[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (289)

Search Parameters:
Keywords = HTLV-1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 1483 KiB  
Brief Report
Evaluation of QuantiFERON-TB Gold for the Diagnosis of Mycobacterium tuberculosis Infection in HTLV-1-Infected Patients
by Luana Leandro Gois, Natália Barbosa Carvalho, Fred Luciano Neves Santos, Carlos Gustavo Regis-Silva, Thainá Gonçalves Tolentino Figueiredo, Bernardo Galvão-Castro, Edgar Marcelino Carvalho and Maria Fernanda Rios Grassi
Viruses 2024, 16(12), 1873; https://doi.org/10.3390/v16121873 - 30 Nov 2024
Viewed by 669
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is associated with an increased risk of tuberculosis (TB). This study aimed to evaluate the performance of the QuantiFERON-TB Gold (QFT) test for the diagnosis of Mycobacterium tuberculosis (MTB) infection in HTLV-1-infected individuals. HTLV-1-infected participants were [...] Read more.
Human T-cell leukemia virus type 1 (HTLV-1) is associated with an increased risk of tuberculosis (TB). This study aimed to evaluate the performance of the QuantiFERON-TB Gold (QFT) test for the diagnosis of Mycobacterium tuberculosis (MTB) infection in HTLV-1-infected individuals. HTLV-1-infected participants were divided into four groups: HTLV-1-infected individuals with a history of tuberculosis (HTLV/TB), individuals with positive HTLV and tuberculin skin tests (HTLV/TST+) or negative TST (HTLV/TST−), and HTLV-1-negative individuals with positive TST results (HN/TST+). We compared the diagnostic performance of the QFT assay with that of the TST as a reference and evaluated test sensitivity, specificity, accuracy, likelihood ratio, and diagnostic odds ratio. The results showed a higher frequency of positive TST results and induration diameter ≥10 mm in HTLV-1-infected individuals than in the controls. The QFT test was more frequently positive in the HTLV/TB group than in the other groups, while a combined analysis of HTLV/TB and HTLV/TST+ indicated a QFT sensitivity of 57.5%. No significant differences were found in the other diagnostic performance measures, as QFT test results were in agreement with TST results, particularly in TST-negative individuals. Given the low sensitivity of QFT for LTBI in individuals infected with HTLV-1, the TST may be preferable in regions where both infections are endemic. Full article
(This article belongs to the Special Issue Human T-Cell Leukemia Virus (HTLV) Infection and Treatment)
Show Figures

Figure 1

Figure 1
<p>Flowchart illustrating study design in conformity with the Standards for Reporting of Diagnostic Accuracy Studies (STARD).</p>
Full article ">Figure 2
<p>Diagnostic parameters used to evaluate the performance of the QuantiFERON-TB (QFT) assay in detecting TST positivity and previous tuberculosis infection (TB) among individuals with HTLV-1. HN (HTLV-1-negative); HTLV (HTLV-1-infected individuals); TB (tuberculosis); TST (tuberculin skin test).</p>
Full article ">
19 pages, 4881 KiB  
Article
A Novel Tax-Responsive Reporter T-Cell Line to Analyze Infection of HTLV-1
by Stefanie Heym, Pauline Krebs, Kristin Ott, Norbert Donhauser, Laura M. Kemeter, Florian Simon, Sebastian Millen and Andrea K. Thoma-Kress
Pathogens 2024, 13(11), 1015; https://doi.org/10.3390/pathogens13111015 - 19 Nov 2024
Viewed by 663
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) infects CD4+ T-cells through close cell–cell contacts. The viral Tax-1 (Tax) protein regulates transcription by transactivating the HTLV-1 U3R promoter in the 5′ long terminal repeat of the integrated provirus. Here, we generated a clonal [...] Read more.
Human T-cell leukemia virus type 1 (HTLV-1) infects CD4+ T-cells through close cell–cell contacts. The viral Tax-1 (Tax) protein regulates transcription by transactivating the HTLV-1 U3R promoter in the 5′ long terminal repeat of the integrated provirus. Here, we generated a clonal Tax-responsive T-cell line to track HTLV-1 infection at the single-cell level using flow cytometry, bypassing intracellular viral protein staining. Jurkat T-cells stably transduced with the SMPU vector carrying green fluorescent protein (GFP) under control of 18 × 21 bp Tax-responsive element repeats of the U3R were evaluated. Among 40 clones analyzed for Tax responsiveness, the top two were characterized. Upon overexpression of Tax, over 40% of the cells showed GFP positivity, and approximately 90% of the Tax-positive cells were GFP-positive, indicating efficient reporter activity. However, with CREB-deficient Tax mutant M47, both total GFP-positive cell counts and those within the Tax-positive group significantly decreased. Co-culture with chronically HTLV-1-infected MT-2 or C91-PL cells led to an average of 0.9% or 2.4% GFP-positive cells, respectively, confirming the suitability to monitor HTLV-1 transmission and that HTLV-1 infection is very low. Thus, the novel Tax-responsive reporter T-cell line is a suitable tool to monitor infection of HTLV-1 on the single-cell level. Full article
(This article belongs to the Special Issue New Directions in HTLV-1 Research)
Show Figures

Figure 1

Figure 1
<p>Generation of a Tax-responsive Jurkat–SMPU reporter cell line. (<b>A</b>,<b>B</b>) Transient expression of the SMPU-18 × 21-eGFP reporter. First, (<b>A</b>) 293T or (<b>B</b>) Jurkat T-cells were transiently transfected with either the pSG5 empty vector, the Tax expression vector pSG5-Tax, the SMPU-18 × 21-eGFP reporter vector, or co-transfected with pSG5-Tax and SMPU-18 × 21-eGFP. Flow cytometry (<b>left</b> side) and Western Blot analysis (<b>right</b> side) were performed at 48 h post-transfection. For flow cytometry, forward and side scatter (SSC) were used to gate on living cells. The resulting gates are illustrated, and GFP is plotted against SSC. Western Blots of Tax and GAPDH are depicted. (<b>C</b>) Schematic overview on cell sorting of Jurkat T-cells after transduction. Sorting resulted in 40 individual clones (22 were initially eGFP<sup>−</sup>, 18 had background eGFP signal (eGFP<sup>+</sup>) during sorting). Clones were tested for their Tax responsiveness by transient transfection with pSG5-Tax, which led to the identification of 21 Tax-responsive clones. Created with BioRender.com.</p>
Full article ">Figure 2
<p>Tax activates GFP expression in Jurkat–SMPU reporter T-cell clones at higher levels than the CREB-deficient Tax mutant M47 despite comparable expression levels. Jurkat T-cells, Jurkat–SMPU clones 21, 29, 31, and 34, and a mixed clone of the respective clones were transfected with either the wildtype expression plasmid pSG5-Tax, with the expression plasmid of the CREB-deficient Tax mutant M47 (pSG5-M47), or with the pSG5 empty vector. (<b>A</b>) eGFP expression was analyzed at 48 h post-transfection using a Nikon Eclipse TS2-FL inverted fluorescence microscope and TCapture software. (<b>B</b>) Representative Western Blots at 48 h post-transfection depicting Tax and GAPDH.</p>
Full article ">Figure 3
<p>Jurkat–SMPU reporter T-cell clones are Tax-responsive and show high efficiency in activating eGFP expression. Jurkat T-cells, Jurkat–SMPU reporter T-cell clones 21, 29, and the mixed clone of 21, 29, 31, and 34 were transfected with either the wildtype expression plasmid pSG5-Tax, with the expression plasmid of the CREB-deficient Tax mutant M47 (pSG5-M47), or with the pSG5 empty vector. After 48 h, cells were fixed, intracellularly stained with primary anti-Tax and secondary anti-mouse IgG AF647-coupled antibodies, and analyzed via flow cytometry. (<b>A</b>) Gating strategy depicting representative dot plots of clone 29 transfected with pSG5-Tax. Upper row: Cells were plotted as size (FSC-A; forward scatter area) against size (FSC-H; forward scatter height) to exclude doublets. Then, living cells were determined by plotting size (FSC-A) against granularity (SSC-A; side scatter area). Within the live cell gate, eGFP- and Tax-positive cells were assessed. Lower row, left: After gating for Tax-positive cells by plotting Tax against granularity (SSC), the eGFP-positive cells within the Tax-positive population were assessed. Lower row, right: After gating for eGFP-positive cells by plotting GFP against SSC, the Tax-positive cells within the eGFP-positive population were assessed. (<b>B</b>) eGFP-positive, (<b>C</b>) Tax-positive, and (<b>D</b>) both eGFP- and Tax-positive cells, as well as (<b>E</b>) eGFP-positive cells within the Tax-positive population and (<b>F</b>) vice versa were assessed by flow cytometry analysis. Mean values of five independent experiments ± SE are depicted. Data were analyzed for normal distribution using the Shapiro–Wilk Test followed by (<b>B</b>,<b>D</b>) one-way ANOVA and Tukey’s test (more than two groups) or (<b>E</b>) an unpaired Student’s <span class="html-italic">t</span>-test (two groups) (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 4
<p>Determination of background reporter activity after stimulation of Jurkat–SMPU reporter T-cell clones using different stimuli. (<b>A</b>) Jurkat–SMPU reporter clone 21, (<b>B</b>) clone 29, or (<b>C</b>) the mixed clone of clones 21, 29, 31 and 34 were stimulated with PMA (20 nM) and Ionomycin (1 µM), PHA-P (5 µg/mL) and IL-2 (25 U/mL) or respective solvent controls (DMSO, PBS). Cells were analyzed after d1, d2, d5, and d7 of stimulation. GFP-positive cells were assessed by FACS analysis. Mean values of three independent experiments ± SD are depicted.</p>
Full article ">Figure 5
<p>Jurkat–SMPU reporter T-cell cones 21 and 29 are suitable tools to study infection using C91-PL and MT-2 cells as donor cells. (<b>A</b>) Schematic overview of experimental setup. Jurkat–SMPU reporter T-cell clones 21 and 29 were stimulated with 25 U/mL IL-2 and 2 µg/mL PHA-P. After 48 h, stimulated cells were stained with CellTrace<sup>TM</sup> Violet (CTV) and used as acceptor cells in co-culture experiments. Before co-culture, donor cells were irradiated (77 Gray (Gy)). On days 0 and 3 of co-culture, cells were additionally stimulated with 25 U/mL IL-2. Flow cytometry analysis of eGFP reporter activity in CTV<sup>+</sup> cells was performed on days 0, 3, and 6 of co-culture. As negative controls, donor cells were replaced with either stimulated or unstimulated Jurkat T-cells (not depicted). Created with BioRender.com. (<b>B</b>) Gating strategy illustrating representative dot plots of clone 29 co-cultured with irradiated C91-PL donor cells for 3–6 days. (1) Cells were first plotted size (FSC; forward scatter area) against granularity (SSC; side scatter) to determine living cells of co-culture. (2) Within the co-culture gate, CTV-positive cells were assessed. (3,4) Infection of CTV<sup>+</sup> Jurkat–SMPU clones was determined by plotting the CTV<sup>+</sup> gate against granularity (SSC) on days 0, 3, and 6 of co-culture (highlighted in pink box). (<b>C</b>) Flow cytometry analysis depicting the frequency of eGFP-positive cells within the CTV<sup>+</sup> population (<b>left</b> column) or mean fluorescent intensity (MFI) of eGFP<sup>+</sup> CTV<sup>+</sup> cells (<b>right</b> column). Mean values of three independent experiments ± SE are depicted.</p>
Full article ">
21 pages, 3331 KiB  
Article
Characterization of HTLV-1 Infectious Molecular Clone Isolated from Patient with HAM/TSP and Immortalization of Human Primary T-Cell Lines
by Marcia Bellon, Pooja Jain and Christophe Nicot
Viruses 2024, 16(11), 1755; https://doi.org/10.3390/v16111755 - 9 Nov 2024
Viewed by 802
Abstract
Human T-cell leukemia virus (HTLV-1) is the etiological agent of lymphoproliferative diseases such as adult T-cell leukemia and T-cell lymphoma (ATL) and a neurodegenerative disease known as HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). While several molecular clones of HTLV-1 have been published, all were [...] Read more.
Human T-cell leukemia virus (HTLV-1) is the etiological agent of lymphoproliferative diseases such as adult T-cell leukemia and T-cell lymphoma (ATL) and a neurodegenerative disease known as HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). While several molecular clones of HTLV-1 have been published, all were isolated from samples derived from patients with adult T-cell leukemia. Here, we report the characterization of an HTLV-1 infectious molecular clone isolated from a sample of a patient with HAM/TSP disease. Genetic comparative analyses of the HAM/TSP molecular clone (pBST) revealed unique genetic alterations and specific viral mRNA expression patterns. Interestingly, our clone also harbors characteristics previously published to favor the development of HAM/TSP disease. The molecular clone is capable of infection and immortalization of human primary T cells in vitro. Our studies further demonstrate that the HTLV-1 virus produced from primary T cells transfected with pBST or ACH molecular clones cannot sustain long-term expansion, and cells cease to proliferate after 3–4 months in culture. In contrast, long-term proliferation and immortalization were achieved if the virus was transmitted from dendritic cells to primary T cells, and secondary infection of 729B cells in vitro was demonstrated. In both primary T cells and 729B cells, pBST and ACH were latent, and only hbz viral RNA was detected. This study suggests that HTLV-1 transmission from DC to T cells favors the immortalization of latently infected cells. Full article
(This article belongs to the Special Issue Chronic Infection by Oncogenic Viruses)
Show Figures

Figure 1

Figure 1
<p>Cloning pBST into pBR327. (<b>A</b>) Agarose gel of p.4.39 DNA digested with 1-Sac1+BstEII. 2- BstEII. 3- Sac1. 4- Lambda HindIII DNA marker. Schematic representation of p4.39 with restriction sites for Sac1 (S) or BstEII (B) and pBR327 digested with EcoR1 and Sal1 for cloning. (<b>B</b>–<b>D</b>) Alignment between pBST and MT-2 sequences of different regions of the viral LTR. (<b>E</b>) Alignment between pBST and MT-2 sequence of RexRE REM3 loop. (<b>F</b>) The enhancer region conserved sites for transcription factors SRF and ELK-1 in HTLV-1 ACH (<b>top</b>) and pBST sequence (<b>bottom</b>).</p>
Full article ">Figure 2
<p>Schematic representation of pBST mutations present in viral proteins. (<b>A</b>) Phylogenetic tree analysis, generated with the maximum likelihood (PhyML) method on a 710-nt-long fragment of the LTR. The numbers at the nodes correspond to the bootstrap value, obtained after 1000 repeats. The branch lengths are drawn to scale. HTLV-1c strains were used as outgroup. (<b>B</b>) Phylogenetic tree generated by the maximum likelihood method (PhyML) on 6366 nt corresponding to the concatenation of gag-pol-env-tax ORFs. The numbers at the nodes correspond to the bootstrap value, obtained after 1000 repeats. The branch lengths are drawn to scale. HTLV-1c strains were used as outgroup. (<b>C</b>) Amino acid sequence of structural and enzymatic proteins GAG, PRO, POL, and ENV TAX, HBZ, p12, p30, and p13 from pBST and HTLV-1A prototype ATK. Mutation positions are indicated. (<b>D</b>) Amino acid sequence alignment compared to full-length genome sequences from Japanese HAM/TSP (n = 12) or Brazilian HAM/TSP (n = 10) (Supplemental S4–S12). See material and methods. Red triangle and (*) indicate stop codon.</p>
Full article ">Figure 3
<p>Expression of viral mRNAs from pBST. (<b>A</b>) Schematic representation of HTLV-1 mRNAs with positions of splice donors and splice acceptor sites noted. (<b>B</b>) pBST was transfected into 293T cells, and total RNAs were extracted after 48 h. “(-)” represents control, pCDNA 3.1 transfected 293T cells, and “PBST” represents pBST transfected 293T cells. Specific primers (<a href="#viruses-16-01755-t001" class="html-table">Table 1</a>) were used, and RT-PCR products were resolved onto agarose gels. p13 PCR products are approximately 130 bp. HTLV-1 transformed cells LAF, MT4, and HUT102 were used as controls. (<b>C</b>) Schematic of the pBST LTR cloned into the luciferase vector. Luciferase assays representing fold change activation of pBST-LTR-Luciferase activated by Tax produced in the context of the molecular clone. (<b>D</b>) The pBST envelope gene was cloned into pCDNA 3.1 expression vector and transfected into high-density HeLa cells. Syncytia were visualized after 48 h by staining with Crystal Violet.</p>
Full article ">Figure 4
<p>pBST produces infectious virus particles. (<b>A</b>) Electron microscopy images of 293T cells transfected with ACH or pBST. Digital images were acquired with an AMT digital camera, with magnification scale bars indicated in the figures. White triangles indicate virus particles, with size in nanometers (nm). (<b>B</b>) 293T cells were transfected with increasing amounts of pBST molecular clones. After 48 h, supernatant was cleared by centrifugation for 5 min at 10,000 rpm, and supernatant was tested for GAG p19 by ELISA. (<b>C</b>) 729B cells were transfected with pBST by Amaxa, and after 48 h, cells were co-cultivated with BHK1E6 HTLV-1LTR-LacZ reporter cells for 48 h. Cells were washed, fixed, and stained with X-Gal to reveal HTLV-1-infected beta-galactosidase-positive cells.</p>
Full article ">Figure 5
<p>Immortalization of primary human T cell lines and secondary transmission to 729B cells. (<b>A</b>) Images of cultures from PBMCs at 3 weeks of culture and pBST immortalized T cell lines. (<b>B</b>) FACS analyses of cell surface markers for CD4, CD8, and CD25 expression on activated PBMCs or pBST immortalized cells. Cells were blocked for 30 min in BSA buffer and incubated for 2 h with the appropriate conjugated antibody. Cells were washed and fixed in 1% PFA overnight before analyses. CD4-APC (cat#551980) BD Pharmingen; IgG1-k-Isotype control-APC (cat#550854) BD Pharmingen. CD8-PE (cat#555367) BD Pharmingen; IgG1-k-Isotype control-PE (cat#555749) BD Pharmingen. Results were acquired on instrument BC-Accuri C6 Plus Flow Cytometer (BD Biosciences). (<b>C</b>) PCR analyses of genomic DNA from PBMC controls or PBMCs immortalized with pBST. HTLV-1 LAF cell line was used as a control. Chromatograms show specific genetic variations (black arrow) present in the pBST immortalized cell lines. The ACH sequence was used as a control. (<b>D</b>) RT-PCR analysis of <span class="html-italic">hbz</span> expression from 729B cell controls or 729B cells chronically infected with pBST. The HTLV-1 LAF cell line was used as a control.</p>
Full article ">
12 pages, 1553 KiB  
Article
Radiological Changes in the Spinal Cord and Brain of Patients with HTLV-1-Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP)
by Emily H. Stack, Serhat V. Okar, Tianxia Wu, Mallory Stack, Yair Mina, María Gaitán, Shila Azodi, Will Frazier, Joan Ohayon, Irene C. M. Cortese, Daniel S. Reich, Govind Nair and Steven Jacobson
Pathogens 2024, 13(11), 920; https://doi.org/10.3390/pathogens13110920 - 22 Oct 2024
Viewed by 661
Abstract
HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a chronic, progressive neurological disorder and shares many radiological and clinical features with other more prevalent myelopathies. Here, we quantified spinal cord and brain volumes in adults with HAM/TSP in comparison with healthy volunteers (HVs) and individuals [...] Read more.
HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a chronic, progressive neurological disorder and shares many radiological and clinical features with other more prevalent myelopathies. Here, we quantified spinal cord and brain volumes in adults with HAM/TSP in comparison with healthy volunteers (HVs) and individuals diagnosed with relapsing–remitting or progressive multiple sclerosis (RRMS or P-MS). Clinical disability and MRI were assessed in 24 HVs, 43 HAM/TSP subjects, and 46 MS subjects. Spinal cord cross-sectional area (SCCSA) and brain tissue volumes were measured and compared. HAM/TSP subjects had significantly lower SCCSA corresponding to cervical levels 2 and 3 (C2–3) (54.0 ± 8 mm2), cervical levels 4 and 5 (C4–5) (57.8 ± 8 mm2), and thoracic levels 4 to 9 (T4–9) (22.7 ± 4 mm2) and significantly elevated brain white matter hyperintensity (WMH) fraction (0.004 ± 0.008) compared to the HVs (C2–3: 69.4 ± 8 mm2, C4–5: 75.1 ± 9 mm2, T4–9: 34.1 ± 4 mm2; all p < 0.0001; and WMH: 0.0005 ± 0.0007; p < 0.001). In the HAM/TSP subjects, SCCSA at all levels but not WMH showed a significant correlation with clinical disability scores. WMH in HAM/TSP subjects, therefore, may not be related to clinical disability. SCCSA in our limited RRMS cohort was higher than the HAM/TSP cohort (C2–3: 67.6 ± 8 mm2, C4–5: 72.7 ± 9 mm2, T4–9: 33.4 ± 5 mm2; all p < 0.0001) and WMH was lower than in P-MS subjects (p = 0.0067). Principal component analysis suggested that SCCSA and WMH may be used to differentiate HAM/TSP from MS. Understanding these differences msay help establish early diagnostic criteria for HAM/TSP patients. Full article
(This article belongs to the Special Issue New Directions in HTLV-1 Research)
Show Figures

Figure 1

Figure 1
<p>Global spinal cord atrophy. (<b>A</b>) Representative mid-sagittal T1-weighted MR images of the cervical (upper row) and thoracolumbar (lower low) from healthy volunteers (HVs), HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), relapsing–remitting multiple sclerosis (RRMS), and progressive multiple sclerosis (P-MS). Group-averaged spinal cord cross-sectional areas (SCCSA) from (<b>B</b>) C2–3, (<b>C</b>) C4–5, and (<b>D</b>) T4–9 showing significantly lower values in the HAM/TSP subjects at all regions of the cord. ** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Brain volumes. (<b>A</b>) FLAIR image (top row) and brain segmentation results overlaid on FLAIR images showing segmented (bottom row) grey matter (brown), white matter (beige), cerebrospinal fluid (CSF, green), and white matter hyperintensities or lesions (teal) from a representative participant in each diagnosis group. Group-averaged analysis of (<b>B</b>) grey matter, (<b>C</b>) white matter, (<b>D</b>) CSF, and (<b>E</b>) lesion or white matter hyperintensity volumes expressed as a fraction of total intracranial volume and adjusted for age showing differences only in the WMH volumes. ** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Correlation of radiological and clinical scores. (<b>A</b>) Plots showing a statistically significant correlation (partial Pearson’s) between Scripps neurologic rating scale (SNRS) and spinal cord cross-sectional area (SCCSA) at all cord levels (in columns) for HAM/TSP (top row) and RRMS (bottom row) subjects with disability increasing with reducing SCCSA. (<b>B</b>) SCCSA showed a significant correlation only in the C4–5 region in the P-MS group (left). Brain white matter hyperintensity (or lesion) volumes were significantly correlated with a clinical disability only in the relapsing–remitting multiple sclerosis group (correlation with EDSS shown on the right).</p>
Full article ">Figure 4
<p>Principal component analysis. Principal components 1 (dominated by SCCSA) and 3 (dominated by median WMH volume from individuals) derived from brain and spine radiological variables show the ability to separate the diagnosis groups.</p>
Full article ">
41 pages, 2642 KiB  
Review
Current State of Therapeutics for HTLV-1
by Tiana T. Wang, Ashley Hirons, Marcel Doerflinger, Kevin V. Morris, Scott Ledger, Damian F. J. Purcell, Anthony D. Kelleher and Chantelle L. Ahlenstiel
Viruses 2024, 16(10), 1616; https://doi.org/10.3390/v16101616 - 15 Oct 2024
Cited by 1 | Viewed by 1309
Abstract
Human T cell leukaemia virus type-1 (HTLV-1) is an oncogenic retrovirus that causes lifelong infection in ~5–10 million individuals globally. It is endemic to certain First Nations populations of Northern and Central Australia, Japan, South and Central America, Africa, and the Caribbean region. [...] Read more.
Human T cell leukaemia virus type-1 (HTLV-1) is an oncogenic retrovirus that causes lifelong infection in ~5–10 million individuals globally. It is endemic to certain First Nations populations of Northern and Central Australia, Japan, South and Central America, Africa, and the Caribbean region. HTLV-1 preferentially infects CD4+ T cells and remains in a state of reduced transcription, often being asymptomatic in the beginning of infection, with symptoms developing later in life. HTLV-1 infection is implicated in the development of adult T cell leukaemia/lymphoma (ATL) and HTLV-1-associated myelopathies (HAM), amongst other immune-related disorders. With no preventive or curative interventions, infected individuals have limited treatment options, most of which manage symptoms. The clinical burden and lack of treatment options directs the need for alternative treatment strategies for HTLV-1 infection. Recent advances have been made in the development of RNA-based antiviral therapeutics for Human Immunodeficiency Virus Type-1 (HIV-1), an analogous retrovirus that shares modes of transmission with HTLV-1. This review highlights past and ongoing efforts in the development of HTLV-1 therapeutics and vaccines, with a focus on the potential for gene therapy as a new treatment modality in light of its successes in HIV-1, as well as animal models that may help the advancement of novel antiviral and anticancer interventions. Full article
(This article belongs to the Special Issue HIV and HTLV Infections and Coinfections)
Show Figures

Figure 1

Figure 1
<p>Worldwide distribution of HTLV-1 infection. The map specifies the prevalence by region. Created with mapchart.net (2022).</p>
Full article ">Figure 2
<p>The genomic structure of HTLV-1. The 3′ region is described as the pX region. Tax, rex, and hbz are spliced. Created in BioRender.</p>
Full article ">Figure 3
<p>The HTLV-1 life cycle in a cell. The virus fuses into the host cell, CD4<sup>+</sup>/CD8<sup>+</sup> T cells, through interaction with cell surface receptors. The viral core is transported into the cytoplasm where it is reverse transcribed into a double-stranded DNA. This is then transported into the nucleus and integrated into the host genome. The viral proteins are transcribed and translated by the host cell machinery and become infectious after undergoing viral budding. Created in BioRender.</p>
Full article ">Figure 4
<p>Disease progression of HTLV-1 and the rates of infection. HTLV-1 mostly presents in infected patients as a lifelong subtle immune dysfunction without a defined associated disease, with a broadly estimated 10–30% of populations developing symptomatic HTLV-1-associated disease. There is, however, a broad spectrum of diseases that are reported with infection other than HAM and ATL. Understanding the determinants of if, when, and what disease afflicts infected individuals needs to be further addressed. Created in BioRender.</p>
Full article ">Figure 5
<p>Mechanisms of siRNA-induced gene silencing. Both RNAi pathways can exert genetic repression by viral and non-viral mechanisms of delivery. PTGS mostly occurs in the cytoplasm as the RISC machinery initiates specific mRNA cleavage. TGS is localised to the nucleus and is mediated by the RITS complex, acting through repressive epigenetic modifications. Ago1, Argonaute 1; Ago2, Argonaute 2; shRNA, short hairpin RNA; RISC, RNA-induced silencing complex; RITS, RNA-induced transcriptional silencing complex; TRBP, transactivating response (TAR) RNA-binding protein; TNRC6, trinucleotide repeat containing 6 protein faily. Created in BioRender.</p>
Full article ">Figure 6
<p>Routes of administration for nanoparticle delivery of RNAi therapeutics. (<b>A</b>) Chemical modifications, siRNA conjugations, and nanoparticle formulations can be used to improve the outcomes of delivery. Nanoparticle formulation helps to facilitate cell entry and endosomal escape to yield enhanced epigenetic silencing. (<b>B</b>) The different routes of administration for nanoparticle formulation. Created in BioRender.</p>
Full article ">
15 pages, 2244 KiB  
Article
Diagnostic Value of Anti-HTLV-1-Antibody Quantification in Cerebrospinal Fluid for HTLV-1-Associated Myelopathy
by Tomoo Sato, Naoko Yagishita, Natsumi Araya, Makoto Nakashima, Erika Horibe, Katsunori Takahashi, Yasuo Kunitomo, Yukino Nawa, Isao Hamaguchi and Yoshihisa Yamano
Viruses 2024, 16(10), 1581; https://doi.org/10.3390/v16101581 - 8 Oct 2024
Viewed by 873
Abstract
The diagnostic accuracy of cerebrospinal fluid (CSF) anti-human T-cell leukemia virus type I (HTLV-1) antibody testing for HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM) remains unclear. Therefore, we measured the anti-HTLV-1 antibody levels in CSF using various test kits, evaluated the stability of CSF antibodies, [...] Read more.
The diagnostic accuracy of cerebrospinal fluid (CSF) anti-human T-cell leukemia virus type I (HTLV-1) antibody testing for HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM) remains unclear. Therefore, we measured the anti-HTLV-1 antibody levels in CSF using various test kits, evaluated the stability of CSF antibodies, and performed a correlation analysis using the particle agglutination (PA) method, as well as a receiver operating characteristic (ROC) analysis between patients with HAM and carriers. The CSF anti-HTLV-1 antibody levels were influenced by freeze–thaw cycles but remained stable when the CSF was refrigerated at 4 °C for up to 48 h. Measurements from 92 patients (69 patients with HAM and 23 carriers) demonstrated a strong correlation (r > 0.9) with the PA method across all six quantifiable test kits. All six test kits, along with CSF neopterin and CXCL10, exhibited areas under the ROC curve greater than 0.9, indicating a high diagnostic performance for HAM. Among these, five test kits, Lumipulse and Lumipulse Presto HTLV-I/II, HISCL-UD (a kit under development), HTLV-Abbott, and Elecsys HTLV-I/II, established a cutoff with 100% sensitivity and maximum specificity, achieving a sensitivity of 100% and a specificity ranging from 43.5% to 56.5%. This cutoff value, in combination with clinical findings, will aid in the accurate diagnosis of HAM. Full article
(This article belongs to the Special Issue Chronic Infection by Oncogenic Viruses)
Show Figures

Figure 1

Figure 1
<p>Effect of freeze–thaw cycles on the measurement of anti-HTLV-1 antibody quantitative data in CSF. Six cases of CSF—two low, two medium, and two high titers—were employed in the assay for each test kit (see the <a href="#sec2dot5-viruses-16-01581" class="html-sec">Section 2.5</a> for details). The CSF underwent one, two, or three freeze–thawing cycles, and the anti-HTLV-1 antibody titers were assessed. Measurements for each time point are displayed as the mean ± SD. A one-way repeated-measures ANOVA was used to compare the corresponding data among the three conditions. <span class="html-italic">p</span> ≤ 0.05 was considered statistically significant. COI, cutoff index; S/CO, signal-to-cutoff ratio. Information on each test kit is shown in <a href="#viruses-16-01581-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 2
<p>Effect of storage time at 4 °C on the measurement of anti-HTLV-1 antibody quantitative data in CSF. Six cases of CSF—two low, two medium, and two high titers—were employed in the assay for each test kit (see the <a href="#sec2dot5-viruses-16-01581" class="html-sec">Section 2.5</a> for details). The CSF was stored at 4 °C for 0 h, 24 h, and 48 h, and the anti-HTLV-1 antibody titers were assessed. Measurements for each time point are displayed as the mean ± SD. A one-way repeated-measures ANOVA was used to compare the corresponding data among the three conditions. <span class="html-italic">p</span> ≤ 0.05 was considered statistically significant. COI, cutoff index; S/CO, signal-to-cutoff ratio. Information on each test kit is shown in <a href="#viruses-16-01581-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 3
<p>Distribution of CSF anti-HTLV-1 antibody titers in patients with HAM. The vertical axis represents the number of patients, and the horizontal axis represents the antibody titer measured using the PA method. Of the 322 patients with HAM, 248 were not on steroid therapy (black), and 74 were on steroid therapy (gray).</p>
Full article ">Figure 4
<p>ROC analysis to assess the diagnostic performance of the five markers in discriminating patients with HAM from HTLV-1 carriers. We collected the past data of five known markers, the PVL in PBMCs, the PVL in CSF cells, the ratio of these PVLs, CSF CXCL10, and CSF neopterin, in 92 individuals (69 patients with HAM and 23 HTLV-1 carriers). PVL, HTLV-1 proviral load; PBMCs, peripheral blood mononuclear cells; AUC, area under the curve.</p>
Full article ">Figure 5
<p>ROC analysis of CSF anti-HTLV-1 antibody levels. CSF anti-HTLV-1 antibody levels in 92 individuals (69 patients with HAM and 23 HTLV-1 carriers) were measured using seven different quantifiable anti-HTLV-1 antibody test kits, and an ROC analysis was performed for each. Information on each test kit is shown in <a href="#viruses-16-01581-t001" class="html-table">Table 1</a>. AUC, area under the curve.</p>
Full article ">Figure 6
<p>Correlations with the PA method. Using the results of anti-HTLV-1 antibody titers from 92 CSF samples (69 patients with HAM [red] and 23 carriers [blue]), we examined the correlations between the PA method and six other test kits. Antibody titers for the PA method are indicated as Log<sub>2</sub> values, and antibody levels (COI and S/CO) for all other test kits are demonstrated as Log<sub>10</sub> values. The linear equation, Spearman’s rank correlation coefficient (rs), 95% confidence interval, and <span class="html-italic">p</span>-value are shown in the figure. COI, cutoff index; S/CO, signal-to-cutoff ratio. Information on each test kit is shown in <a href="#viruses-16-01581-t001" class="html-table">Table 1</a>.</p>
Full article ">
7 pages, 238 KiB  
Article
Cognitive Assessment in HTLV-1 Patients Followed Up at a Reference Center in Salvador, Brazil
by Luísa Bordallo, Iris Montaño-Castellón, Liliane Lins-Kusterer and Carlos Brites
Viruses 2024, 16(10), 1569; https://doi.org/10.3390/v16101569 - 5 Oct 2024
Viewed by 934
Abstract
Introduction: Human T-cell lymphotropic virus type 1 (HTLV-1) is endemic to Brazil, and there is still no specific treatment for these patients. The literature shows that few studies have described the cognitive impairment associated with an HTLV-1 infection, with none of them examining [...] Read more.
Introduction: Human T-cell lymphotropic virus type 1 (HTLV-1) is endemic to Brazil, and there is still no specific treatment for these patients. The literature shows that few studies have described the cognitive impairment associated with an HTLV-1 infection, with none of them examining the population of Salvador, where there are approximately forty thousand people infected with the virus. Objectives: To determine the prevalence of cognitive impairment among individuals with HTLV-1. In addition, investigate whether sociodemographic aspects, time since the diagnosis of infection, and the diagnosis of HTLV-Associated Myelopatia/Tropical Spastic Paraparesis (HAM/TSP) or depression are associated with cognitive impairment in this population. Methods: This was an observational, cross-sectional study that consisted of consecutively approaching 100 HTLV-1 patients during outpatient care at a referral center followed by the administration of three questionnaires— the Mini Mental State Examination (MMSE), the Montreal Cognitive Assessment (MoCA), and Beck’s Depression Inventory. Results: The prevalence of cognitive impairment found was 71% using the MMSE and 82% using the MoCA. There was a statistically significant association between the cognitive dysfunction and the variables of age and education according to the MoCA analysis but not the MMSE data. Diagnosis of HAM/TSP was correlated with cognitive impairment using the MMSE but not the MoCA. The prevalence of depression was 20%, and there was no association between cognitive impairment and depressive symptoms in these patients. Conclusions: The findings of this study demonstrate a correlation between cognitive dysfunction and HTVL-1 infection, with a more evident involvement of executive functions and memory. Larger studies are needed to clarify the association between cognitive dysfunction, age, education, and the diagnosis of HAM/TSP. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
25 pages, 5680 KiB  
Review
The Assembly of HTLV-1—How Does It Differ from HIV-1?
by Dominik Herrmann, Shuyu Meng, Huixin Yang, Louis M. Mansky and Jamil S. Saad
Viruses 2024, 16(10), 1528; https://doi.org/10.3390/v16101528 - 27 Sep 2024
Viewed by 1927
Abstract
Retroviral assembly is a highly coordinated step in the replication cycle. The process is initiated when the newly synthesized Gag and Gag-Pol polyproteins are directed to the inner leaflet of the plasma membrane (PM), where they facilitate the budding and release of immature [...] Read more.
Retroviral assembly is a highly coordinated step in the replication cycle. The process is initiated when the newly synthesized Gag and Gag-Pol polyproteins are directed to the inner leaflet of the plasma membrane (PM), where they facilitate the budding and release of immature viral particles. Extensive research over the years has provided crucial insights into the molecular determinants of this assembly step. It is established that Gag targeting and binding to the PM is mediated by interactions of the matrix (MA) domain and acidic phospholipids such as phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). This binding event, along with binding to viral RNA, initiates oligomerization of Gag on the PM, a process mediated by the capsid (CA) domain. Much of the previous studies have focused on human immunodeficiency virus type 1 (HIV-1). Although the general steps of retroviral replication are consistent across different retroviruses, comparative studies revealed notable differences in the structure and function of viral components. In this review, we present recent findings on the assembly mechanisms of Human T-cell leukemia virus type 1 and highlight key differences from HIV-1, focusing particularly on the molecular determinants of Gag–PM interactions and CA assembly. Full article
Show Figures

Figure 1

Figure 1
<p>HTLV-1 and HTLV-1 replication cycles. (<b>A</b>) Mature HTLV-1 virion attaches to the host cell receptor complex containing NRP-1, GLUT-1, and HSPGs. The mechanisms of reverse transcription and uncoating have long been thought to occur in the cytoplasm but recent advances on the mechanisms of HIV-1 reverse transcription and uncoating (below) raised similar questions about other retroviruses, including HTLV-1. Subsequent nuclear import and integration into the host genome yields the provirus. Transcription and translation produce Gag, Gag-Pol, Env, accessory proteins, and viral gRNA. Gag is trafficked to the PM for assembly via the MTOC, while Env is post-translationally processed and trafficked to the cell surface through the ER and Golgi apparatus. Virus assembly and maturation yield a new, infectious virus. (<b>B</b>) Mature HIV-1 virion attaches to the host CD4 receptor and co-receptors (CCR5 or CXCR4). The virus core is then transported to the nucleus via microtubules, a process that appears to be accompanied by reverse transcription. Recent studies indicated that CA core uncoating occurs in the nucleus near the integration sites. Transcription and translation produce Gag, Gag-Pol, Env, accessory proteins, and viral gRNA. Gag is then trafficked to the PM for assembly, while Env is post-translationally processed and trafficked to the cell surface through the ER and Golgi apparatus. Virus assembly and maturation yield a new, infectious virus.</p>
Full article ">Figure 2
<p>HTLV-1 genome and RNA transcripts. Genome encodes for Gag, Pro, Pol, Env, Tax, Rex, and pX genes. pX region contains genes of Rex, Tax, p30, p12, p13, and HBZ (antisense transcript). mRNA transcripts are 5′-capped and 3′-polyadenylated. Alternative splicing yields mRNA for Env, Tax, Rex, p12, p13, p30, and HBZ.</p>
Full article ">Figure 3
<p>MA-membrane binding models for HIV-1 and HTLV-1. (<b>A</b>) Structures of HIV-1 myrMA (PDB code 2H3I) and HTLV-1 myr(–)MA (PDB code 7M1W). Structures highlight the HBR implicated in membrane binding (blue sticks). For HIV-1 myrMA, the following residues are not shown for clarity: myr group, residues 2–3 and 115–132. For HTLV-1 myr(–)MA, the following residues are not shown for clarity: 1–2 and 94–99. (<b>B</b>) Surface representation of the HIV-1 myrMA structure (PDB code 2H3I) highlighting residues that exhibited substantial chemical shift changes upon binding of <span class="html-italic">tr</span>-P(4,5)P<sub>2</sub> (PDB code 2H3V) and IP<sub>3</sub> (left and middle, respectively). Structures are viewed in identical orientations. The structure of HTLV-1 myr(–)MA bound to IP<sub>3</sub> is shown on the right. (<b>C</b>) Models of HIV-1 myrMA and HTLV-1 myr(–)MA bound to membrane showing interactions between PI(4,5)P<sub>2</sub> and/or PS and the HBR. Membrane bilayer was constructed by CHARMM-GUI [<a href="#B169-viruses-16-01528" class="html-bibr">169</a>].</p>
Full article ">Figure 4
<p>HIV-1 and HTLV-1 Gag hexamer structures. (<b>A</b>) The two HIV-1 CA molecules are displayed on the side of the hexamer, with CA<sub>NTD</sub> in cyan and CA<sub>CTD</sub> in orange. HIV-1 SP1 domains are shown in blue. The PDB codes are HIV-1 (5L93) [<a href="#B109-viruses-16-01528" class="html-bibr">109</a>], HTLV-1 CA<sub>NTD</sub> (8PUG) [<a href="#B183-viruses-16-01528" class="html-bibr">183</a>], and HTLV-1 CA<sub>CTD</sub> (8PUH) [<a href="#B183-viruses-16-01528" class="html-bibr">183</a>]. The cross-section of the HTLV-1 Gag lattice reconstruction map suggests a distinctive arrangement of the CA<sub>NTD</sub> and CA<sub>CTD</sub> compared to HIV-1. (<b>B</b>) Shown is the top view of the HIV-1 hexamer structure, which was generated by fitting HIV-1 CA (5L93) into the EM density of the immature HIV-1 lattice (EMD: 4017). The top view of the HTLV-1 Gag hexamer structure shown was generated by fitting CA<sub>NTD</sub> and CA<sub>CTD</sub> separately into the EM density of the immature HTLV-1 CA lattice (EMD: 17942). The flexible linker between HTLV-1 CA<sub>NTD</sub> and CA<sub>CTD</sub> is unstructured and is therefore not shown.</p>
Full article ">Figure 5
<p>Comparison of Env CT. (<b>A</b>) Schematic representation of the gp41 subunits, indicating the lengths of their respective cytoplasmic tails (25 and 150 amino acids for HTLV-1 and HIV-1, respectively. (<b>B</b>) Secondary structure representation of the HIV-1 gp41CT protein based on the NMR data [<a href="#B192-viruses-16-01528" class="html-bibr">192</a>]. (<b>C</b>) HIV-1 Env incorporation is mediated by interaction between the MA domain of the Gag lattice and gp41CT. For HTLV-1, the CT appears to contain functional motifs that play important roles in cell-to-cell infection and syncytium formation.</p>
Full article ">Figure 6
<p>Comparison of MA lattices based on structural data. (<b>A</b>) Schematic representation of the myrMA lattice in the immature and mature states based on the cryo-ET data [<a href="#B170-viruses-16-01528" class="html-bibr">170</a>]. The trimer–trimer interactions are mediated by the N-terminal domain in the vicinity of the myr group, while the PI(4,5)P<sub>2</sub> binding pocket is empty. In the mature myrMA lattice, PI(4,5)P<sub>2</sub> is bound to the cleft and myrMA trimer–trimer interactions are formed by the HBR and PI(4,5)P<sub>2</sub>. (<b>B</b>) Schematic illustration of the myrMA lattice based on the X-ray structure of myrMA. In this lattice, myrMA–myrMA interaction at the trimer–trimer interface is mediated by the N-terminal residues. Of note, myrMA–myrMA interaction at the trimer–trimer interface places the myr groups (red) in juxtaposition. The HBR and PI(4,5)P<sub>2</sub> binding cleft are also shown. Hexagons and triangles denote C6 and C3 symmetry, respectively.</p>
Full article ">
16 pages, 883 KiB  
Systematic Review
The Global Prevalence of HTLV-1 and HTLV-2 Infections among Immigrants and Refugees—A Systematic Review and Meta-Analysis
by Thaís Augusto Marinho, Michele Tiemi Okita, Rafael Alves Guimarães, Ana Laura de Sene Amâncio Zara, Karlla Antonieta Amorim Caetano, Sheila Araújo Teles, Márcia Alves Dias de Matos, Megmar Aparecida dos Santos Carneiro and Regina Maria Bringel Martins
Viruses 2024, 16(10), 1526; https://doi.org/10.3390/v16101526 - 27 Sep 2024
Viewed by 1684
Abstract
This is the first systematic review and meta-analysis to estimate the prevalence of human T-lymphotropic virus 1 and 2 (HTLV-1 and 2) infections among immigrants and refugees worldwide. PubMed/MEDLINE, Scopus, EMBASE, Web of Science, and Virtual Health Library (VHL) databases were searched for [...] Read more.
This is the first systematic review and meta-analysis to estimate the prevalence of human T-lymphotropic virus 1 and 2 (HTLV-1 and 2) infections among immigrants and refugees worldwide. PubMed/MEDLINE, Scopus, EMBASE, Web of Science, and Virtual Health Library (VHL) databases were searched for studies published from their inception to 6 January 2023. A meta-analysis using a generalized linear mixed model with a random effect was performed for HTLV-1 and HTLV-2. Subgroup analyses were performed based on the decade of study, sample size, confirmatory methods, region of study, risk group, and region of origin. Of the 381 studies initially identified, 21 were included. The pooled prevalence of HTLV-1 and HTLV-2 was 1.28% (95% CI: 0.58, 2.81) and 0.11% (95% CI: 0.04, 0.33), respectively. HTLV-1 prevalence differed significantly by region of origin, with the highest prevalence among those from the Western Pacific Region (7.27%; 95% CI: 2.94, 16.83). The subgroup analysis also showed significant differences between the estimates of HTLV-1 considering the decade of study, sample size, and region of study. For HTLV-2, significant differences were shown in relation to sample size, confirmatory methods, and risk group. The higher HTLV-1 prevalence found deserves public health attention in immigrant and refugee-receiving non-endemic countries. Full article
(This article belongs to the Special Issue Viral Infections in Special Populations)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram.</p>
Full article ">Figure 2
<p>Forest plot of the prevalence of HTLV-1 infection among immigrants and refugees [<a href="#B46-viruses-16-01526" class="html-bibr">46</a>,<a href="#B47-viruses-16-01526" class="html-bibr">47</a>,<a href="#B48-viruses-16-01526" class="html-bibr">48</a>,<a href="#B49-viruses-16-01526" class="html-bibr">49</a>,<a href="#B50-viruses-16-01526" class="html-bibr">50</a>,<a href="#B52-viruses-16-01526" class="html-bibr">52</a>,<a href="#B53-viruses-16-01526" class="html-bibr">53</a>,<a href="#B54-viruses-16-01526" class="html-bibr">54</a>,<a href="#B55-viruses-16-01526" class="html-bibr">55</a>,<a href="#B56-viruses-16-01526" class="html-bibr">56</a>,<a href="#B57-viruses-16-01526" class="html-bibr">57</a>,<a href="#B58-viruses-16-01526" class="html-bibr">58</a>,<a href="#B59-viruses-16-01526" class="html-bibr">59</a>,<a href="#B60-viruses-16-01526" class="html-bibr">60</a>,<a href="#B61-viruses-16-01526" class="html-bibr">61</a>,<a href="#B62-viruses-16-01526" class="html-bibr">62</a>,<a href="#B63-viruses-16-01526" class="html-bibr">63</a>,<a href="#B64-viruses-16-01526" class="html-bibr">64</a>,<a href="#B65-viruses-16-01526" class="html-bibr">65</a>,<a href="#B66-viruses-16-01526" class="html-bibr">66</a>,<a href="#B67-viruses-16-01526" class="html-bibr">67</a>].</p>
Full article ">Figure 3
<p>Forest plot of the prevalence of HTLV-2 infection among immigrants and refugees [<a href="#B46-viruses-16-01526" class="html-bibr">46</a>,<a href="#B47-viruses-16-01526" class="html-bibr">47</a>,<a href="#B48-viruses-16-01526" class="html-bibr">48</a>,<a href="#B49-viruses-16-01526" class="html-bibr">49</a>,<a href="#B50-viruses-16-01526" class="html-bibr">50</a>,<a href="#B52-viruses-16-01526" class="html-bibr">52</a>,<a href="#B53-viruses-16-01526" class="html-bibr">53</a>,<a href="#B54-viruses-16-01526" class="html-bibr">54</a>,<a href="#B55-viruses-16-01526" class="html-bibr">55</a>,<a href="#B56-viruses-16-01526" class="html-bibr">56</a>,<a href="#B62-viruses-16-01526" class="html-bibr">62</a>,<a href="#B63-viruses-16-01526" class="html-bibr">63</a>,<a href="#B64-viruses-16-01526" class="html-bibr">64</a>].</p>
Full article ">
21 pages, 3920 KiB  
Article
Intrafamilial Transmission of HTLV-1 and HTLV-2 in Indigenous Peoples of the Brazilian Amazon: Molecular Characterization and Phylogenetic Analysis
by Isabella Nogueira Abreu, Felipe Bonfim Freitas, Eliene Rodrigues Putira Sacuena, Gabriel dos Santos Pereira Neto, Bruno José Sarmento Botelho, Carlos Neandro Cordeiro Lima, Vanessa de Oliveira Freitas, Mike Barbosa dos Santos, Sandra Souza Lima, Ricardo Ishak, João Farias Guerreiro, Antonio Carlos Rosário Vallinoto and Izaura Maria Cayres Vallinoto
Viruses 2024, 16(10), 1525; https://doi.org/10.3390/v16101525 - 26 Sep 2024
Viewed by 1097
Abstract
Human T-limphotropic virus 1 infection has a global distribution, with a high prevalence in some regions of Brazil and the world, while HTLV-2 infection is endemic mainly among indigenous people and drug users. To analyze intrafamilial transmission of HTLV-1/2 in five Kayapó indigenous [...] Read more.
Human T-limphotropic virus 1 infection has a global distribution, with a high prevalence in some regions of Brazil and the world, while HTLV-2 infection is endemic mainly among indigenous people and drug users. To analyze intrafamilial transmission of HTLV-1/2 in five Kayapó indigenous peoples (Gorotire, Kararaô, Kokraimoro, Kubenkokre, and Xikrin do Bacajá), we investigated 1452 individuals who underwent serological and molecular tests. Among the 276 indigenous people with positive results, we identified intrafamily transmission in 42.7% of cases, representing 38 families. It was possible to suggest horizontal and vertical transmissions in 15.8% (6/38) and 47.4% (18/38) of the family groups, respectively. In 15.8%, it was not possible to suggest the route, which indicated that the transmission may have occurred through both vertical and horizontal routes. Through phylogenetic analyses, 35 samples positive for HTLV-2 were sequenced and classified as subtype 2c, and the two samples that tested positive for HTLV-1 were shown to belong to the cosmopolitan subtype, transcontinental subgroup (HTLV-1aA). This study confirms the intrafamilial transmission of HTLV-1/2 infection in indigenous people of the Brazilian Amazon, highlighting the importance of the sexual and mother-to-child transmission routes in maintaining the virus in these people. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

Figure 1
<p>A flowchart of the analysis of the individuals investigated highlights the viral type and the occurrence of intrafamily transmission. I.T.—Intrafamily transmission; N.I.—No information.</p>
Full article ">Figure 2
<p>Pedigree showing the possible routes of horizontal transmission (Families 2, 15, 19, 20, 21, and 34), vertical transmission (Families 1, 3–6, 8–12, 14, 17, 24, 25, 27, 31, 37 and 38), vertical and horizontal transmission (Families 23, 28, 30, 33, 35 and 36), vertical or horizontal transmission (Families 16, 18, 22, 26, 29 and 32) and uncommon cases (Families 7 and 13) of HTLV-1 and HTLV-2 among the indigenous peoples Kubenkokre (Families 1 to 18), Xikrin do Bacajá (Families 19 to 30), Kararaô (Family 31), Kokraimoro (Family 32) and Gorotire (Families 33 to 38) of the Kayapó ethnic group located in the Brazilian Amazon.</p>
Full article ">Figure 3
<p>Rooted phylogenetic tree showing the relationships between the HTLV-1 samples available in GenBank and those described in the present study (BRPA_78_Gorotire_R; BRPA_69_Gorotire_R; highlighted in red). The tree was constructed using the maximum likelihood method after partial alignment of nucleotides in the 5‘LTR-1 region. The statistical sustainability test (bootstrap) was applied with 1000 replicates from the sequence bank.</p>
Full article ">Figure 4
<p>Rooted phylogenetic tree showing the relationships between the HTLV-2 samples available in GenBank and the 35 samples (highlighted in red) described in the present study. The tree was constructed using the maximum likelihood method after partial alignment of nucleotides in the 5‘LTR-2 region. The statistical sustainability test (bootstrap) was applied with 1000 replicates from the sequence bank.</p>
Full article ">Figure 5
<p>Heatmap showing the distance matrix between the nucleotide sequences of the 5‘LTR region of HTLV-2 in the five family groups. (<b>A</b>) family 4; (<b>B</b>) family 23; (<b>C</b>) 29; (<b>D</b>) family 32 and (<b>E</b>) family 36.</p>
Full article ">Figure 6
<p>Heatmap showing the comparison between the samples sequenced for HTLV-2 in this study.</p>
Full article ">Figure 7
<p>Heatmap showing the comparison between samples sequenced for HTLV-2 among four indigenous peoples (Gorotire, Kokraimoro, Kubenkokre, and Xikrin do Bacajá).</p>
Full article ">
10 pages, 3671 KiB  
Case Report
Long-Term Survival of Patients with Adult T-Cell Leukemia/Lymphoma Treated with Amplified Natural Killer Cell Therapy
by Yuji Okubo, Sho Nagai, Yuta Katayama, Kunihiro Kitamura, Kazuhisa Hiwaki and Keisuke Teshigawara
Reports 2024, 7(3), 80; https://doi.org/10.3390/reports7030080 - 19 Sep 2024
Viewed by 1366
Abstract
Background: Adult T-cell leukemia/lymphoma (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1) after a long latent infection. HTLV-1 induces the indolent or aggressive type of leukemia in 5% of HTLV-1 carriers. ATL, especially the aggressive type, is resistant to multi-agent [...] Read more.
Background: Adult T-cell leukemia/lymphoma (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1) after a long latent infection. HTLV-1 induces the indolent or aggressive type of leukemia in 5% of HTLV-1 carriers. ATL, especially the aggressive type, is resistant to multi-agent chemotherapy. The indolent type often progresses to the aggressive type. Even in the most indolent-type cases, that is, smoldering ATL, the average survival time is 55.0 months. Case Presentation: Five patients with ATL were followed up for their clinical course after amplified natural killer cell (ANK) therapy. Four patients who received ANK therapy as first-line therapy achieved complete remission and showed long-term survival without aggressive conversion or relapse for more than 5 years. One patient was treated with multiagent chemotherapy due to acute exacerbation but relapsed 2 months later. She was subsequently treated with radiation and ANK therapy and survived for more than 6 years. Furthermore, ANK therapy enhanced the immune function of ATL patients to a level higher than that of normal individuals. Conclusions: ANK therapy has great potential as first-line treatment for ATL. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

Figure 1
<p>ANK therapy and overall survival. Light blue bars: time from clinical diagnosis to initiation of ANK therapy. Yellow bars: time from initiation of ANK therapy to death or last follow-up day (September 2020). Red bars: duration of ANK therapy. Small numbers: number of intravenous injections of ANK cells.</p>
Full article ">Figure 2
<p>Fluctuation of serum sIL-2R levels in Patient 2 (blue broken line). Serum levels of sIL-2R before ANK therapy. Patient 2 developed acute exacerbation and was treated with multi-agent chemotherapy. She had CR, and her serum sIL-2R levels decreased to 1018 U/mL. However, her ATL relapsed two months later.</p>
Full article ">Figure 3
<p>Fluctuation of serum sIL-2R levels in Patient 1 with ANK therapy after acute exacerbation of smoldering ATL. Reprinted/adapted with permission from Ref. [<a href="#B13-reports-07-00080" class="html-bibr">13</a>]. Copyright 2018, copyright Dr. Keisuke Teshigawara.</p>
Full article ">Figure 4
<p>The level of serum sIL-2R in patients receiving ANK therapy as first-line treatment. Serum sIL-2R levels in the patients who received ANK therapy as first-line treatment and their status of watchful waiting.</p>
Full article ">Figure 5
<p>Changes in serum sIL-2R levels in patient 2. Two months after the introduction of ANK therapy, ATL recurred and skin tumors appeared frequently. Radiotherapy was administered until the seventh course of ANK therapy, but then ANK therapy alone was administered for the next 9 months, and serum sIL-2R levels decreased.</p>
Full article ">Figure 6
<p>The effect of conventional and ANK therapies on the clinical course of Patient 2. Healing of multiple skin tumor and reduction in serum sIL-2R in Patient 2. Patient 2 relapsed and showed multiple skin tumors on her left forearm (Photo 1). These skin tumors were almost cured seven months after ANK therapy (Photo 2). Thereafter, the healthy status of the left forearm continued for five years (Photo 3).</p>
Full article ">Figure 7
<p>Comparison of NK cell activity between patents achieved complete remission and healthy controls (<span class="html-italic">n</span> = 20). Yellow line: Patient 4, red line: Patient 5.</p>
Full article ">
10 pages, 265 KiB  
Case Report
Modified Prophylactic Donor Lymphocyte Infusion (DLI) in an Adult T Cell Lymphoma/Leukemia (ATLL) Patient—Modality of Relapse Prevention
by Alexandra Ionete, Alexandru Bardas, Zsofia Varady, Madalina Vasilica, Orsolya Szegedi and Daniel Coriu
Diseases 2024, 12(9), 210; https://doi.org/10.3390/diseases12090210 - 11 Sep 2024
Viewed by 901
Abstract
Adult T-cell Leukemia/Lymphoma (ATLL) is a rare but aggressive malignancy associated with the human T-cell lymphotropic virus type 1 (HTLV-1). ATLL is a challenging malignancy characterized by its aggressive nature and poor prognosis. Despite advancements in treatment, relapse rates remain high. Donor lymphocyte [...] Read more.
Adult T-cell Leukemia/Lymphoma (ATLL) is a rare but aggressive malignancy associated with the human T-cell lymphotropic virus type 1 (HTLV-1). ATLL is a challenging malignancy characterized by its aggressive nature and poor prognosis. Despite advancements in treatment, relapse rates remain high. Donor lymphocyte infusion (DLI) is a promising therapeutic option post-hematopoietic stem cell transplantation (HSCT) to prevent relapse. However, the prophylactic use of DLI in ATLL patients remains underexplored. We report the case of a 45-year-old female diagnosed with ATLL. Following induction chemotherapy and successful HSCT, a modified prophylactic DLI regimen was administered, consisting of gradually increasing doses of donor lymphocytes. The patient demonstrated a favorable response with no significant graft-versus-host disease (GVHD) and maintained remission over a 40-month follow-up period, suggesting a potential benefit of this approach. This case highlights the potential efficacy and safety of modified prophylactic DLI in ATLL patients, warranting further investigation. Our findings suggest that modified prophylactic DLI is a viable option for ATLL patients post-HSCT, offering a balance between efficacy and safety. Future research should focus on optimizing DLI protocols and exploring biomarkers for response prediction. Full article
(This article belongs to the Section Oncology)
16 pages, 5075 KiB  
Article
The Oncoprotein Fra-2 Drives the Activation of Human Endogenous Retrovirus Env Expression in Adult T-Cell Leukemia/Lymphoma (ATLL) Patients
by Julie Tram, Laetitia Marty, Célima Mourouvin, Magali Abrantes, Ilham Jaafari, Raymond Césaire, Philippe Hélias, Benoit Barbeau, Jean-Michel Mesnard, Véronique Baccini, Laurent Chaloin and Jean-Marie Jr. Peloponese
Cells 2024, 13(18), 1517; https://doi.org/10.3390/cells13181517 - 10 Sep 2024
Viewed by 1036
Abstract
Human endogenous retroviruses (HERVs) are retroviral sequences integrated into 8% of the human genome resulting from ancient exogenous retroviral infections. Unlike endogenous retroviruses of other mammalian species, HERVs are mostly replication and retro-transposition defective, and their transcription is strictly regulated by epigenetic mechanisms [...] Read more.
Human endogenous retroviruses (HERVs) are retroviral sequences integrated into 8% of the human genome resulting from ancient exogenous retroviral infections. Unlike endogenous retroviruses of other mammalian species, HERVs are mostly replication and retro-transposition defective, and their transcription is strictly regulated by epigenetic mechanisms in normal cells. A significant addition to the growing body of research reveals that HERVs’ aberrant activation is often associated with offsetting diseases like autoimmunity, neurodegenerative diseases, cancers, and chemoresistance. Adult T-cell leukemia/lymphoma (ATLL) is a very aggressive and chemoresistant leukemia caused by the human T-cell leukemia virus type 1 (HTLV-1). The prognosis of ATLL remains poor despite several new agents being approved in the last few years. In the present study, we compare the expression of HERV genes in CD8+-depleted PBMCs from HTLV-1 asymptomatic carriers and patients with acute ATLL. Herein, we show that HERVs are highly upregulated in acute ATLL. Our results further demonstrate that the oncoprotein Fra-2 binds the LTR region and activates the transcription of several HERV families, including HERV-H and HERV-K families. This raises the exciting possibility that upregulated HERV expression could be a key factor in ATLL development and the observed chemoresistance, potentially leading to new therapeutic strategies and significantly impacting the field of oncology and virology. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Lymphomas)
Show Figures

Figure 1

Figure 1
<p>Prevalence of Abs against human HERV Env antigens in patients with acute ATL and the respective control groups. (<b>A</b>) HERV envelope (ENV) antigenemia in sera from non-infected patients NI (<span class="html-italic">n</span> = 7; black circle), HTLV-1 asymptomatic carriers (<span class="html-italic">n</span> = 7; green square), and acute ATLL patients ATLL (<span class="html-italic">n</span> = 7; red diamond). The dotted lines represent positivity thresholds calculated by ROC analysis. (<b>B</b>) The area under the curve (AUC) and its statistical significance are reported (ns <span class="html-italic">p</span> ≤ 0.05 and **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>Detection of human HERV mRNA by qRT-PCR in ATL-derived cell lines. (<b>A</b>–<b>C</b>) Expression of HERV mRNA was assessed in one HTLV-1-negative cell line (Jurkat) and three HTLV-1-derived cell lines (HUT102, C81–66, and ATL-2). (<b>D</b>,<b>E</b>) HERV gag mRNA expression was compared in Jurkat cells and HTLV-1-derived cell lines. (<b>F</b>) HERV-R-Pol mRNA was compared in Jurkat cells and HTLV-1-derived cell lines (** <span class="html-italic">p</span> ≤ 0.001). (<b>G</b>–<b>I</b>) Relative Fra-2, Tax, and HBZ expression in Jurkat cells and HTLV-1-derived cell lines (statistical significance was determined using a one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 3
<p>Detection of human HERV mRNA in CD8<sup>+</sup>-depleted PBMCs from HTLV-1-infected patients. (<b>A</b>–<b>D</b>) HERV mRNA was expressed in acute ATLL patients (red square) compared to HTLV-1 asymptomatic carriers (ACs) (black dot). (<b>E</b>,<b>F</b>) HERV gag mRNA expression was compared in AC (black dot) and ATLL patients (red square) (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.00001). (<b>G</b>) HERV-R-Pol was measured in AC and ATLL patients compared to ACs (** <span class="html-italic">p</span> ≤ 0.001). (<b>H</b>,<b>I</b>) The relative expression of Tax and HBZ in AC patients and ATL patients.</p>
Full article ">Figure 4
<p>HBZ does not activate HERV LTR. HEK293T was co-transfected with a plasmid carrying the luciferase reporter gene under the control of an NF-kB-dependent promoter (<b>A</b>), the collagenase promoter (<b>B</b>), or different HERV LTRs (<b>C</b>–<b>F</b>) and Tax-Flag, p65-Flag, or HBZ-Myc expression vectors, in addition to pRcActin-LacZ for the normalization of transfection efficiency. Cells were harvested 48 h. post-transfection and assayed for luciferase activity. The results show a fold increase compared to the mock control (set at a value of 1) and represent the mean values of three independently transfected cells. (<b>A</b>,<b>B</b>) Western blot analyses assessed the expression of Tax, p65, and HBZs. Actin is shown as a loading control (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, * <span class="html-italic">p</span> ≤ 0.01, ** <span class="html-italic">p</span> ≤ 0.001; *** <span class="html-italic">p</span> ≤ 0.00001).</p>
Full article ">Figure 5
<p>Fra-2 but not cFos activate the HERV LTR. HEK293T cells were co-transfected with a plasmid carrying the luciferase reporter gene under the control of the collagenase promoter in triplicate as shown (<b>A</b>) or different HERV 5′LTRs (<b>B</b>–<b>E</b>), different combinations of binding partners of AP-1-related transcription factors, and pRcActin-LacZ. The cells were harvested 48 h post-transfection and assayed for luciferase activity. The results show a fold increase in the mock control and represent the mean values of three independently transfected cell samples. (<b>F</b>) Western blot analyses were carried out to assess the expression of AP-1 transcription factors. Actin is shown as a loading control (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05,* <span class="html-italic">p</span> ≤ 0.01, ** <span class="html-italic">p</span> ≤ 0.001; *** <span class="html-italic">p</span> ≤ 0.0001 and **** <span class="html-italic">p</span> ≤ 0.00001).</p>
Full article ">Figure 6
<p>HBZ does not alter the activation of type I and type II HERV-H LTRs by Fra-2. (<b>A</b>) HEK293T cells were co-transfected with the different AP-1 in the absence or presence of HBZ and a plasmid carrying the luciferase reporter gene under the control of the collagenase promoter. (<b>B</b>) Western blot analyses were carried out to assess the expression of AP-1 transcription factors and HBZ. Actin is shown as a loading control. HEK293T cells were co-transfected with a plasmid carrying the luciferase reporter gene under the control of the collagenase promoter (<b>A</b>) or different HERV 5′LTRs (<b>C</b>–<b>E</b>), different AP-1 expression vectors, and pRcActin-LacZ in the presence or absence of HBZ. The cells were harvested 48 h post-transfection and assayed for luciferase activity. The results show a fold increase in the mock control and represent the mean values of three independently transfected cell samples (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, * <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 7
<p>HERV Env mRNA detected in HEK293T cells stably expressing Fra-2. (<b>A</b>) Western blot analyses were carried out on the lysate of two pools of HEK293-Fra2 to assess the expression of Fra-2. Beta actin is shown as a loading control. (<b>B</b>–<b>E</b>) HEK293T stably expressing Fra2 was harvested at different passages (from p3 to p8), and the expression of HERV-H Env, HERV-R Env, HERV-K Env, and HERV-E gag mRNAs was assessed by qRT-PCR (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, * <span class="html-italic">p</span> ≤ 0.01; ** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">Figure 8
<p>Kinetic analysis of Fra-2 and HERV Env mRNA in CD8<sup>+</sup>-depleted PBMCs from asymptomatic carriers and ATL patients. CD8<sup>+</sup>-depleted PBMCs from five ATL patients were cultivated ex vivo for five days, and Fra-2 (closed black square) (<b>A</b>), HERV-H Env (closed green triangle) (<b>B</b>), and HERV-K Env mRNA (closed red triangle) (<b>C</b>) were quantified at different time points using qRT-PCR (one-way ANOVA test with Dunn‘s multiple comparisons post-test; *** <span class="html-italic">p</span> ≤ 0.0001 and **** <span class="html-italic">p</span> ≤ 0.00001 (<b>D</b>,<b>E</b>) Relevance of Fra-2 and HERV Env mRNA expression in ATL patients, as analyzed with the Pearson correlation Test.</p>
Full article ">Figure 9
<p>Fra-2 binds to HERV-H-LTR in ATL-2 cells. ChIP assays were performed on chromatin prepared from the indicated ATL-2 cell lines using antibodies against Fra-2. Data are presented as fold enrichment relative to the IgG control. Data are an average of three independent experiments. Error bars represent the SEM (two-way ANOVA <span class="html-italic">t</span>-test, ns <span class="html-italic">p</span> ≤ 0.05 ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
18 pages, 5236 KiB  
Article
Dendritic Cells Pulsed with HAM/TSP Exosomes Sensitize CD4 T Cells to Enhance HTLV-1 Infection, Induce Helper T-Cell Polarization, and Decrease Cytotoxic T-Cell Response
by Julie Joseph, Thomas A. Premeaux, Ritesh Tandon, Edward L. Murphy, Roberta Bruhn, Christophe Nicot, Bobby Brooke Herrera, Alexander Lemenze, Reem Alatrash, Prince Baffour Tonto, Lishomwa C. Ndhlovu and Pooja Jain
Viruses 2024, 16(9), 1443; https://doi.org/10.3390/v16091443 - 10 Sep 2024
Viewed by 1210
Abstract
HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a progressive demyelinating disease of the spinal cord due to chronic inflammation. Hallmarks of disease pathology include dysfunctional anti-viral responses and the infiltration of HTLV-1-infected CD4+ T cells and HTLV-1-specific CD8+ T cells in the central nervous [...] Read more.
HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a progressive demyelinating disease of the spinal cord due to chronic inflammation. Hallmarks of disease pathology include dysfunctional anti-viral responses and the infiltration of HTLV-1-infected CD4+ T cells and HTLV-1-specific CD8+ T cells in the central nervous system. HAM/TSP individuals exhibit CD4+ and CD8+ T cells with elevated co-expression of multiple inhibitory immune checkpoint proteins (ICPs), but ICP blockade strategies can only partially restore CD8+ T-cell effector function. Exosomes, small extracellular vesicles, can enhance the spread of viral infections and blunt anti-viral responses. Here, we evaluated the impact of exosomes isolated from HTLV-1-infected cells and HAM/TSP patient sera on dendritic cell (DC) and T-cell phenotypes and function. We observed that exosomes derived from HTLV-infected cell lines (OSP2) elicit proinflammatory cytokine responses in DCs, promote helper CD4+ T-cell polarization, and suppress CD8+ T-cell effector function. Furthermore, exosomes from individuals with HAM/TSP stimulate CD4+ T-cell polarization, marked by increased Th1 and regulatory T-cell differentiation. We conclude that exosomes in the setting of HAM/TSP are detrimental to DC and T-cell function and may contribute to the progression of pathology with HTLV-1 infection. Full article
(This article belongs to the Special Issue Human T-Cell Leukemia Virus (HTLV) Infection and Treatment)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Exosomes activate and elicit proinflammatory responses in dendritic cells. (<b>A</b>) Representative phenotype of myeloid dendritic cell (mDC) and plasmacytoid dendritic cell (pDC) population differences with exosome stimulation (<span class="html-italic">n</span> = 4). GMFI levels of CD40, CD80, and CD86 in mDC and pDC populations untreated or stimulated with exosomes. Bars represent mean ± standard deviation (SD). (<b>B</b>) Expression of cytokines associated with Th1, Th2, Th17, and Treg polarization in total DCs stimulated with OSP2 cell-derived exosomes or LPS. Cytokines were grouped according to associated Th1 (IFN-γ, TNF-α, and IL-2), Th2 (IL-4, IL-12a, and IL-13), Th17 (IL-6 and IL-17a), and Treg (IL-10 and TGF-β) subsets. (<b>C</b>) Cytokine levels (pg/mL) in supernatants of mDCs (<b>left</b>) and pDCs (<b>right</b>) untreated or exosome-stimulated. Statistical differences were determined by one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 2
<p>Dendritic cell-pulsed exosomes polarize T cells. (<b>A</b>) Exosome stimulation schematic and representative gating strategy. Donor CD3+ T cells and matched total DCs were isolated (<span class="html-italic">n</span> = 4). (<b>B</b>) DCs were exposed to OSP2-derived exosomes for 24 h and subsequently co-incubated with donor-matched T cells for another 24 h. Absolute count of CD4+ T-cells expressing subtype-associated markers: Th1 (IFN-γ, CCR5), Th2 (IL-4, CCR4), Th17 (IL-17, CCR6), and Treg (CD25, FoxP3). Quantification (pg/mL) of IFN-γ, TGF-β, and TNF-α cytokine levels in exosome-stimulated DC–T-cell co-culture (<span class="html-italic">n</span> = 3). (<b>C</b>) Representative flow plots and quantification (<span class="html-italic">n</span> = 4) of percent positive CD4+ T cells expressing functional markers of IFN-γ, IL-4, IL-17a, CD25, CCR5, and CCR6 after stimulation with OSP2 cell-derived exosomes. Bars represent mean ± standard deviation (SD). Statistical differences were determined by one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2 Cont.
<p>Dendritic cell-pulsed exosomes polarize T cells. (<b>A</b>) Exosome stimulation schematic and representative gating strategy. Donor CD3+ T cells and matched total DCs were isolated (<span class="html-italic">n</span> = 4). (<b>B</b>) DCs were exposed to OSP2-derived exosomes for 24 h and subsequently co-incubated with donor-matched T cells for another 24 h. Absolute count of CD4+ T-cells expressing subtype-associated markers: Th1 (IFN-γ, CCR5), Th2 (IL-4, CCR4), Th17 (IL-17, CCR6), and Treg (CD25, FoxP3). Quantification (pg/mL) of IFN-γ, TGF-β, and TNF-α cytokine levels in exosome-stimulated DC–T-cell co-culture (<span class="html-italic">n</span> = 3). (<b>C</b>) Representative flow plots and quantification (<span class="html-italic">n</span> = 4) of percent positive CD4+ T cells expressing functional markers of IFN-γ, IL-4, IL-17a, CD25, CCR5, and CCR6 after stimulation with OSP2 cell-derived exosomes. Bars represent mean ± standard deviation (SD). Statistical differences were determined by one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Immune checkpoint and anti-viral cytokine expression of exosomes from individuals with HAM/TSP. (<b>A</b>) Representative NTA of exosomes isolated from asymptomatic carrier (AC, <span class="html-italic">n</span> = 1) and HAM/TSP (HAM, <span class="html-italic">n</span> = 3) patient sera in individuals infected with HTLV-1 or HTLV-2. (<b>B</b>) Quantification of immune checkpoint proteins BTLA, LAG-3, PD-1, and PD-L2 in exosomes from AC and HAM/TSP patient sera from individuals infected with HTLV-1, left, or HTLV-2, right. (<b>C</b>) Quantification of IFN-γ and TNF-α levels in exosomes from AC and HAM/TSP patient sera from individuals infected with HTLV-1, left, or HTLV-2, right. Bars represent mean ± standard deviation (SD). Statistical differences were determined by unpaired two-tailed T-tests of technical replicates, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>HAM/TSP exosomes skew Th1/Treg responses and sensitize cells toward infection. (<b>A</b>) Exosome stimulation schematic and representative gating strategy. CD3+ T cells and total DCs were isolated from matched donors (<span class="html-italic">n</span> = 4). DCs were stimulated with exosomes from HTLV-1 patient sera for 24 h and subsequently co-incubated with donor-matched T cells for another 24 h. Differences in GMFI of representative markers are plotted. Bottom, absolute count of CD4+ T cells expressing subtype-associated markers: Th1 (IFN-y and CCR5), Th2 (IL-4 and CCR4), Th17 (IL-17 and CCR6), and Treg (CD25 and FoxP3). (<b>B</b>) Quantification (pg/mL) of IFN-γ, TGF-β, and TNF-α cytokine levels in patient exosome-stimulated DC–T-cell co-culture. (<b>C</b>) Representative flow plots and quantification (<span class="html-italic">n</span> = 4) of percent positive CD4+ T cells expressing functional markers of IFN-γ, IL-4, IL-17a, CD25, CCR5, and CCR6 after stimulation with HTLV-1 patient-derived exosomes. Bars represent mean ± standard deviation (SD). Statistical differences were determined by one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4 Cont.
<p>HAM/TSP exosomes skew Th1/Treg responses and sensitize cells toward infection. (<b>A</b>) Exosome stimulation schematic and representative gating strategy. CD3+ T cells and total DCs were isolated from matched donors (<span class="html-italic">n</span> = 4). DCs were stimulated with exosomes from HTLV-1 patient sera for 24 h and subsequently co-incubated with donor-matched T cells for another 24 h. Differences in GMFI of representative markers are plotted. Bottom, absolute count of CD4+ T cells expressing subtype-associated markers: Th1 (IFN-y and CCR5), Th2 (IL-4 and CCR4), Th17 (IL-17 and CCR6), and Treg (CD25 and FoxP3). (<b>B</b>) Quantification (pg/mL) of IFN-γ, TGF-β, and TNF-α cytokine levels in patient exosome-stimulated DC–T-cell co-culture. (<b>C</b>) Representative flow plots and quantification (<span class="html-italic">n</span> = 4) of percent positive CD4+ T cells expressing functional markers of IFN-γ, IL-4, IL-17a, CD25, CCR5, and CCR6 after stimulation with HTLV-1 patient-derived exosomes. Bars represent mean ± standard deviation (SD). Statistical differences were determined by one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>HAM/TSP exosomes skew T cells to Th1/Treg profiles in patient PBMCs. (<b>A</b>) Representative flow plots and quantification (<span class="html-italic">n</span> = 4) of the polarization of T cells from individuals with HTLV-1 with or without HTLV-1 sera-derived exosomes. (<b>B</b>) Representative flow plots and quantification (<span class="html-italic">n</span> = 4) of HTLV-1-infected patient CD4+ T cells with or without HTLV-1 sera-derived exosome stimulation. Bars represent mean ± standard deviation (SD). Statistical differences were determined by one-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Exosomes diminish CD8+ T-cell activity. (<b>A</b>) Schematic of treatment and gating strategy of CD8+ T cells. CD3+ T cells and total DCs were isolated from matched donors (<span class="html-italic">n</span> = 4). DCs were stimulated with OSP2-derived exosomes for 24 h and subsequently co-incubated with donor-matched T cells for another 24 h. Absolute counts of CD8+ T cells expressing MIP-1a, Granzyme B, Perforin, IFN-γ, PD-1, and Ki67 after co-incubation with exosome-pulsed DCs. (<b>B</b>) Absolute counts of CD8+ T cells after CD3+/CD28+ activation and stimulation with OSP2 exosomes alone, or exosomes incubated with a cocktail of anti-PD-L2, anti-BTLA, anti-LAG-3, and anti-PD-1 blocking antibodies. (<b>C</b>) Absolute counts of CD8+ T cells expressing MIP-1a, Granzyme B, Perforin, IFN-γ, PD-1, and Ki67 in HTLV-1-infected patient T cells (<span class="html-italic">n</span> = 4) stimulated with patient sera exosomes alone or exosomes incubated with a cocktail of anti-PD-L2, anti-BTLA, anti-LAG-3, and anti-PD-1 blocking antibodies. Counts were determined by experiments with <span class="html-italic">n</span> = 3/4 healthy donors, and error bars represent SEM of donor variation. Bars represent mean ± standard deviation (SD). Statistical differences were determined by one-way ANOVA. (<b>D</b>) Western blot and densitometry of CD3/CD28-stimulated T cells treated with Jurkat and OSP2 exosomes alone, or exosomes incubated with a cocktail of anti-PD-L2, anti-BTLA, anti-LAG-3, and anti-PD-1 blocking antibodies. Blot was probed for phosphorylated AKT or ERK. Error bars represent SEM of blot variation; statistical differences were determined by paired two-tailed T-test, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
21 pages, 8221 KiB  
Article
MicroRNA Profiling in Papillary Thyroid Cancer
by Richard Armos, Bence Bojtor, Marton Papp, Ildiko Illyes, Balazs Lengyel, Andras Kiss, Balazs Szili, Balint Tobias, Bernadett Balla, Henriett Piko, Anett Illes, Zsuzsanna Putz, Andras Kiss, Erika Toth, Istvan Takacs, Janos P. Kosa and Peter Lakatos
Int. J. Mol. Sci. 2024, 25(17), 9362; https://doi.org/10.3390/ijms25179362 - 29 Aug 2024
Cited by 2 | Viewed by 1268
Abstract
Genetic alterations are well known to be related to the pathogenesis and prognosis of papillary thyroid carcinoma (PTC). Some miRNA expression dysregulations have previously been described in the context of cancer development including thyroid carcinoma. In our study, we performed original molecular diagnostics [...] Read more.
Genetic alterations are well known to be related to the pathogenesis and prognosis of papillary thyroid carcinoma (PTC). Some miRNA expression dysregulations have previously been described in the context of cancer development including thyroid carcinoma. In our study, we performed original molecular diagnostics on tissue samples related to our own patients. We aimed to identify all dysregulated miRNAs in potential association with PTC development via sequencing much higher numbers of control-matched PTC tissue samples and analyzing a wider variety of miRNA types than previous studies. We analyzed the expression levels of 2656 different human miRNAs in the context of 236 thyroid tissue samples (118 tumor and control pairs) related to anonymized PTC cases. Also, KEGG pathway enrichment analysis and GO framework analysis were used to establish the links between miRNA dysregulation and certain biological processes, pathways of signaling, molecular functions, and cellular components. A total of 30 significant differential miRNA expressions with at least ±1 log2 fold change were found related to PTC including, e.g., miR-551b, miR-146b, miR-221, miR-222, and miR-375, among others, being highly upregulated, as well as miR-873 and miR-204 being downregulated. In addition, we identified miRNA patterns in vast databases (KEGG and GO) closely similar to that of PTC including, e.g., miRNA patterns of prostate cancer, HTLV infection, HIF-1 signaling, cellular responses to growth factor stimulus and organic substance, and negative regulation of gene expression. We also found 352 potential associations between certain miRNA expressions and states of clinicopathological variables. Our findings—supported by the largest case number of original matched-control PTC–miRNA relation research—suggest a distinct miRNA expression profile in PTC that could contribute to a deeper understanding of the underlying molecular mechanisms promoting the pathogenesis of the disease. Moreover, significant miRNA expression deviations and their signaling pathways in PTC presented in our study may serve as potential biomarkers for PTC diagnosis and prognosis or even therapeutic targets in the future. Full article
(This article belongs to the Special Issue Mechanisms of Thyroid Hormone Signaling in Human Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>This workflow diagram illustrates the steps of the miRNA analysis of PTC patients. We reviewed 161 anonymized PTC cases of the tissue archives, from which 129 were selected as eligible based on histopathological evaluation. A total of 258 thyroid tissue samples (129-129 tumor and control samples, respectively) related to these cases were then collected and subjected to sectioning. Then, the sections underwent miRNA isolation and quality control of RNA concentrations, leading to the exclusion of samples being evaluated as inapplicable isolate specimens. The remaining samples were then subjected to sequencing, after which a bioinformatic and statistical assessment was conducted on the data in the context of 2656 different miRNA types in total. Bioinformatic evaluation led to the further exclusion of 1 sample pair (both tumor and control) due to insufficient sequencing yield detected. Finally, we were able to establish those miRNAs which show significantly different expression patterns in PTC and non-PTC tissues related to 118 patients in total.</p>
Full article ">Figure 2
<p>The bars of this chart present the log<sub>2</sub> fold change of the top 20 miRNAs (named on the vertical axis) selected based on their significantly different expression profiles between the cancer and control groups. Bars that extend to the right of the zero line (red) show overexpression of the particular miRNA in tumor tissue, while those to the left (blue) indicate underexpression.</p>
Full article ">Figure 3
<p>This volcano plot illustrates the different expressions of the miRNAs. On the horizontal axis, the log<sub>2</sub> fold change is represented, highlighting the magnitude of expression deviations. The vertical axis illustrates the negative logarithm of the <span class="html-italic">p</span>-value (−log<sub>10</sub>P), reflecting the statistical significance of the expression change related to each miRNA. Dots above the horizontal threshold line (blue and red) represent miRNAs that pass the significance criterion. Dots to the right or left of the vertical threshold lines (red) indicate not only high significance levels but also a substantial overexpression or underexpression of the corresponding miRNAs, respectively. Dots below the horizontal threshold line represent miRNAs with large fold changes that are not statistically significant (green) or miRNAs that do not meet any of the threshold values (gray).</p>
Full article ">Figure 4
<p>In this heatmap, the rows correspond to the “top miRNAs” (n = 30) of this study, selected based on their significantly different expression levels between tumor (red) and control (blue) groups categorized by histopathological characteristics. (<b>A</b>) Each column represents one tissue sample (n = 236) subjected to molecular analysis. The color intensity within each cell reflects the Z-score derived from the normalized number of reads aligned to significant “top miRNAs”, with more red shades indicating higher expression and more blue indicating a lower expression pattern of the particular miRNA of the row. (<b>B</b>) Hierarchical clustering is applied to both “top miRNAs” and samples of the two groups, as shown by the black branches, grouping similar expression profiles together. The vertical dendrogram (black lines on the vertical axis) illustrates the hierarchical clustering of “top miRNAs”, categorizing them based on the similarity in their expression patterns across all samples, while the horizontal dendrogram (black branches on the horizontal axis) represents the hierarchical clustering of samples, highlighting that the samples with similar miRNA expression profiles tend to fall into the same (either control or tumor) group (<b>C</b>,<b>D</b>).</p>
Full article ">Figure 5
<p>Comparative principal component analysis of miRNA expressions in tumor and control samples. In plot (<b>A</b>), a PCA of all miRNA expressions tested is shown, with the horizontal axis representing Principal Component 1 (PC1), which accounts for 44.27% of the variance, and the vertical axis representing Principal Component 2 (PC2), accounting for 17.78% of the variance. Variables of the control group are marked in red and the tumor group in blue, indicating moderate separation along PC1, suggesting differential expression patterns between the two states. Plot (<b>B</b>) however displays a PCA focused exclusively on miRNA expressions found to be significant previously, with PC1 explaining a dominant 86.07% of the variance and PC2 accounting for 12.14%. Here, the separation between the two groups is more pronounced along PC1, indicating an explicit distinction in the expression profiles. The juxtaposition of these two plots highlights that specific miRNAs (marked as significant) contribute mostly to the molecular variance between the tumor and non-tumor conditions. The comparison illustrates the utility of focusing on significant miRNAs for a more targeted understanding of the molecular background of PTC.</p>
Full article ">Figure 6
<p>KEGG and Gene Ontology (GO) enrichment analyses (ORA—over-representation analysis) based on statistically significant (<span class="html-italic">p</span> ≤ 0.05) miRNAs of this study. Associations were found between the miRNA expression patterns in PTC marked as significant and the molecular patterns of pathways (<b>A</b>) listed in the KEGG database as well as biological processes (<b>B</b>), cellular components (<b>C</b>), and molecular functions (<b>D</b>) listed in the GO database. Based on the strength of significance, the plot visualizes the top 20 molecular patterns of the KEGG and GO databases showing potential correlation with PTC. Each bar represents a pathway, a biological process, a cellular component, or a molecular function of these databases (vertical axes), with the length of the bar reflecting the significance of a possible association with PTC as indicated by the −log10 of the adjusted <span class="html-italic">p</span>-value (P.adj) (horizontal axes). The color gradient conveys the adjusted <span class="html-italic">p</span>-value, transitioning from yellow (less significant) to dark purple (more significant). The data suggest that these molecular patterns (<b>A</b>–<b>D</b>) may be influenced by the same miRNAs as the development and/or progression of PTC.</p>
Full article ">Figure 6 Cont.
<p>KEGG and Gene Ontology (GO) enrichment analyses (ORA—over-representation analysis) based on statistically significant (<span class="html-italic">p</span> ≤ 0.05) miRNAs of this study. Associations were found between the miRNA expression patterns in PTC marked as significant and the molecular patterns of pathways (<b>A</b>) listed in the KEGG database as well as biological processes (<b>B</b>), cellular components (<b>C</b>), and molecular functions (<b>D</b>) listed in the GO database. Based on the strength of significance, the plot visualizes the top 20 molecular patterns of the KEGG and GO databases showing potential correlation with PTC. Each bar represents a pathway, a biological process, a cellular component, or a molecular function of these databases (vertical axes), with the length of the bar reflecting the significance of a possible association with PTC as indicated by the −log10 of the adjusted <span class="html-italic">p</span>-value (P.adj) (horizontal axes). The color gradient conveys the adjusted <span class="html-italic">p</span>-value, transitioning from yellow (less significant) to dark purple (more significant). The data suggest that these molecular patterns (<b>A</b>–<b>D</b>) may be influenced by the same miRNAs as the development and/or progression of PTC.</p>
Full article ">Figure 7
<p>Triple circle network graph illustrating the most relevant miRNA expression differences between certain states of the examined clinicopathological variables such as age, sex, ATA risk, and stages (TNM and AJCC eighth edition) (middle circle, black nodes). Differentially expressed miRNAs within the control samples are represented as blue nodes (outermost circle), whereas they are indicated as red nodes in the context of tumor samples (innermost circle). All red and blue nodes represent a significant change (<span class="html-italic">p</span> &lt; 0.05) in miRNA expression in relation to at least one clinicopathological variable. The significant associations are indicated by lines, with blue indicating negative changes and red indicating positive changes in miRNA expressions. The color gradient of the lines from blue to red represents the log<sub>2</sub> fold change (log<sub>2</sub>FC) of miRNA expressions, with darker shades representing greater expression differences and thus stronger links. To provide a clear and uncluttered visual representation of the network structure, the graph is devoid of any node labels related to associations with log<sub>2</sub>FC values between 10 and −10.</p>
Full article ">
Back to TopTop