family harboring iciHHV-6A and/or iciHHV-6B. The arrow indicates the consultant patient (II:2) suffering from fulminant hepatic failure. (<b>B</b>) iciHHV-6A and/or iciHHV-6B viral loads determined in hair follicles and expressed in copy number per cell are indicated for each family member. ID, identification; y, year.</p>
Full article ">
10 pages, 1919 KiB  
Article
Viral Proteins U41 and U70 of Human Herpesvirus 6A Are Dispensable for Telomere Integration
by Darren J. Wight, Nina Wallaschek, Anirban Sanyal, Sandra K. Weller, Louis Flamand and Benedikt B. Kaufer
Viruses 2018, 10(11), 656; https://doi.org/10.3390/v10110656 - 21 Nov 2018
Cited by 14 | Viewed by 4804
Abstract
Human herpesvirus-6A and -6B (HHV-6A and -6B) are two closely related betaherpesviruses that infect humans. Upon primary infection they establish a life-long infection termed latency, where the virus genome is integrated into the telomeres of latently infected cells. Intriguingly, HHV-6A/B can integrate into [...] Read more.
Human herpesvirus-6A and -6B (HHV-6A and -6B) are two closely related betaherpesviruses that infect humans. Upon primary infection they establish a life-long infection termed latency, where the virus genome is integrated into the telomeres of latently infected cells. Intriguingly, HHV-6A/B can integrate into germ cells, leading to individuals with inherited chromosomally-integrated HHV-6 (iciHHV-6), who have the HHV-6 genome in every cell. It is known that telomeric repeats flanking the virus genome are essential for integration; however, the protein factors mediating integration remain enigmatic. We have previously shown that the putative viral integrase U94 is not essential for telomere integration; thus, we set out to assess the contribution of potential viral recombination proteins U41 and U70 towards integration. We could show that U70 enhances dsDNA break repair via a homology-directed mechanism using a reporter cell line. We then engineered cells to produce shRNAs targeting both U41 and U70 to inhibit their expression during infection. Using these cells in our HHV-6A in vitro integration assay, we could show that U41/U70 were dispensable for telomere integration. Furthermore, additional inhibition of the cellular recombinase Rad51 suggested that it was also not essential, indicating that other cellular and/or viral factors must mediate telomere integration. Full article
(This article belongs to the Section Animal Viruses)
Show Figures

Figure 1

Figure 1
<p>The role of HHV-6A U70 and U41 in double strand DNA break repair by single strand annealing (SSA). (<b>A</b>) An SSA reporter was integrated into 293T cells and the cells were transfected with the indicated expression plasmids. The frequency of repair was calculated and normalized to the mock transfected cells (control plasmid). Displayed are the mean frequency of repair for each transfected construct (<span class="html-italic">n</span> = 3 ± SEM). (<b>B</b>) The C-terminal HA-tag on U70 was removed and the same assay as in (<b>A</b>) was performed (<span class="html-italic">n</span> = 3 ± SEM).</p>
Full article ">Figure 2
<p>Validation of 293T HHV-6A U41/U70 knockdown cells. (<b>A</b>) 293T, Kd poly or single cell clones were individually transfected with U41-HA (upper panels) or U70-HA (lower panels) expression plasmids. One day post-transfection, cells were lysed and the proteins separated by SDS-PAGE. Immunoblotting was then performed using anti-HA or anti-beta-actin (beta-actin; loading control) antibodies. (<b>B</b>) Optical densities for HA bands were extracted using BIO-1D software and normalized for protein loading (β-actin). Displayed in the histogram is the viral protein expression in the poly clonal cells (Kd poly) and the best clonal cell (Kd C5) relative to expression in 293T control cells.</p>
Full article ">Figure 3
<p>The effect of HHV-6A U41/U70 knockdown on HHV-6A integration. 293T or Kd polyclonal (Kd poly) cells were infected with the HHV-6A ∆U94 virus and the GFP-positive cells sorted. Samples were taken immediately after sorting and 14 days later. (<b>A</b>) Mean virus DNA copies per million cells was quantified by qPCR using primers against viral U86 and cellular B2M. This number is displayed in the histogram (<span class="html-italic">n</span> = 4 ± SEM). RI-1 is a specific inhibitor of the Rad51 cellular recombinase. (<b>B</b>) The 14-day samples were analyzed by FISH to detect the HHV-6A genome (green). Representative images are displayed for metaphase and interphase cells (DAPI staining shown in grey). Red arrows indicate the location of HHV-6A signals. Scale bars for the top and bottom images are 10 µm and 4 µm for the images in the middle.</p>
Full article ">Figure 4
<p>The effect of HHV-6A U41/U70 knockdown on telomere integration in a clonal knockdown cell line. The integration assay was performed as described in <a href="#viruses-10-00656-f003" class="html-fig">Figure 3</a>A but using the U41/U70 single cell clone (Kd C5). Mean virus DNA copies per million cells was quantified by qPCR against viral U86 and cellular B2M. This number is shown in the histogram (<span class="html-italic">n</span> = 3 ± SEM).</p>
Full article ">
751 KiB  
Review
Latency, Integration, and Reactivation of Human Herpesvirus-6
by Shara N. Pantry and Peter G. Medveczky
Viruses 2017, 9(7), 194; https://doi.org/10.3390/v9070194 - 24 Jul 2017
Cited by 75 | Viewed by 9910
Abstract
Human herpesvirus-6A (HHV-6A) and human herpesvirus-6B (HHV-6B) are two closely related viruses that infect T-cells. Both HHV-6A and HHV-6B possess telomere-like repeats at the terminal regions of their genomes that facilitate latency by integration into the host telomeres, rather than by episome formation. [...] Read more.
Human herpesvirus-6A (HHV-6A) and human herpesvirus-6B (HHV-6B) are two closely related viruses that infect T-cells. Both HHV-6A and HHV-6B possess telomere-like repeats at the terminal regions of their genomes that facilitate latency by integration into the host telomeres, rather than by episome formation. In about 1% of the human population, human herpes virus-6 (HHV-6) integration into germline cells allows the viral genome to be passed down from one generation to the other; this condition is called inherited chromosomally integrated HHV-6 (iciHHV-6). This review will cover the history of HHV-6 and recent works that define the biological differences between HHV-6A and HHV-6B. Additionally, HHV-6 integration and inheritance, the capacity for reactivation and superinfection of iciHHV-6 individuals with a second strain of HHV-6, and the role of hypomethylation of human chromosomes during integration are discussed. Overall, the data suggest that integration of HHV-6 in telomeres represent a unique mechanism of viral latency and offers a novel tool to study not only HHV-6 pathogenesis, but also telomere biology. Paradoxically, the integrated viral genome is often defective especially as seen in iciHHV-6 harboring individuals. Finally, gaps in the field of HHV-6 research are presented and future studies are proposed. Full article
(This article belongs to the Special Issue Viruses and Telomeres)
Show Figures

Figure 1

Figure 1
<p>Genome organization of human herpesvirus-6 (HHV-6). Solid lines represent the unique length (U) regions of the genome, boxed regions represent repetitive elements of the genome, and the black boxes represent the perfect telomere repeats, while the gray boxes represent the imperfect telomere repeats. DR<sub>L</sub>: Direct repeat left; DR<sub>R</sub>: Direct repeat right; pac1, pac2: Packaging signals; R0–3: Repetitive elements.</p>
Full article ">Figure 2
<p>Model for integration and reactivation of HHV-6. The linear genome of HHV-6 integrates into the telomeres of the host chromosome, and during reactivation the integrated genome is liberated from the host chromosome, forming a circular intermediate. Rolling circle replication of the circular intermediate generates concatemers of the viral genome.</p>
Full article ">Figure 3
<p>Possible integration configurations of the HHV-6 genome. (<b>A</b>) The HHV-6 genome contains four telomere-like repeats flanking the direct repeats; (<b>B</b>) Integration of the HHV-6 genome by recombination with the host telomeres can potentially occur at either of the four telomere repeats and five possible integration configurations may be generated.</p>
Full article ">
2293 KiB  
Review
HHV-6A/B Integration and the Pathogenesis Associated with the Reactivation of Chromosomally Integrated HHV-6A/B
by Vanessa Collin and Louis Flamand
Viruses 2017, 9(7), 160; https://doi.org/10.3390/v9070160 - 26 Jun 2017
Cited by 24 | Viewed by 6420
Abstract
Unlike other human herpesviruses, human herpesvirus 6A and 6B (HHV-6A/B) infection can lead to integration of the viral genome in human chromosomes. When integration occurs in germinal cells, the integrated HHV-6A/B genome can be transmitted to 50% of descendants. Such individuals, carrying one [...] Read more.
Unlike other human herpesviruses, human herpesvirus 6A and 6B (HHV-6A/B) infection can lead to integration of the viral genome in human chromosomes. When integration occurs in germinal cells, the integrated HHV-6A/B genome can be transmitted to 50% of descendants. Such individuals, carrying one copy of the HHV-6A/B genome in every cell, are referred to as having inherited chromosomally-integrated HHV-6A/B (iciHHV-6) and represent approximately 1% of the world’s population. Interestingly, HHV-6A/B integrate their genomes in a specific region of the chromosomes known as telomeres. Telomeres are located at chromosomes’ ends and play essential roles in chromosomal stability and the long-term proliferative potential of cells. Considering that the integrated HHV-6A/B genome is mostly intact without any gross rearrangements or deletions, integration is likely used for viral maintenance into host cells. Knowing the roles played by telomeres in cellular homeostasis, viral integration in such structure is not likely to be without consequences. At present, the mechanisms and factors involved in HHV-6A/B integration remain poorly defined. In this review, we detail the potential biological and medical impacts of HHV-6A/B integration as well as the possible chromosomal integration and viral excision processes. Full article
(This article belongs to the Special Issue Viruses and Telomeres)
Show Figures

Figure 1

Figure 1
<p>HHV-6A/B genomes and their integrated forms. Schematic representation of human herpesvirus 6A and 6B (HHV-6A/B) genomes and the reported integrated forms. (<b>A</b>) The unique region (U) of the 160 kbp HHV-6A/B genomes is flanked by identical direct repeats (DR<sub>L</sub> and DR<sub>R</sub>) of 8–9 kbp. The DRs possess a <span class="html-italic">pac1</span> (yellow) and <span class="html-italic">pac2</span> (red) sequences, adjacent to imperfect telomeric repeats impTMR (blue) and TMR (green) sequences, respectively. The genome is not drawn to scale; (<b>B</b>) Chromosomally integrated HHV-6A/B (ciHHV-6A/B) genome (with loss of <span class="html-italic">pac2</span> in DR<sub>R</sub> and <span class="html-italic">pac1</span> in DR<sub>L</sub>) with elongated telomeres at the DR<sub>L</sub>; (<b>C</b>) Single integrated DR<sub>L</sub> with elongated telomere; (<b>D</b>) Integrated HHV-6A/B concatemers. Genomes are not drawn to scale.</p>
Full article ">Figure 2
<p>Possible mechanisms of HHV-6A/B genome excision from telomeres. Schematic representation of hypothetic processes of HHV-6A/B genome excision from telomeres. (<b>A</b>) Telomeric repeats form a t-loop in the TMR of HHV-6A/B DR<sub>L</sub>, followed by recombination and excision, resulting into a first t-loop excision: a telomeric circle and a chromosomally integrated HHV-6A/B lacking a DR but still possessing TMR sequences. (<b>B</b>) A second t-loop formation is made by recombination of the TMR at the end of the genome into HHV-6A/B DR<sub>R</sub>, resulting in a fully excised and circular HHV-6A/B genome containing a single DR with a single <span class="html-italic">pac1</span>, <span class="html-italic">pac2</span>, impTMR and TMR sequence. (<b>C</b>) Invasion of the telomeric repeats into the TMR of the DR<sub>R</sub>, resulting into a HHV-6A/B free chromosome and a full viral genome with a complete DR.</p>
Full article ">Figure 3
<p>Possible mechanisms for HHV-6A/B integration. Schematic representation of HHV-6A/B chromosomal integration process. (<b>A</b>) Unfolding of the chromosome t-loop and invasion by the telomeric 3′ overhang into HHV-6A/B’s DR. This mechanism is unlikely to occur since all ciHHV-6A/B reported so far have lost most telomeric repeats. (<b>B</b>) Break induced replication (BIR) repair mechanism caused by G quadruplexes (G4) structure (or other blockage) in the lagging strand. (<b>C</b>) The free 3′ strand is rescued by Rad51 protein that searches for proximal homologous sequences. If a HHV-6A/B genome is close to proximity, Rad51 invades the viral TMR, displacing one strand of the HHV-6A/B genome to allow the synthesis of the complementary strand. Upon cell divisions, the DR<sub>L</sub> would lose <span class="html-italic">pac1</span> due to end replication problem and the impTMR would serve as telomeric template to elongate telomeres at the end of the genome. (<b>D</b>) Single stranded annealing (SSA) repair mechanism. Upon virus entry in the cell, DNA damage response is triggered, at the same time a break caused by a stalled replication fork at the human telomeres activate SSA. SSA activation leads to resection of both the viral and human DNA in a 5′ to 3′ direction to create complementary sequences. Meanwhile, the 3′ strands of both genomes are protected by the replication protein A (RPA). Rasd52 binds the RPA and searched for pairing in which <span class="html-italic">pac2</span> will be lost. Annealed sequences then lead to the copying of the viral genome. Genomes are not drawn to scale.</p>
Full article ">
Back to TopTop