ijms-logo

Journal Browser

Journal Browser

Host and Human Oncovirus Interaction

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Microbiology".

Deadline for manuscript submissions: closed (25 July 2023) | Viewed by 21763

Special Issue Editors


E-Mail Website
Guest Editor
Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
Interests: HTLV-1; viral protein functions; viral replication; maintenance of latency; immune escaping; ATL; viral oncogenesis; antibody-drug conjugate (ADC); host mRNA quality control (nonsense-mediated mRNA decay, NMD)

E-Mail Website
Guest Editor
Department of Practical Management of Medical Information, Graduate School of Medicine, St. Marianna University, Kawasaki 216-8511, Japan
Interests: HTLV-1; ATL; hematology; oncology; epidemiology; medical treatment

Special Issue Information

Dear Colleagues,

This special issue is open to all authors, who are interested in any aspects of interactions between the host and human oncovirus, e.g. HTLV-1, HIV-1, HPV, HBV, and EBV. The direction of interactions can be on both sides, i.e., from the virus to the host or from the host to the virus. Especially, research articles and reviews on the virus-host interactions in the following fields are welcome; 1) Molecular biology of viral replication in the host cell, 2) Influences of viral replication in the host cells and its implication to the onset of diseases, 3) Cellular responses to viral infection, and 4) Immune responses against infected cells. Also, topics in 5) Virus-host co-evolution and viral pathogenesis will be an important part of this special issue. Furthermore, we welcome manuscripts in 6) Development of therapeutic approaches for the elimination of viruses and for virus-caused diseases including anti-virus drugs, vaccinations, and neutralizing antibodies.

Dr. Nakano Kazumi
Prof. Dr. Toshiki Watanabe
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • human oncoviruses
  • functions of viral proteins
  • viral replication
  • latency
  • oncogenesis
  • host cell responses
  • host cell genetic manipulations
  • inflammation
  • immune responses
  • immune escaping of infected cells
  • co-evolution and mutations in the viral genome
  • neutralizing antibody

Published Papers (12 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

20 pages, 5017 KiB  
Article
The Pleiotropic Effects of YBX1 on HTLV-1 Transcription
by Susan Smith, Jaideep Seth, Amanda Midkiff, Rachel Stahl, Yu-Ci Syu, Nikoloz Shkriabai, Mamuka Kvaratskhelia, Karin Musier-Forsyth, Pooja Jain, Patrick L. Green and Amanda R. Panfil
Int. J. Mol. Sci. 2023, 24(17), 13119; https://doi.org/10.3390/ijms241713119 - 23 Aug 2023
Cited by 1 | Viewed by 1074
Abstract
HTLV-1 is an oncogenic human retrovirus and the etiologic agent of the highly aggressive ATL malignancy. Two viral genes, Tax and Hbz, are individually linked to oncogenic transformation and play an important role in the pathogenic process. Consequently, regulation of HTLV-1 gene [...] Read more.
HTLV-1 is an oncogenic human retrovirus and the etiologic agent of the highly aggressive ATL malignancy. Two viral genes, Tax and Hbz, are individually linked to oncogenic transformation and play an important role in the pathogenic process. Consequently, regulation of HTLV-1 gene expression is a central feature in the viral lifecycle and directly contributes to its pathogenic potential. Herein, we identified the cellular transcription factor YBX1 as a binding partner for HBZ. We found YBX1 activated transcription and enhanced Tax-mediated transcription from the viral 5′ LTR promoter. Interestingly, YBX1 also interacted with Tax. shRNA-mediated loss of YBX1 decreased transcript and protein abundance of both Tax and HBZ in HTLV-1-transformed T-cell lines, as well as Tax association with the 5′ LTR. Conversely, YBX1 transcriptional activation of the 5′ LTR promoter was increased in the absence of HBZ. YBX1 was found to be associated with both the 5′ and 3′ LTRs in HTLV-1-transformed and ATL-derived T-cell lines. Together, these data suggest that YBX1 positively influences transcription from both the 5′ and 3′ promoter elements. YBX1 is able to interact with Tax and help recruit Tax to the 5′ LTR. However, through interactions with HBZ, YBX1 transcriptional activation of the 5′ LTR is repressed. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

22 pages, 1470 KiB  
Article
Human Ad19a/64 HERV-W Vaccines Uncover Immunosuppression Domain-Dependent T-Cell Response Differences in Inbred Mice
by Isabella Skandorff, Emeline Ragonnaud, Jasmin Gille, Anne-Marie Andersson, Silke Schrödel, Lara Duvnjak, Louise Turner, Christian Thirion, Ralf Wagner and Peter Johannes Holst
Int. J. Mol. Sci. 2023, 24(12), 9972; https://doi.org/10.3390/ijms24129972 - 09 Jun 2023
Cited by 1 | Viewed by 1383
Abstract
Expression of human endogenous retrovirus type W (HERV-W) has been linked to cancer, making HERV-W antigens potential targets for therapeutic cancer vaccines. In a previous study, we effectively treated established tumours in mice by using adenoviral-vectored vaccines targeting the murine endogenous retrovirus envelope [...] Read more.
Expression of human endogenous retrovirus type W (HERV-W) has been linked to cancer, making HERV-W antigens potential targets for therapeutic cancer vaccines. In a previous study, we effectively treated established tumours in mice by using adenoviral-vectored vaccines targeting the murine endogenous retrovirus envelope and group-specific antigen (Gag) of melanoma-associated retrovirus (MelARV) in combination with anti-PD-1. To break the immunological tolerance to MelARV, we mutated the immunosuppressive domain (ISD) of the MelARV envelope. However, reports on the immunogenicity of the HERV-W envelope, Syncytin-1, and its ISD are conflicting. To identify the most effective HERV-W cancer vaccine candidate, we evaluated the immunogenicity of vaccines encoding either the wild-type or mutated HERV-W envelope ISD in vitro and in vivo. Here, we show that the wild-type HERV-W vaccine generated higher activation of murine antigen-presenting cells and higher specific T-cell responses than the ISD-mutated counterpart. We also found that the wild-type HERV-W vaccine was sufficient to increase the probability of survival in mice subjected to HERV-W envelope-expressing tumours compared to a control vaccine. These findings provide the foundation for developing a therapeutic cancer vaccine targeting HERV-W-positive cancers in humans. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

27 pages, 3866 KiB  
Article
Chimeric Human Papillomavirus-16 Virus-like Particles Presenting HIV-1 P18I10 Peptide: Expression, Purification, Bio-Physical Properties and Immunogenicity in BALB/c Mice
by Chun-Wei Chen, Narcís Saubi and Joan Joseph-Munné
Int. J. Mol. Sci. 2023, 24(9), 8060; https://doi.org/10.3390/ijms24098060 - 29 Apr 2023
Cited by 2 | Viewed by 1798
Abstract
Human papillomavirus (HPV) vaccines based on HPV L1 virus-like particles (VLPs) are already licensed but not accessible worldwide. About 38.0 million people were living with HIV in 2020 and there is no HIV vaccine yet. Therefore, safe, effective, and affordable vaccines against both [...] Read more.
Human papillomavirus (HPV) vaccines based on HPV L1 virus-like particles (VLPs) are already licensed but not accessible worldwide. About 38.0 million people were living with HIV in 2020 and there is no HIV vaccine yet. Therefore, safe, effective, and affordable vaccines against both viruses are an urgent need. In this study, the HIV-1 P18I10 CTL peptide from the V3 loop of HIV-1 gp120 glycoprotein was inserted into the HPV16 L1 protein to construct chimeric HPV:HIV (L1:P18I10) VLPs. Instead of the traditional baculovirus expression vector/insect cell (BEVS/IC) system, we established an alternative mammalian 293F cell-based expression system using cost-effective polyethylenimine-mediated transfection for L1:P18I10 protein production. Compared with conventional ultracentrifugation, we optimized a novel chromatographic purification method which could significantly increase L1:P18I10 VLP recovery (~56%). Chimeric L1:P18I10 VLPs purified from both methods were capable of self-assembling to integral particles and shared similar biophysical and morphological properties. After BALB/c mice immunization with 293F cell-derived and chromatography-purified L1:P18I10 VLPs, almost the same titer of anti-L1 IgG (p = 0.6409) was observed as Gardasil anti-HPV vaccine-immunized mice. Significant titers of anti-P18I10 binding antibodies (p < 0.01%) and P18I10-specific IFN-γ secreting splenocytes (p = 0.0002) were detected in L1:P18I10 VLP-immunized mice in comparison with licensed Gardasil-9 HPV vaccine. Furthermore, we demonstrated that insertion of HIV-1 P18I10 peptide into HPV16 L1 capsid protein did not affect the induction in anti-L1 antibodies. All in all, we expected that the mammalian cell expression system and chromatographic purification methods could be time-saving, cost-effective, scalable platforms to engineer bivalent VLP-based vaccines against HPV and HIV-1 Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

11 pages, 1815 KiB  
Article
Distinctive Kaposi Sarcoma-Associated Herpesvirus Serological Profile during Acute Plasmodium falciparum Malaria Episodes
by Peter O. Oluoch, Catherine S. Forconi, Cliff I. Oduor, Dominic A. Ritacco, Hoseah M. Akala, Jeffrey A. Bailey, Jonathan J. Juliano, John M. Ong’echa, Christian Münz and Ann M. Moormann
Int. J. Mol. Sci. 2023, 24(7), 6711; https://doi.org/10.3390/ijms24076711 - 04 Apr 2023
Cited by 1 | Viewed by 1291
Abstract
The seroprevalence of Kaposi sarcoma-associated herpesvirus (KSHV) and the incidence of endemic Kaposi sarcoma (KS) overlap with regions of malaria endemicity in sub-Saharan Africa. Multiple studies have shown an increased risk of KSHV seroconversion in children from high malaria compared to low malaria [...] Read more.
The seroprevalence of Kaposi sarcoma-associated herpesvirus (KSHV) and the incidence of endemic Kaposi sarcoma (KS) overlap with regions of malaria endemicity in sub-Saharan Africa. Multiple studies have shown an increased risk of KSHV seroconversion in children from high malaria compared to low malaria regions; however, the impact of acute episodes of Plasmodium falciparum (P. falciparum) malaria on KSHV’s biphasic life cycle and lytic reactivation has not been determined. Here, we examined KSHV serological profiles and viral loads in 134 children with acute malaria and 221 healthy children from high malaria regions in Kisumu, as well as 77 healthy children from low malaria regions in Nandi. We assayed KSHV, Epstein–Barr virus (EBV), and P. falciparum malaria antibody responses in these three by multiplexed Luminex assay. We confirmed that KSHV seroprevalence was significantly associated with malaria endemicity (OR = 1.95, 1.18–3.24 95% CI, p = 0.01) with 71–77% seropositivity in high-malaria (Kisumu) compared to 28% in low-malaria (Nandi) regions. Furthermore, KSHV serological profiles during acute malaria episodes were distinct from age-matched non-malaria-infected children from the same region. Paired IgG levels also varied after malaria treatment, with significantly higher anti-ORF59 at day 0 but elevated ORF38, ORF73, and K8.1 at day 3. Acute malaria episodes is characterized by perturbation of KSHV latency in seropositive children, providing further evidence that malaria endemicity contributes to the observed increase in endemic KS incidence in sub-Saharan Africa. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

11 pages, 286 KiB  
Article
Role of Low-Risk HPV PCR Monoinfection in Screening for HSIL and Anal Cancer in Men Who Have Sex with Men Living with HIV
by Carmen María García-Martínez, Inmaculada Calle-Gómez, Javier López-Hidalgo, Patricia Gómez-Ronquillo, Mohamed Omar-Mohamed Balgahata and Carmen Hidalgo-Tenorio
Int. J. Mol. Sci. 2023, 24(6), 5642; https://doi.org/10.3390/ijms24065642 - 15 Mar 2023
Viewed by 1451
Abstract
To determine the value of low-risk human papillomavirus (HPV) PCR to screen for “high-grade anal squamous intraepithelial lesion and anal cancer” (HSIL-plus), rate of patients with low-grade anal squamous intraepithelial lesion (LSIL) progressing to HSIL-plus, and progression-related factors. Prospective, longitudinal study of consecutive [...] Read more.
To determine the value of low-risk human papillomavirus (HPV) PCR to screen for “high-grade anal squamous intraepithelial lesion and anal cancer” (HSIL-plus), rate of patients with low-grade anal squamous intraepithelial lesion (LSIL) progressing to HSIL-plus, and progression-related factors. Prospective, longitudinal study of consecutive MSM-LHIV attended between May 2010 and December 2021 and followed for 43 months (IQR: 12–76). HIV-related variables were gathered at baseline, performing anal cytology for HPV detection/genotyping, thin-layer cytological study, and high-resolution anoscopy (HRA). Follow-up was annual when HRA was normal or LSIL, and post-treatment in cases of HSIL-plus, re-evaluating sexual behavior, viral-immunological status, and HPV infection of anal mucosa. The 493 participants had mean age of 36 years: CD4 nadir < 200 cells/uL in 23.1%, virological failure in 4.1%, and tetravalent HPV vaccine > 5 years earlier in 15%. HSIL-plus was ruled out in patients with monoinfection by low-risk HPV genotype and normal cytology (100% sensitivity, 91.9% specificity, PPV 2.9%, and NPV 100%). Progression from LISL to HSIL-plus occurred in 4.27% of patients within 12 months (IQR: 12–12): risk factors were acquisition of high-risk (HR: 4.15; 95% CI: 1.14–15.03) and low-risk (HR: 3.68 95% CI: 1.04–12.94) HPV genotypes, specifically genotype 6 (HR: 4.47, 95% CI: 1.34–14.91), and history of AIDS (HR: 5.81 95% CI: 1.78–18.92). Monoinfection by LR-HPV genotypes in patients with normal cytology is not associated with anal cancer or precursor lesions. Progression from LSIL to HSIL-plus, observed in <5% of patients, was related to acquisition of HR and LR HPV genotypes, especially 6, and a history of AIDS. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
16 pages, 2240 KiB  
Article
Reovirus Type 3 Dearing Variants Do Not Induce Necroptosis in RIPK3-Expressing Human Tumor Cell Lines
by Diana J. M. van den Wollenberg, Vera Kemp, Martijn J. W. E. Rabelink and Rob C. Hoeben
Int. J. Mol. Sci. 2023, 24(3), 2320; https://doi.org/10.3390/ijms24032320 - 24 Jan 2023
Viewed by 1601
Abstract
Reoviruses are used as oncolytic viruses to destroy tumor cells. The concomitant induction of anti-tumor immune responses enhances the efficacy of therapy in tumors with low amounts of immune infiltrates before treatment. The reoviruses should provoke immunogenic cell death (ICD) to stimulate a [...] Read more.
Reoviruses are used as oncolytic viruses to destroy tumor cells. The concomitant induction of anti-tumor immune responses enhances the efficacy of therapy in tumors with low amounts of immune infiltrates before treatment. The reoviruses should provoke immunogenic cell death (ICD) to stimulate a tumor cell-directed immune response. Necroptosis is considered a major form of ICD, and involves receptor-interacting protein kinase 1 (RIPK1), RIPK3 and phosphorylation of mixed-lineage kinase domain-like protein (MLKL). This leads to cell membrane disintegration and the release of damage-associated molecular patterns that can activate immune responses. Reovirus Type 3 Dearing (T3D) can induce necroptosis in mouse L929 fibroblast cells and mouse embryonic fibroblasts. Most human tumor cell lines have a defect in RIPK3 expression and consequently fail to induce necroptosis as measured by MLKL phosphorylation. We used the human colorectal adenocarcinoma HT29 cell line as a model to study necroptosis in human cells since this cell line has frequently been described in necroptosis-related studies. To stimulate MLKL phosphorylation and induce necroptosis, HT29 cells were treated with a cocktail consisting of TNFα, the SMAC mimetic BV6, and the caspase inhibitor Z-VAD-FMK. While this treatment induced necroptosis, three different reovirus T3D variants, i.e., the plasmid-based reverse genetics generated virus (T3DK), the wild-type reovirus T3D isolate R124, and the junction adhesion molecule-A-independent reovirus mutant (jin-1) failed to induce necroptosis in HT29 cells. In contrast, these viruses induced MLKL phosphorylation in murine L929 cells, albeit with varying efficiencies. Our study shows that while reoviruses efficiently induce necroptosis in L929 cells, this is not a common phenotype in human cell lines. This study emphasizes the difficulties of translating the results of ICD studies from murine cells to human cells. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

Review

Jump to: Research, Other

25 pages, 1250 KiB  
Review
The Impact of Co-Infections for Human Gammaherpesvirus Infection and Associated Pathologies
by Prishanta Chinna, Katrin Bratl, Humaira Lambarey, Melissa J. Blumenthal and Georgia Schäfer
Int. J. Mol. Sci. 2023, 24(17), 13066; https://doi.org/10.3390/ijms241713066 - 22 Aug 2023
Cited by 3 | Viewed by 1991
Abstract
The two oncogenic human gammaherpesviruses Epstein-Barr virus (EBV) and Kaposi’s sarcoma-associated herpesvirus (KSHV) cause significant disease burden, particularly in immunosuppressed individuals. Both viruses display latent and lytic phases of their life cycle with different outcomes for their associated pathologies. The high prevalence of [...] Read more.
The two oncogenic human gammaherpesviruses Epstein-Barr virus (EBV) and Kaposi’s sarcoma-associated herpesvirus (KSHV) cause significant disease burden, particularly in immunosuppressed individuals. Both viruses display latent and lytic phases of their life cycle with different outcomes for their associated pathologies. The high prevalence of infectious diseases in Sub-Saharan Africa (SSA), particularly HIV/AIDS, tuberculosis, malaria, and more recently, COVID-19, as well as their associated inflammatory responses, could potentially impact either virus’ infectious course. However, acute or lytically active EBV and/or KSHV infections often present with symptoms mimicking these predominant diseases leading to misdiagnosis or underdiagnosis of oncogenic herpesvirus-associated pathologies. EBV and/or KSHV infections are generally acquired early in life and remain latent until lytic reactivation is triggered by various stimuli. This review summarizes known associations between infectious agents prevalent in SSA and underlying EBV and/or KSHV infection. While presenting an overview of both viruses’ biphasic life cycles, this review aims to highlight the importance of co-infections in the correct identification of risk factors for and diagnoses of EBV- and/or KSHV-associated pathologies, particularly in SSA, where both oncogenic herpesviruses as well as other infectious agents are highly pervasive and can lead to substantial morbidity and mortality. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

19 pages, 1089 KiB  
Review
Mouse Models for HTLV-1 Infection and Adult T Cell Leukemia
by Shinsuke Nakajima and Kazu Okuma
Int. J. Mol. Sci. 2023, 24(14), 11737; https://doi.org/10.3390/ijms241411737 - 21 Jul 2023
Cited by 1 | Viewed by 1446
Abstract
Adult T cell leukemia (ATL) is an aggressive hematologic disease caused by human T cell leukemia virus type 1 (HTLV-1) infection. Various animal models of HTLV-1 infection/ATL have been established to elucidate the pathogenesis of ATL and develop appropriate treatments. For analyses employing [...] Read more.
Adult T cell leukemia (ATL) is an aggressive hematologic disease caused by human T cell leukemia virus type 1 (HTLV-1) infection. Various animal models of HTLV-1 infection/ATL have been established to elucidate the pathogenesis of ATL and develop appropriate treatments. For analyses employing murine models, transgenic and immunodeficient mice are used because of the low infectivity of HTLV-1 in mice. Each mouse model has different characteristics that must be considered before use for different HTLV-1 research purposes. HTLV-1 Tax and HBZ transgenic mice spontaneously develop tumors, and the roles of both Tax and HBZ in cell transformation and tumor growth have been established. Severely immunodeficient mice were able to be engrafted with ATL cell lines and have been used in preclinical studies of candidate molecules for the treatment of ATL. HTLV-1-infected humanized mice with an established human immune system are a suitable model to characterize cells in the early stages of HTLV-1 infection. This review outlines the characteristics of mouse models of HTLV-1 infection/ATL and describes progress made in elucidating the pathogenesis of ATL and developing related therapies using these mice. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

20 pages, 2961 KiB  
Review
Oncogenic Viruses and the Epigenome: How Viruses Hijack Epigenetic Mechanisms to Drive Cancer
by Signe A. MacLennan and Marco A. Marra
Int. J. Mol. Sci. 2023, 24(11), 9543; https://doi.org/10.3390/ijms24119543 - 31 May 2023
Cited by 2 | Viewed by 1915
Abstract
Globally, viral infections substantially contribute to cancer development. Oncogenic viruses are taxonomically heterogeneous and drive cancers using diverse strategies, including epigenomic dysregulation. Here, we discuss how oncogenic viruses disrupt epigenetic homeostasis to drive cancer and focus on how virally mediated dysregulation of host [...] Read more.
Globally, viral infections substantially contribute to cancer development. Oncogenic viruses are taxonomically heterogeneous and drive cancers using diverse strategies, including epigenomic dysregulation. Here, we discuss how oncogenic viruses disrupt epigenetic homeostasis to drive cancer and focus on how virally mediated dysregulation of host and viral epigenomes impacts the hallmarks of cancer. To illustrate the relationship between epigenetics and viral life cycles, we describe how epigenetic changes facilitate the human papillomavirus (HPV) life cycle and how changes to this process can spur malignancy. We also highlight the clinical impact of virally mediated epigenetic changes on cancer diagnosis, prognosis, and treatment. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

12 pages, 1293 KiB  
Review
CD30 Expression and Its Functions during the Disease Progression of Adult T-Cell Leukemia/Lymphoma
by Makoto Nakashima and Kaoru Uchimaru
Int. J. Mol. Sci. 2023, 24(10), 8731; https://doi.org/10.3390/ijms24108731 - 13 May 2023
Cited by 5 | Viewed by 2969
Abstract
CD30, a member of the tumor necrosis factor receptor superfamily, plays roles in pro-survival signal induction and cell proliferation in peripheral T-cell lymphoma (PTCL) and adult T-cell leukemia/lymphoma (ATL). Previous studies have identified the functional roles of CD30 in CD30-expressing malignant lymphomas, not [...] Read more.
CD30, a member of the tumor necrosis factor receptor superfamily, plays roles in pro-survival signal induction and cell proliferation in peripheral T-cell lymphoma (PTCL) and adult T-cell leukemia/lymphoma (ATL). Previous studies have identified the functional roles of CD30 in CD30-expressing malignant lymphomas, not only PTCL and ATL, but also Hodgkin lymphoma (HL), anaplastic large cell lymphoma (ALCL), and a portion of diffuse large B-cell lymphoma (DLBCL). CD30 expression is often observed in virus-infected cells such as human T-cell leukemia virus type 1 (HTLV-1). HTLV-1 is capable of immortalizing lymphocytes and producing malignancy. Some ATL cases caused by HTLV-1 infection overexpress CD30. However, the molecular mechanism-based relationship between CD30 expression and HTLV-1 infection or ATL progression is unclear. Recent findings have revealed super-enhancer-mediated overexpression at the CD30 locus, CD30 signaling via trogocytosis, and CD30 signaling-induced lymphomagenesis in vivo. Successful anti-CD30 antibody-drug conjugate (ADC) therapy for HL, ALCL, and PTCL supports the biological significance of CD30 in these lymphomas. In this review, we discuss the roles of CD30 overexpression and its functions during ATL progression. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

21 pages, 1108 KiB  
Review
HBV Infection and Host Interactions: The Role in Viral Persistence and Oncogenesis
by Riccardo Nevola, Domenico Beccia, Valerio Rosato, Rachele Ruocco, Davide Mastrocinque, Angela Villani, Pasquale Perillo, Simona Imbriani, Augusto Delle Femine, Livio Criscuolo, Maria Alfano, Marco La Montagna, Antonio Russo, Raffaele Marfella, Domenico Cozzolino, Ferdinando Carlo Sasso, Luca Rinaldi, Aldo Marrone, Luigi Elio Adinolfi and Ernesto Claar
Int. J. Mol. Sci. 2023, 24(8), 7651; https://doi.org/10.3390/ijms24087651 - 21 Apr 2023
Cited by 5 | Viewed by 2805
Abstract
Hepatitis B virus (HBV) is a major cause of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Despite the advent of vaccines and potent antiviral agents able to suppress viral replication, recovery from chronic HBV infection is still an extremely difficult goal to achieve. [...] Read more.
Hepatitis B virus (HBV) is a major cause of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Despite the advent of vaccines and potent antiviral agents able to suppress viral replication, recovery from chronic HBV infection is still an extremely difficult goal to achieve. Complex interactions between virus and host are responsible for HBV persistence and the risk of oncogenesis. Through multiple pathways, HBV is able to silence both innate and adaptive immunological responses and become out of control. Furthermore, the integration of the viral genome into that of the host and the production of covalently closed circular DNA (cccDNA) represent reservoirs of viral persistence and account for the difficult eradication of the infection. An adequate knowledge of the virus–host interaction mechanisms responsible for viral persistence and the risk of hepatocarcinogenesis is necessary for the development of functional cures for chronic HBV infection. The purpose of this review is, therefore, to analyze how interactions between HBV and host concur in the mechanisms of infection, persistence, and oncogenesis and what are the implications and the therapeutic perspectives that follow. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

Other

Jump to: Research, Review

15 pages, 2927 KiB  
Case Report
Small RNA Profiling in an HTLV-1-Infected Patient with Acute Adult T-Cell Leukemia-Lymphoma at Diagnosis and after Maintenance Therapy: A Case Study
by Rodrigo Pessôa, Daniela Raguer Valadão de Souza, Youko Nukui, Juliana Pereira, Lorena Abreu Fernandes, Rosa Nascimento Marcusso, Augusto César Penalva de Oliveira, Jorge Casseb, Alberto José da Silva Duarte and Sabri Saeed Sanabani
Int. J. Mol. Sci. 2023, 24(13), 10643; https://doi.org/10.3390/ijms241310643 - 26 Jun 2023
Viewed by 921
Abstract
Small RNAs (sRNAs) are epigenetic regulators of essential biological processes associated with the development and progression of leukemias, including adult T-cell leukemia/lymphoma (ATLL) caused by human T-cell lymphotropic virus type 1 (HTLV-1), an oncogenic human retrovirus originally discovered in a patient with adult [...] Read more.
Small RNAs (sRNAs) are epigenetic regulators of essential biological processes associated with the development and progression of leukemias, including adult T-cell leukemia/lymphoma (ATLL) caused by human T-cell lymphotropic virus type 1 (HTLV-1), an oncogenic human retrovirus originally discovered in a patient with adult T-cell leukemia/lymphoma. Here, we describe the sRNA profile of a 30-year-old woman with ATLL at the time of diagnosis and after maintenance therapy with the aim of correlating expression levels with response to therapy. Full article
(This article belongs to the Special Issue Host and Human Oncovirus Interaction)
Show Figures

Figure 1

Back to TopTop