[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
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (87)

Search Parameters:
Keywords = retargeting

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2709 KiB  
Article
PD1-Targeted Transgene Delivery to Treg Cells
by Vladislav A. Zhuchkov, Yulia E. Kravchenko, Elena I. Frolova and Stepan P. Chumakov
Viruses 2024, 16(12), 1940; https://doi.org/10.3390/v16121940 - 19 Dec 2024
Viewed by 378
Abstract
Achieving the precise targeting of lentiviral vectors (LVs) to specific cell populations is crucial for effective gene therapy, particularly in cancer treatment where the modulation of the tumor microenvironment can enhance anti-tumor immunity. Programmed cell death protein 1 (PD-1) is overexpressed on activated [...] Read more.
Achieving the precise targeting of lentiviral vectors (LVs) to specific cell populations is crucial for effective gene therapy, particularly in cancer treatment where the modulation of the tumor microenvironment can enhance anti-tumor immunity. Programmed cell death protein 1 (PD-1) is overexpressed on activated tumor-infiltrating T lymphocytes, including regulatory T cells that suppress immune responses via FOXP3 expression. We developed PD1-targeted LVs by incorporating the anti-PD1 nanobody nb102c3 into receptor-blinded measles virus H and VSV-Gmut glycoproteins. We assessed the retargeting potential of nb102c3 and evaluated transduction efficiency in activated T lymphocytes. FOXP3 expression was suppressed using shRNA delivered by these LVs. Our results demonstrate that PD1-targeted LVs exerted pronounced tropism towards PD1+ cells, enabling the selective transduction of activated T lymphocytes while sparing naive T cells. The suppression of FOXP3 in Tregs reduced their suppressive activity. PD1-targeted glycoprotein H provided greater specificity, whereas the VSV-Gmut, together with the anti-PD1 pseudoreceptor, achieved higher viral titers but was less selective. Our study demonstrates that PD1-targeted LVs may offer a novel strategy to modulate immune responses within the tumor microenvironment with the potential for developing new therapeutic strategies aimed at enhancing anti-tumor immunity. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic representations of retargeted measles (Hc∆18AA-102c3, 4AHc∆24AA-102c3), Nipah (Nip∆34Gm4-102c3), and VSV (VSVG<sub>mut</sub>+102c3R) glycoproteins tested for lentivector retargeting. (<b>B</b>) Predicted structure of the homodimer of the measles H-protein head fused with nb102c3. (<b>C</b>) Microphotographs of the syncytia formation test performed on HEK-293T and HEK-293T-PD1 cells transfected with Hc∆18AA-102c3, 4AHc∆24AA-102c3, and Nip∆34Gm4-102c3 with corresponding F-protein-encoding plasmids and the lentivector plasmid encoding tagRFP (red channel). (<b>D</b>) Infectious titers of all retargeted LVs (i.u.) and selectivity (fold). (<b>E</b>) FACS plots of HEK-293T and HEK-293T-PD1 cells transduced with a corresponding lentivector carrying the tagRFP sequence (1 mL of non-concentrated lentivirus sample per 35 mm well).</p>
Full article ">Figure 2
<p>(<b>A</b>) qPCR quantitation of PDCD1 levels in PBMCs cultured with IL-2 (ctrl), IL-2 + PHA-M (1), PMA + ionomycin + PHA-M (2), IL-2 + PHA-M + PMA + ionomycin (3), and IL-2 + PHA-M + PMA + ionomycin + IFNγ + IL-4 + IL-12 (4), **—<span class="html-italic">p</span> = 0.007. (<b>B</b>) nanoLuc luminescence levels normalized to cell count after transduction with lentiviral vectors of CD4<sup>+</sup> T cells with or without transduction enhancers (Treatment 3, as in (<b>A</b>), and Treatment 3-PB-F108), and in the presence of 50 µg/mL azidothymidine (Treatment 3 + AZT). (<b>C</b>) nanoLuc luminescence levels normalized to cell count after transduction with control (4AHc∆24) and targeted (4AHc∆24AA-102c3, VSVGmut+102c3R) lentivectors, **—<span class="html-italic">p</span> &lt; 0.0075. (<b>D</b>) nanoLuc luminescence levels in transduced CD4<sup>+</sup> T cells, **—<span class="html-italic">p</span> = 0.0018, ***—<span class="html-italic">p</span> = 0.0005.</p>
Full article ">Figure 3
<p>shRNA knockdown of FOXP3 with a non-targeted lentivector. (<b>A</b>) Schematic representation of shRNA-expressing lentivector and its cassette integrated in the genome. (<b>B</b>) qPCR quantitation of FOXP3 expression in stimulated CD4<sup>+</sup> T cells and Tregs transduced with shFOXP3 and control shRNA, **—<span class="html-italic">p</span> = 0.0065. (<b>C</b>) qPCR quantitation of FOXP3 expression in PD1<sup>+</sup> CD4<sup>+</sup> T cells and PD1<sup>+</sup> Tregs transduced with shFOXP3 and control shRNA, *—<span class="html-italic">p</span> = 0.015. (<b>D</b>) TGFβ levels in media of PD1<sup>−</sup> and PD1<sup>+</sup> Tregs transduced with shFOXP3 and the control, ****—<span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Cytometry measurements of CD4<sup>+</sup> PD1<sup>+</sup> T lymphocytes co-cultured with U937-tagGFP and transduced with 4AHc∆24AA-102c3-pseudotyped (<b>A</b>) VSVG<sub>mut</sub>+102c3R-pseudotyped (<b>B</b>) and 4AHc∆24-pseudotyped (<b>C</b>) LVs carrying the tagRFP sequence. (<b>D</b>) FOXP3 expression in PD1<sup>+</sup> Tregs co-cultured with U937 cells and transduced with different pseudotypes of LVs carrying shFOXP3 or control shRNA. *—<span class="html-italic">p</span> = 0.0485, ns—<span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">
6 pages, 610 KiB  
Case Report
Multiple Renal Infarctions in Spontaneous Double Renal Artery Dissection: A Case Report
by Gaetano Ferrara, Michelangelo Nasuto, Francesco Napolitano, Giovanni Ciccarese and Filippo Aucella
J. Clin. Med. 2024, 13(23), 7307; https://doi.org/10.3390/jcm13237307 - 1 Dec 2024
Viewed by 430
Abstract
Background: As spontaneous renal artery dissection (SRAD) is a rare cause of abdominal pain, bilateral dissection is an extremely rare event. Only approximately two hundred cases of SRAD have been reported in the literature. The diagnosis is often delayed due to the rarity [...] Read more.
Background: As spontaneous renal artery dissection (SRAD) is a rare cause of abdominal pain, bilateral dissection is an extremely rare event. Only approximately two hundred cases of SRAD have been reported in the literature. The diagnosis is often delayed due to the rarity of the disease and non-specific clinical presentations such as flank pain, hypertension, fever, nausea, vomiting, and hematuria, which can be often misdiagnosed as a genito-urinary infection or gastrointestinal or bowel disease. Before 1980, the diagnosis of SRAD was mostly confirmed via autopsy or, rarely, via angiography. At present, the diagnosis is made using advanced imaging approaches, including computed tomography angiography (CTA) and magnetic resonance angiography (MRA), with a higher number of incidentally diagnosed SRADs. Methods: we performed laboratory tests and radiological examinations (computed abdominal tomography and multiplanar reconstruction) that revealed multiple infarctions and ischemic areas with hypoperfusion in the upper middle third of the left kidney and in a large part of middle and lower areas of the right kidney; the left renal artery exhibited increased intimal thickening and arteritis. Results: The multiplanar reconstruction revealed bilateral renal artery dissection and multiple arterial infarctions disseminated throughout both kidneys. After a clinical follow-up and hypertension retargeting, the patient was discharged with dual antiplatelet therapy and ACE inhibitor drugs. No lipid-lowering therapy was needed. Conclusions: Spontaneous renal artery dissection (SRAD) is a rare clinical event that typically presents with acute low-back or flank pain, hypertension, fever, hematuria, and acute renal failure. The condition could be misdiagnosed or receive a delayed diagnosis due to its relative rarity and non-specific presentation. The gold standard is enhanced computed tomography (CT) scans, and if the diagnosis is positive, vascular multiplanar reconstruction is generally suggested, as it can display lesions more clearly. Over 300 cases have been reported since the first characterization of SRAD; however, to date, a consensus has not been reached on the most appropriate treatment. Conservative therapy, open surgery, and intravascular intervention have been reported as treatments for SRAD. Full article
(This article belongs to the Special Issue Advanced Imaging Techniques for Nephrology and Urology)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>–<b>d</b>): Multiplanar reconstructions on Anglo-CT scan, arterial phase. Right kidney: focal ischemic area (white arrow) associated with sub-acute dissection of the inferior polar artery (blue arrow). Left kidney: acute dissection of renal artery (blue arrow with dots) with focal acute ischemic area (red arrow).</p>
Full article ">Figure 2
<p>(<b>a</b>,<b>b</b>): Digital subtraction angiography (DSA). Right kidney (<b>a</b>): segmental renal artery occlusion (white arrow) consequent to distal dissection (blue arrows) of the inferior polar artery originating from the abdominal aorta. Left kidney (<b>b</b>): renal artery dissection (blue arrow with dots) with occlusion of smaller superior and inferior segmental branches and their corresponding focal de-vascularized areas (red arrows).</p>
Full article ">
14 pages, 3424 KiB  
Article
Directorial Editing: A Hybrid Deep-Learning Approach to Content-Aware Image Retargeting and Resizing
by Elliot Dickman and Paul Diefenbach
Electronics 2024, 13(22), 4459; https://doi.org/10.3390/electronics13224459 - 14 Nov 2024
Viewed by 576
Abstract
Image retargeting is a common computer graphics task which involves manipulating the size or aspect ratio of an image. This task often presents a challenge to the artist or user, because manipulating the size of an image necessitates some degree of data loss [...] Read more.
Image retargeting is a common computer graphics task which involves manipulating the size or aspect ratio of an image. This task often presents a challenge to the artist or user, because manipulating the size of an image necessitates some degree of data loss as pixels need to be removed to accommodate a different image size. We present an image retargeting framework which implements a confidence map generated by a segmentation model for content-aware resizing, allowing users to specify which subjects in an image to preserve using natural language prompts much like the role of an art director conversing with their artist. Using computer vision models to detect object positions also provides additional control over the composition of the retargeted image at various points in the image-processing pipeline. This object-based approach to energy map augmentation is incredibly flexible, because only minor adjustments to the processing of the energy maps can provide a significant degree of control over where seams—paths of pixels through the image—are removed, and how seam removal is prioritized in different sections of the image. It also provides additional control with techniques for object and background separation and recomposition. This research explores how several different types of deep-learning models can be integrated into this pipeline in order to easily make these decisions, and provide different retargeting results on the same image based on user input and compositional considerations. Because this is a framework based on existing machine-learning models, this approach will benefit from advancements in the rapidly developing fields of computer vision and large language models and can be extended for further natural language directorial controls over images. Full article
(This article belongs to the Special Issue Deep Learning in Image Processing and Computer Vision)
Show Figures

Figure 1

Figure 1
<p>Original image (<b>a</b>), Retargeted image with seam carving (<b>b</b>), scaling (<b>c</b>), and cropping (<b>d</b>) [<a href="#B4-electronics-13-04459" class="html-bibr">4</a>].</p>
Full article ">Figure 2
<p>Distortions from naïve seam carving, with noticeable distortion in the ibex and the diagonal lines of the vehicle.</p>
Full article ">Figure 3
<p>Original image (<b>a</b>) with confidence maps for “flower” (<b>b</b>), “houseplant” (<b>c</b>), and “man” (<b>d</b>) classes using the automated method.</p>
Full article ">Figure 4
<p>Original image (<b>a</b>) with NLP confidence map using prompt “Woman and metal bar. The woman is more important.” (<b>b</b>), compared to the automated segmentation output map (<b>c</b>).</p>
Full article ">Figure 5
<p>Original image (<b>a</b>) and the combined <math display="inline"><semantics> <mrow> <mi>e</mi> <mo>_</mo> <mi>a</mi> <mi>u</mi> <mi>g</mi> </mrow> </semantics></math> energy map using the NLP segmentation output shown in <a href="#electronics-13-04459-f004" class="html-fig">Figure 4</a> (<b>b</b>).</p>
Full article ">Figure 6
<p>Retargeting an image with clearly defined object placement demonstrates this method well. Original image (<b>a</b>), the segmentation output overlaid with red lines denoting the edges of each object’s predicted bounding box (<b>b</b>), and the image retargeted using the above algorithm to maintain the relative positioning of each object (<b>c</b>). While only the horizontal bounds of objects were calculated here because only vertical seams were removed, the full bounding box would be required for both vertical and horizontal retargeting.</p>
Full article ">Figure 7
<p>Original image (<b>a</b>), retargeted image using naïve seam carving (<b>b</b>), our proposed hybrid energy method (<b>c</b>), and retargeting the subjects separately from the rest of the image using the proposed layered method (<b>d</b>). The layered method uses SAM and CLIPSeg to segment the layers, and DALL-E is used to inpaint the missing areas of overlap. The layered method results in significantly less distortion to the background, and better preserves the primary subjects in the image.</p>
Full article ">Figure 8
<p>Original image (<b>a</b>); results of naïve seam carving (<b>b</b>); results of our method using the automatic (<b>c</b>) and manual (<b>d</b>) confidence maps as seen earlier in <a href="#electronics-13-04459-f004" class="html-fig">Figure 4</a>.</p>
Full article ">Figure 9
<p>Our proposed method (<b>a</b>) compared with cropping (<b>b</b>) and scaling (<b>c</b>) to the same aspect ratio.</p>
Full article ">Figure 10
<p>Original image (<b>a</b>) with four variations demonstrating how different prompts can yield vastly different retargeting results. Prompts: “People and flowers” (<b>b</b>), “People and doorway” (<b>c</b>), “People and doorway, but the doorway is more important” (<b>d</b>), “Only small flowers in flowerpots” (<b>e</b>).</p>
Full article ">
17 pages, 2194 KiB  
Article
Long Terminal Repeats of Gammaretroviruses Retain Stable Expression after Integration Retargeting
by Dalibor Miklík, Martina Slavková, Dana Kučerová, Chahrazed Mekadim, Jakub Mrázek and Jiří Hejnar
Viruses 2024, 16(10), 1518; https://doi.org/10.3390/v16101518 - 25 Sep 2024
Cited by 1 | Viewed by 881
Abstract
Retroviruses integrate into the genomes of infected host cells to form proviruses, a genetic platform for stable viral gene expression. Epigenetic silencing can, however, hamper proviral transcriptional activity. As gammaretroviruses (γRVs) preferentially integrate into active promoter and enhancer sites, the high transcriptional activity [...] Read more.
Retroviruses integrate into the genomes of infected host cells to form proviruses, a genetic platform for stable viral gene expression. Epigenetic silencing can, however, hamper proviral transcriptional activity. As gammaretroviruses (γRVs) preferentially integrate into active promoter and enhancer sites, the high transcriptional activity of γRVs can be attributed to this integration preference. In addition, long terminal repeats (LTRs) of some γRVs were shown to act as potent promoters by themselves. Here, we investigate the capacity of different γRV LTRs to drive stable expression within a non-preferred epigenomic environment in the context of diverse retroviral vectors. We demonstrate that different γRV LTRs are either rapidly silenced or remain active for long periods of time with a predominantly active proviral population under normal and retargeted integration. As an alternative to the established γRV systems, the feline leukemia virus and koala retrovirus LTRs are able to drive stable, albeit intensity-diverse, transgene expression. Overall, we show that despite the occurrence of rapid silencing events, most γRV LTRs can drive stable expression outside of their preferred chromatin landscape after retrovirus integrations. Full article
(This article belongs to the Section General Virology)
Show Figures

Figure 1

Figure 1
<p>MLV is stably expressed after integration retargeting. Comparison of expression stability of MLV-derived vectors after integration retargeting. (<b>A</b>) A schematic depiction of the LTR-dGFP-LTR vectors and integrase (IN) variants used in the experiment. (<b>B</b>) A dot plot representing the flow cytometry measurement of K562 cells transduced by MLV-derived vectors 3 dpi. Numbers correspond to the percentage of GFP+ cells. For each vector variant, 2400 live cells were selected to construct the dot plot. (<b>C</b>) Fold change in the fraction of GFP+ cells in the transduced population during 30 days of culture. The <span class="html-italic">y</span>-axis is on a log<sub>2</sub> scale. Timepoint 3 dpi represents the data in panel (<b>B</b>). (<b>D</b>) Fraction of GFP+ cells after the cultivation of bulk populations sorted for GFP expression at 3 dpi. Panels (<b>E</b>,<b>F</b>) demonstrate characteristics of clonal populations expanded from single cells sorted for GFP expression at 3 dpi. The fraction of cells expressing GFP (<b>E</b>) and the mean fluorescence intensity (MFI) (<b>F</b>) were measured at 30 dpi. In each category, 201 clonal populations were characterized. Schemes in panels (<b>C</b>–<b>E</b>) show a time course of experiments, with flow cytometry (F) or FACS sorting (S) performed at a given dpi.</p>
Full article ">Figure 2
<p>Integration site (IS) profile of active MLV proviruses after retargeting. IS distribution analysis of MLV IN<sup>wt</sup> and Bin vectors in K562 cells expressing GFP. Panels (<b>A</b>,<b>B</b>) represent an analysis of IS distances to defined chromatin segments. (<b>A</b>) Dot plot representing the results of the “Impact” effect-size analysis comparing IS of Bin<sup>CBX</sup> and IN<sup>wt</sup> vector usage on the distribution of ISs. Points represent individual segments grouped into categories differentiated by colors. CTDiff marks the change in the central tendency of the distribution. Shown are the names of the segments with Impact absolute value ≥ 0.5. (<b>B</b>) Plot showing the distribution of IS distances to the active transcription start site (Tss) and strong enhancer (Enh) chromatin segments. Each dot represents an individual IS, box plots represent medians and quartile range of the distance distributions. (<b>C</b>–<b>E</b>) Panels represent the targeting of chromatin A/B subcompartments and lamina-associated domains (LAD). (<b>C</b>) A bar plot representing a fraction of proviruses integrated into the subcompartments and LADs. Fold frequency changes in subcompartment targeting of W390A and CBX IN variants to wt IN. The <span class="html-italic">x</span>-axis is depicted in the log<sub>2</sub> scale. (<b>E</b>) Frequency of shuffled sites in the chromatin subcompartments representing a random targeting control. Each dot represents a shuffled site set prepared for each of the IN variant samples. The height of the bar represents the mean targeting frequency.</p>
Full article ">Figure 3
<p>Expression stability of non-MLV gammaretroviral vectors after retargeting. (<b>A</b>) Gammaretroviral vectors carrying LTRs from five different retroviruses were constructed. Viral stocks were produced with Gag-Pol variants, and expression of the proviruses in transduced cell population was observed for two weeks. (<b>B</b>) Expression of d2GFP by gammaretroviral vector-transduced cells at 3 dpi. For each sample, 10,000 cells are shown. GFP-positive cells are displayed in green. Box plots show the median and quartile range of GFP intensity for the GFP-positive population. Numbers specify the percentage of GFP-positive cells in the transduced population. Shown data represent experiment E4. Log<sub>10</sub> transformed GFP and FSC.A signal is used in a graph. (<b>C</b>) Bar plot showing the change in % of GFP-positive cells in time. The values are relative to the level of expression observed at 3 dpi of a particular experiment. (<b>D</b>) Ratio of % GFP-positive cells to a copy number (CN) of detected d2GFP-encoding genomes per 100 cells. The flow cytometry and DNA extraction were performed two weeks after transduction.</p>
Full article ">Figure 4
<p>Intensity and stability of αRV vector expression with γRV LTR as an internal promoter. (<b>A</b>) A schematic depiction of an αRV AS vector. The internal promoter was derived from the U3 promoter/enhancer part of the LTR of the studied γRVs. The bottom of the panel contains the scheme of the experiment, where GFP expression was followed every 3–4 days at 3–31 dpi. (<b>B</b>) Intensity of d2GFP expression in transduced K562 cells at 3 dpi. Cells inside the GFP+ gate are colored green. Box plot describes the intensity distribution of GFP+ cells. The numbers show the percentage of GFP+ cells of all alive cells. For each sample, 10,000 cells are shown. Samples are ordered by the median intensity of GFP+ cells. (<b>C</b>) Representation of the time-course experiment where the percentage of GFP+ cells in transduced populations was followed. Values on the <span class="html-italic">y</span>-axis show the percentage of GFP+ cells relative to the percentage of GFP+ cells observed at 3 dpi. Light lines and points show individual transduction experiments with divergent multiplicities of infection. Black-outlined points connected by black lines show the average of all experiments. (<b>D</b>) Copy number of proviruses (GFP copies) as per 100 genomic equivalents (200 copies of RPP30 reference target) measured by the droplet digital PCR (ddPCR). Proviral copy number was established from genomic DNA collected at 14 dpi in samples shown in panel (<b>B</b>). (<b>E</b>) Ratio of the percentage of GFP+ cells per proviral copy number per 100 genome equivalents. The value of 1 marks the point where all proviruses are expected to be active in expression. Points in (<b>D</b>,<b>E</b>) show values of technical duplicates.</p>
Full article ">
29 pages, 1556 KiB  
Review
The Immunotherapy of Acute Myeloid Leukemia: A Clinical Point of View
by Federico Mosna
Cancers 2024, 16(13), 2359; https://doi.org/10.3390/cancers16132359 - 27 Jun 2024
Viewed by 3368
Abstract
The potential of the immune system to eradicate leukemic cells has been consistently demonstrated by the Graft vs. Leukemia effect occurring after allo-HSCT and in the context of donor leukocyte infusions. Various immunotherapeutic approaches, ranging from the use of antibodies, antibody–drug conjugates, bispecific [...] Read more.
The potential of the immune system to eradicate leukemic cells has been consistently demonstrated by the Graft vs. Leukemia effect occurring after allo-HSCT and in the context of donor leukocyte infusions. Various immunotherapeutic approaches, ranging from the use of antibodies, antibody–drug conjugates, bispecific T-cell engagers, chimeric antigen receptor (CAR) T-cells, and therapeutic infusions of NK cells, are thus currently being tested with promising, yet conflicting, results. This review will concentrate on various types of immunotherapies in preclinical and clinical development, from the point of view of a clinical hematologist. The most promising therapies for clinical translation are the use of bispecific T-cell engagers and CAR-T cells aimed at lineage-restricted antigens, where overall responses (ORR) ranging from 20 to 40% can be achieved in a small series of heavily pretreated patients affected by refractory or relapsing leukemia. Toxicity consists mainly in the occurrence of cytokine-release syndrome, which is mostly manageable with step-up dosing, the early use of cytokine-blocking agents and corticosteroids, and myelosuppression. Various cytokine-enhanced natural killer products are also being tested, mainly as allogeneic off-the-shelf therapies, with a good tolerability profile and promising results (ORR: 20–37.5% in small trials). The in vivo activation of T lymphocytes and NK cells via the inhibition of their immune checkpoints also yielded interesting, yet limited, results (ORR: 33–59%) but with an increased risk of severe Graft vs. Host disease in transplanted patients. Therefore, there are still several hurdles to overcome before the widespread clinical use of these novel compounds. Full article
Show Figures

Figure 1

Figure 1
<p>Immunotherapy of acute myeloid leukemia. This figure shows a schematic representation of selected components and agents involved in the immunotherapy of acute myeloid leukemia (AML). Cells exerting immune activity against AML are shown on the left of the figure; opposite to them, immunomodulatory cells and immunosuppressive agents are shown on the right. Potential immunotherapeutic drugs are shown on top of the figure. Besides exposing antigen-derived peptides from MHC to cytotoxic T lymphocytes (CTL, in black) and natural killer cells (NK, in green), AML blasts (in red) actively express immunomodulatory molecules (e.g., indoleamine 2,3 dioxygenase—IDO) and immune checkpoint ligands to evade immune reactions (e.g., PD-L1, CD155/CD112, NKG2DL), as well as maintain sensitivity to potential immunotherapeutic agents (e.g., antibody–drug conjugates—ADCs, bispecific T-cell engagers—BiTEs, and dual-affinity retargeting antibodies—DARTs) by the expression of lineage-restricted antigens (e.g., CD33, CD123). CTL/CAR-T and NK/CAR-NK cells, together with antigen-presenting cells (APCs) are fundamental in activating and eliciting the immune response against AML; opposite to them, T-regulatory cells (T<sub>reg</sub>), mesenchymal stromal cells (MSCs), myeloid-derived suppressor cells (MDSCs), and Ttmor-associated macrophages (TAMs) modulate the immune response by means of contact-dependent as well as soluble factors (represented as yellow circles—soluble IDO (sIDO)—and starlets—immunomodulatory cytokines e.g., IL-10, TGF-β, IL-35). Following therapy, immunotoxins and immune checkpoint inhibitors (ICPI) also contribute to the formation of the milieu.</p>
Full article ">Figure 2
<p>Chimeric antigen receptor T-cells constructs and platforms. (<b>A</b>) Chimeric antigen receptor (CAR) constructs have evolved over time from a basic form that only contained a CD3ζ chain intracellular signal domain (1st generation) to more complex structures that include one (2nd generation) or two (3rd generation) costimulatory domains (most commonly CD28 or 4-1BB). More recent CARs contain interleukin-producing sections (4th generation, e.g., IL-12) or intracellular domains of cytokine receptors (5th generation, e.g., IL-2Rβ). Besides acquiring the CAR, engineered T lymphocytes usually maintain their previous T-cell receptor (TCR), depending on the specific design. Upon recognition by their CAR, CAR-T cells activate against the target cells (e.g., CD33+ AML) by inducing their apoptosis. (<b>B</b>) “Dual CAR” platforms can generate different combinations of CAR-Ts: in the <span class="html-italic">pooled CAR-T</span>, two different clones, each with its specific CAR, are generated; in the <span class="html-italic">compound CAR-T</span>, both CARs, each complete with a costimulatory and an activating domain, are expressed by the same cell; in the <span class="html-italic">split CAR-T</span>, two different CARs (a chimeric antigen receptor and a chimeric costimulatory receptor) are present on the same cell and linked differently to activating or costimulatory domains; finally, in the <span class="html-italic">tandem CAR-T</span>, two antigen-recognizing CARs are both linked to the same costimulatory and activating domains.</p>
Full article ">Figure 3
<p>Mechanisms of immune escape by acute myeloid leukemia cells. <b>The figure</b> shows a schematic representation of selected possible mechanisms of immune escape by acute myeloid leukemia (AML) cells. All these biological changes by AML cells are ultimately responsible for immune evasion and for inducing an exhaustion phenotype in both T lymphocytes and natural killer (NK) cells (not represented).</p>
Full article ">
21 pages, 29279 KiB  
Article
Neutral Facial Rigging from Limited Spatiotemporal Meshes
by Jing Hou, Dongdong Weng, Zhihe Zhao, Ying Li and Jixiang Zhou
Electronics 2024, 13(13), 2445; https://doi.org/10.3390/electronics13132445 - 21 Jun 2024
Viewed by 794
Abstract
Manual facial rigging is time-consuming. Traditional automatic rigging methods lack either 3D datasets or explainable semantic parameters, which makes it difficult to retarget a certain 3D expression to a new face. To address the problem, we automatically generate a large 3D dataset containing [...] Read more.
Manual facial rigging is time-consuming. Traditional automatic rigging methods lack either 3D datasets or explainable semantic parameters, which makes it difficult to retarget a certain 3D expression to a new face. To address the problem, we automatically generate a large 3D dataset containing semantic parameters, joint positions, and vertex positions from a limited number of spatiotemporal meshes. We establish an expression generator based on a multilayer perceptron with vertex constraints from the semantic parameters to the joint positions and establish an expression recognizer based on a generative adversarial structure from the joint positions to the semantic parameters. To enhance the accuracy of key facial area recognition, we add local vertex constraints for the eyes and lips, which are determined by the 3D masks computed by the proposed projection-searching algorithm. We testthe generation and recognition effects on a limited number of publicly available Metahuman meshes and self-collected meshes. Compared with existing methods, our generator has the shortest generation time of 14.78 ms and the smallest vertex relative mean square error of 1.57 × 10−3, while our recognizer has the highest accuracy of 92.92%. The ablation experiment verifies that the local constraints can improve the recognition accuracy by 3.02%. Compared with other 3D mask selection methods, the recognition accuracy is improved by 1.03%. In addition, our method shows robust results for meshes of different levels of detail, and the rig has more dimensions of semantic space. The source code will be made available if this paper is accepted for publication. Full article
(This article belongs to the Special Issue Neural Networks and Deep Learning in Computer Vision)
Show Figures

Figure 1

Figure 1
<p>Overview of our method: (<b>a</b>) dataset expansion, (<b>b</b>) structure of the proposed RigGenNet, and (<b>c</b>) structure of the proposed RigRecogNet.</p>
Full article ">Figure 2
<p>Explanation of joint rotation transformations. The fixed angles of joint <math display="inline"><semantics> <mi mathvariant="bold-italic">B</mi> </semantics></math> are represented by the positions of sub-joints <math display="inline"><semantics> <mrow> <msub> <mi mathvariant="bold-italic">B</mi> <mn>1</mn> </msub> <mo>,</mo> <msub> <mi mathvariant="bold-italic">B</mi> <mn>2</mn> </msub> <mo>,</mo> <msub> <mi mathvariant="bold-italic">B</mi> <mn>3</mn> </msub> </mrow> </semantics></math>.</p>
Full article ">Figure 3
<p>Illustration showing the selection of 3D masks; 3D meshes are projected onto the <math display="inline"><semantics> <mrow> <mi>Z</mi> <mi>O</mi> <mi>X</mi> </mrow> </semantics></math> plane, then the 3D vertices, which have corresponding 2D points that are less distant from the detected 2D points than a certain value, are selected as masks.</p>
Full article ">Figure 4
<p>Mask illustration at different levels of detail: (<b>a</b>) the detected 2D points [<a href="#B45-electronics-13-02445" class="html-bibr">45</a>], (<b>b</b>) the chosen 2D points, and (<b>c</b>) the 3D mask. The green parts are the adopted vertices with a mask value of 1, while the gray parts are the useless vertices with a mask value of 0.</p>
Full article ">Figure 5
<p>The absolute vertex error visualized on the generated meshes of different datasets: (<b>a</b>) ground truth, (<b>b</b>) blendshape-based rig [<a href="#B43-electronics-13-02445" class="html-bibr">43</a>,<a href="#B48-electronics-13-02445" class="html-bibr">48</a>], (<b>c</b>) CTBNET [<a href="#B31-electronics-13-02445" class="html-bibr">31</a>], (<b>d</b>) NFR [<a href="#B20-electronics-13-02445" class="html-bibr">20</a>], (<b>e</b>) SketchMetaFace [<a href="#B16-electronics-13-02445" class="html-bibr">16</a>], and (<b>f</b>) ours.</p>
Full article ">Figure 6
<p>The absolute vertex error visualized on the reconstructed mesh of different datasets. The reconstructed meshes are computed by inputting the recognized semantic parameters into RigGenNet. (<b>a</b>) Ground truth; (<b>b</b>) blendshape-based rig [<a href="#B43-electronics-13-02445" class="html-bibr">43</a>,<a href="#B48-electronics-13-02445" class="html-bibr">48</a>]; (<b>c</b>) FFNet [<a href="#B30-electronics-13-02445" class="html-bibr">30</a>]; (<b>d</b>) BTCNET [<a href="#B31-electronics-13-02445" class="html-bibr">31</a>]; (<b>e</b>) NFR [<a href="#B20-electronics-13-02445" class="html-bibr">20</a>]; (<b>f</b>) Shape Transformer [<a href="#B24-electronics-13-02445" class="html-bibr">24</a>]; (<b>g</b>) ours.</p>
Full article ">Figure 7
<p>The absolute vertex error visualized on the generated mesh and the UV mapping of different levels of detail. The generated meshes were computed by inputting the labeled semantic parameters into RigGenNet. (<b>a</b>) LOD6; (<b>b</b>) LOD3; (<b>c</b>) LOD0.</p>
Full article ">Figure 8
<p>The absolute vertex error visualized on the reconstructed mesh and the UV mapping of different levels of detail. The reconstructed meshes were computed by inputting the recognized semantic parameters into the RigGenNet. (<b>a</b>) Ground truth; (<b>b</b>) recognized semantic parameters; (<b>c</b>) reconstructed rig; (<b>d</b>) error heatmap drawn on the reconstructed mesh; (<b>e</b>) error heatmap drawn on the UV mapping.</p>
Full article ">Figure 9
<p>Unsmooth phenomena in the lips: (<b>a</b>) true expression and (<b>b</b>) the expression reenacted using our method.</p>
Full article ">
33 pages, 6045 KiB  
Article
A Display-Adaptive Pipeline for Dynamic Range Expansion of Standard Dynamic Range Video Content
by Gonzalo Luzardo, Asli Kumcu, Jan Aelterman, Hiep Luong, Daniel Ochoa and Wilfried Philips
Appl. Sci. 2024, 14(10), 4081; https://doi.org/10.3390/app14104081 - 11 May 2024
Cited by 1 | Viewed by 1154
Abstract
Recent advancements in high dynamic range (HDR) display technology have significantly enhanced the contrast ratios and peak brightness of modern displays. In the coming years, it is expected that HDR televisions capable of delivering significantly higher brightness and, therefore, contrast levels than today’s [...] Read more.
Recent advancements in high dynamic range (HDR) display technology have significantly enhanced the contrast ratios and peak brightness of modern displays. In the coming years, it is expected that HDR televisions capable of delivering significantly higher brightness and, therefore, contrast levels than today’s models will become increasingly accessible and affordable to consumers. While HDR technology has gained prominence over the past few years, low dynamic range (LDR) content is still consumed due to a substantial volume of historical multimedia content being recorded and preserved in LDR. Although the amount of HDR content will continue to increase as HDR becomes more prevalent, a large portion of multimedia content currently remains in LDR. In addition, it is worth noting that although the HDR standard supports multimedia content with luminance levels up to 10,000 cd/m2 (a standard measure of brightness), most HDR content is typically limited to a maximum brightness of around 1000 cd/m2. This limitation aligns with the current capabilities of consumer HDR TVs but is a factor approximately five times brighter than current LDR TVs. To accurately present LDR content on a HDR display, it is processed through a dynamic range expansion process known as inverse tone mapping (iTM). This LDR to HDR conversion faces many challenges, including the inducement of noise artifacts, false contours, loss of details, desaturated colors, and temporal inconsistencies. This paper introduces complete inverse tone mapping, artifact suppression, and a highlight enhancement pipeline for video sequences designed to address these challenges. Our LDR-to-HDR technique is capable of adapting to the peak brightness of different displays, creating HDR video sequences with a peak luminance of up to 6000 cd/m2. Furthermore, this paper presents the results of comprehensive objective and subjective experiments to evaluate the effectiveness of the proposed pipeline, focusing on two primary aspects: real-time operation capability and the quality of the HDR video output. Our findings indicate that our pipeline enables real-time processing of Full HD (FHD) video (1920 × 1080 pixels), even on hardware that has not been optimized for this task. Furthermore, we found that when applied to existing HDR content, typically capped at a brightness of 1000 cd/m2, our pipeline notably enhances its perceived quality when displayed on a screen that can reach higher peak luminances. Full article
(This article belongs to the Special Issue Intelligent Systems: Methods and Implementation)
Show Figures

Figure 1

Figure 1
<p>Comparison between LED and OLED display technology.</p>
Full article ">Figure 2
<p>Flowchart of the proposed pipeline for inverse tone mapping. The dynamic range of an 8-bit LDR frame is expanded into a 16-bit HDR frame with a peak luminance of 6000 cd/m<sup>2</sup>.</p>
Full article ">Figure 3
<p>Example of artifact suppression using the fast-guided filter on an image with visible artifacts. The parameters used were <math display="inline"><semantics> <mrow> <mi>r</mi> <mo>=</mo> <mn>32</mn> </mrow> </semantics></math>, <math display="inline"><semantics> <mrow> <mi>ϵ</mi> <mo>=</mo> <mn>0</mn> <mo>.</mo> <msup> <mn>1</mn> <mn>2</mn> </msup> </mrow> </semantics></math>, and <math display="inline"><semantics> <mrow> <mi>s</mi> <mo>=</mo> <mn>4</mn> </mrow> </semantics></math>. In the zoomed region, artifacts in the lapel are suppressed while fine details such as seams and wrinkles in the actor’s face are largely preserved. (Image from the movie trailer “Dawn of the Planet of the Apes”: WETA/Twentieth Century Fox Film Corporation).</p>
Full article ">Figure 4
<p>Dynamic range expansion based on mid-level tone mapping. The overall perception of brightness of the expanded luminance (<math display="inline"><semantics> <msub> <mi>L</mi> <mi mathvariant="normal">w</mi> </msub> </semantics></math>) can be modified by fixing the mid-level in parameter (<math display="inline"><semantics> <msub> <mi>m</mi> <mi mathvariant="normal">i</mi> </msub> </semantics></math>) and varying the mid-level out (<math display="inline"><semantics> <msub> <mi>m</mi> <mi mathvariant="normal">o</mi> </msub> </semantics></math>). Higher values of <math display="inline"><semantics> <msub> <mi>m</mi> <mi mathvariant="normal">o</mi> </msub> </semantics></math> (right side of the image) will make <math display="inline"><semantics> <msub> <mi>L</mi> <mi mathvariant="normal">w</mi> </msub> </semantics></math> brighter, and lower values (left part of the image) will make it darker. Parameters <span class="html-italic">d</span> and <span class="html-italic">a</span> can be used to fine-tune the results, increasing the overall contrast and appearance of the highlights, respectively.</p>
Full article ">Figure 5
<p>False-contour removal method based on iterative projection onto convex sets (POCS). In this example, the pixel value in <math display="inline"><semantics> <msub> <mi>X</mi> <mi>i</mi> </msub> </semantics></math> is filtered using a low-pass filter (first projection operator) and clamped between the quantization boundaries (second projection operator) iteratively.</p>
Full article ">Figure 6
<p>Binary and expansion maps obtained from two different LDR images. The expansion map is used to enhance the highlights of the HDR expanded luminance (<math display="inline"><semantics> <msub> <mi>L</mi> <mi mathvariant="normal">w</mi> </msub> </semantics></math>). (The image on the right was obtained from the movie “Star Wars: Episode III Revenge of the Sith”, property of Lucasfilm).</p>
Full article ">Figure 7
<p>Example of how highlights from the expanded luminance have been enhanced using expansion maps. False-color images are shown below each image for better visualization of the results.</p>
Full article ">Figure 8
<p>Results of the experiment to investigate the most adequate color saturation enhancement parameter value (<span class="html-italic">s</span>) used in Mantiuk’s formulation. Each dot represents a particular scene. The horizontal axis shows the mid-level out parameter (<math display="inline"><semantics> <msub> <mi>m</mi> <mi mathvariant="normal">o</mi> </msub> </semantics></math>) decided by our proposed dynamic range expansion function for that scene. The vertical axis shows the color saturation value (<span class="html-italic">s</span>) an expert user selected as most desirable. Green dots indicate scenes where the expert deemed it unnecessary to increase saturation, while black dots are those where the expert preferred increased saturation.</p>
Full article ">Figure 9
<p>A preview of video sequences utilized in the objective evaluation, obtained from the xDR Dataset.</p>
Full article ">Figure 10
<p>Objective quality scores for HDR video sequences produced by our proposed pipeline and two state-of-the-art methods. Quality scores were obtained from the dynamic range video quality metric (HDRVQM) using the HDR ground truth from the test dataset as a reference. HDRVQM computes one quality score for the entire sequence. Higher values indicate better quality.</p>
Full article ">Figure 11
<p>Sample frames illustrating artifacts in highly saturated regions, with visible distortions generated by the SR-ITM and FMNet methods. These artifacts become more noticeable when the video sequences are displayed on the SIM2 HDR screen. Images have been tone-mapped for presentation.</p>
Full article ">Figure 12
<p>The setup used for the evaluation of the proposed pipeline. The LDR and inverse tone-mapped HDR video sequences were randomly placed on the left (video 1) and on the right (video 2) on the HDR screen in each trial. LDR video sequences were linearized and adjusted to simulate the same appearance when it is displayed on an LDR screen with a limited peak brightness of 250 cd/m<sup>2</sup>.</p>
Full article ">Figure 13
<p>Preview of some video sequences employed in our LDR-to-HDR conversion evaluation experiment sourced from publicly accessible video documentaries and trailers. All video sequences have a resolution of <math display="inline"><semantics> <mrow> <mn>1920</mn> <mo>×</mo> <mn>1080</mn> </mrow> </semantics></math> pixels and 24 fps. (The original producers of the video sequences retain all rights pertaining to the frames shown in this figure).</p>
Full article ">Figure 14
<p>Results of our subjective experiment, comparing user preferences for HDR images generated through our proposed pipeline against the LDR input video sequences. The vertical bars indicate the frequency of participants’ preferences, showing whether they favored one type of video sequence over the other or found both sequences to be of equal quality.</p>
Full article ">Figure 15
<p>Flowchart of the proposed pipeline for HDR-to-xHDR conversion. A 1000 cd/m<sup>2</sup> HDR image is up-converted to extended high dynamic range (xHDR) with a peak luminance up to 6000 cd/m<sup>2</sup>.</p>
Full article ">Figure 16
<p>HDR-to-xHDR conversion: Frames obtained from our test HDR video sequences and the expansion maps computed (on the right). The HDR images were tone-mapped for presentation.</p>
Full article ">Figure 17
<p>Preview of some HDR video sequences employed in our HDR-to-xHDR conversion evaluation experiment, sourced from publicly available HDR video content. All video sequences were down-scaled to <math display="inline"><semantics> <mrow> <mn>1920</mn> <mo>×</mo> <mn>1080</mn> </mrow> </semantics></math> to fit the resolution of the HDR display used in our experiments. (The original producers of the video sequences retain all rights pertaining to the frames shown in this figure).</p>
Full article ">Figure 18
<p>Results of our subjective experiment with numerical data representation for clarity. User preferences are indicated as 0 for HDR, 1 for equivalent (no preference), and 2 for xHDR, facilitating an easier understanding of variability across video sequences. The graph’s bars depict the interquartile range employed to exclude potential outliers in user responses.</p>
Full article ">Figure 19
<p>Results of our subjective experiment, comparing user preferences for xHDR images generated through our proposed HDR to xHDR pipeline against the original HDR video sequences. The vertical bars indicate the frequency of participants’ preferences, showing whether they favored one type of video sequence over the other or found both sequences to be of equal quality.</p>
Full article ">
17 pages, 1614 KiB  
Article
Decrease in Heparan Sulphate Binding in Tropism-Retargeted Oncolytic Herpes Simplex Virus (ReHV) Delays Blood Clearance and Improves Systemic Anticancer Efficacy
by Andrea Vannini, Federico Parenti, Cristina Forghieri, Gaia Vannini, Catia Barboni, Anna Zaghini, Tatiana Gianni and Gabriella Campadelli-Fiume
Cancers 2024, 16(6), 1143; https://doi.org/10.3390/cancers16061143 - 13 Mar 2024
Viewed by 1354
Abstract
The role of the interaction with cell-surface glycosaminoglycans (GAGs) during in vivo HSV infection is currently unknown. The rationale of the current investigation was to improve the anticancer efficacy of systemically administered retargeted oHSVs (ReHVs) by decreasing their binding to GAGs, including those [...] Read more.
The role of the interaction with cell-surface glycosaminoglycans (GAGs) during in vivo HSV infection is currently unknown. The rationale of the current investigation was to improve the anticancer efficacy of systemically administered retargeted oHSVs (ReHVs) by decreasing their binding to GAGs, including those of endothelial cells, blood cells, and off-tumor tissues. As a proof-of-principle approach, we deleted seven amino acids critical for interacting with GAGs from the glycoprotein C (gC) of R-337 ReHV. The modification in the resulting R-399 recombinant prolonged the half-life in the blood of systemically administered R-399 and enhanced its biodistribution to tumor-positive lungs and to the tumor-negative liver. Ultimately, it greatly increased the R-399 efficacy against metastatic-like lung tumors upon IV administration but not against subcutaneous tumors upon IT administration. These results provide evidence that the increased efficacy seen upon R-399 systemic administration correlated with the slower clearance from the circulation. To our knowledge, this is the first in vivo evidence that the partial impairment of the gC interaction with GAGs resulted in a prolonged half-life of circulating ReHV, an increase in the amount of ReHV taken up by tissues and tumors, and, ultimately, an enhanced anticancer efficacy of systemically administered ReHV. Full article
(This article belongs to the Collection Advances and Future Prospects in Oncolytic Virus Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Adsorption of R-399 ReHV and of the parental R-337 to in vitro cultured cells. (<b>A</b>) Schematic representation of the genome of R-337 and its derivative R-399 that harbors a partial deletion of the heparan–sulfate-binding domain in the glycoprotein C (gC) gene. Indicated are the genetic loci of gC (wild type in R-337 and modified in R-399), gB with GCN4 insertion, gD with modifications for the detargeting from HSV-1 natural receptors Herpesvirus Entry Mediator (HVEM) and nectin1 and the retargeting to HER2, the insertion of murine interleukin 12 (mIL-12) gene in the Unique Short 1 (US1) and US2 intergenic locus, and of enhanced green fluorescent Protein (EGFP) in the Unique Long 37 (UL37) and UL38 intergenic locus. (<b>B</b>) Inhibition of virus adsorption to SK-OV-3 cells by low-molecular-weight (LMW) heparin, a heparan sulphate (HS) competitor. R-399 and R-337 viruses were pre-incubated with LMW heparin at the indicated concentrations for 1 h at 4 °C; the preincubated virions were then added to SK-OV-3 cells in the presence of the same amounts of competitor to allow virus adsorption. After the removal of non-adsorbed virus, cells were shifted to 37 °C to allow virus penetration and plaque formation. The number of plaques is expressed as percentage relative to the number of plaques in the heparin-untreated sample. The dashed line indicates the IC<sub>50</sub> value for R-337. (<b>C</b>) Dose-dependent inhibition curve of R-399 or R-337 adsorption to HEp-2 pre-treated with Heparinase I to reduce the overall amount of cell surface HSs and CSs. Triplicate monolayers of HEp-2 cells were pre-treated with Heparinase I at the indicated final concentrations for 1 h at 37 °C. Cells were then put on ice; viral inocula were added to allow 1 h of adsorption at 4 °C. After inoculum removal and rinsing, the viral particles adsorbed to cells were quantified by qPCR by means of a probe annealing to the HSV DNA polymerase gene and expressed as percentage viral genome copies (GC), relative to the GC adsorbed to the heparinase-untreated control. (<b>B</b>,<b>C</b>) Each point (symbol) is the mean of triplicate samples ± SD. Statistical analysis was performed with the unpaired parametric <span class="html-italic">t</span>-test (two-tailed) on the values of area under the curve calculated for each biological replicate (normal distribution, equality of variances from Shapiro–Wilk test and F-test, respectively); results of the tests are reported in the graphs: * = <span class="html-italic">p</span>-value &lt; 0.05; **** = <span class="html-italic">p</span>-value &lt; 0.0001. Color codes: R-337 in blue, R-399 in orange.</p>
Full article ">Figure 2
<p>In vitro characterisation of replicative ability of R-399. (<b>A</b>) Time course of R-337 and R-399 replication in SK-OV-3 and CT26-HER2 cells. Triplicate monolayers of the indicated cells were infected at an input multiplicity of infection of 0.1 PFU/cell, as titrated in the respective cell line and harvested at 24, 48, and 72 h after infection. Progeny virus was titrated in triplicate in SK-OV-3 cells. Each column represents the mean titer ± S.D. (<b>B</b>) Efficiency of R-337 or R-399 infection (plating efficiency) in SK-OV-3 and CT26-HER2 cell lines. For each virus, a suspension containing approximately 5 × 10<sup>8</sup> PFU/mL, and serial 1:10 dilutions thereof, were plated on triplicate monolayers of SK-OV-3 or CT26-HER2 cells; the number of plaques scored in the two different cell lines after 5 d is reported. Each column shows the mean number of plaques ± S.D. (<b>C</b>) Mean size of plaques formed by R-337 or R-399 in SK-OV-3 or CT26-HER2 cell monolayers. Samples were the ones described in Panel B. At the end of the experiment, 20 plaques for each virus were subjected to plaque-size determination by means of Nis Elements–AR–Imaging Software; plaque size was expressed as µm<sup>2</sup>. Each column represents the mean size of plaques in the indicated sample ± S.D. (<b>D</b>) Time course of cytotoxic effect exerted by R-337 or R-399 in SK-OV-3 and CT26-HER2 cells. For each time point, quadruplicate monolayers were infected at 0.1 PFU/cell. Cytotoxicity was determined by alamar blue viability and expressed as the percentage of non-viable cells relative to uninfected viable cells. Each column represents the mean of two independent experiments ± S.D. (<b>E</b>) Secretion of mIL-12 by SK-OV-3 and CT26-HER2 cells infected with R-337 or R-399 recombinants. mIL-12 was quantified by ELISA with the aid of the mouse ELISA kit, relative to a standard curve. Each column represents the mean of three independent experiments where each sample was run in duplicate ± S.D. Color codes: as in <a href="#cancers-16-01143-f001" class="html-fig">Figure 1</a>.</p>
Full article ">Figure 3
<p>Clearance and biodistribution of systemically administered R-399 and parental R-337. (<b>A</b>) Kinetics of R-399 and R-337 blood clearance. huHER2-trangenic mice (herein BALB/c-TG) (<span class="html-italic">n</span> = 10) mice were administered via an IV injection with 1 × 10<sup>6</sup> CT26-HER2 cells to establish metastatic-like lung tumors and, 14 d later, they received a single IV injection of R-337 (<span class="html-italic">n</span> = 5) or R-399 (<span class="html-italic">n</span> = 5) (1 × 10<sup>7</sup> PFU/mouse). Blood samples were collected 5, 30, and 60 min after injection; infectious virus in the blood samples was titrated in SK-OV-3 cells and expressed as PFU/100 µL blood. (<b>B</b>) Biodistribution to indicated tissues of R-399 or R-337 were expressed as viral genome copies (GCs)/100 ng of tissues DNA. Lung, liver, and brain samples (<span class="html-italic">n</span> = 4 for each virus) were collected 60 min after virus injection and homogenized, and the total DNA was purified as detailed in <a href="#sec2-cancers-16-01143" class="html-sec">Section 2</a>. The content in viral GCs was quantified by qPCR using a probe for HSV DNA polymerase and a standard curve to interpolate the results. Data were expressed as GCs/100 ng of DNA. (<b>A</b>,<b>B</b>) Symbols and columns indicate the mean values from the indicated number of mice ± SD. Statistical analysis was performed (<b>A</b>) by means of the unpaired nonparametric two-tailed Mann–Whitney test (non-normal distribution from Shapiro–Wilk test) for each time point, and (<b>B</b>) by means of the unpaired parametric two-tailed <span class="html-italic">t</span>-test with Welch’s correction for each organ (normal distribution, non-equal variances from Shapiro–Wilk test and F-test, respectively). Results of the statistical analyses are reported in the graphs: * = <span class="html-italic">p</span>-value &lt; 0.05; ** = <span class="html-italic">p</span>-value &lt; 0.01; *** = <span class="html-italic">p</span>-value &lt; 0.001; “ns” = no significant difference. Color codes: as in <a href="#cancers-16-01143-f001" class="html-fig">Figure 1</a>.</p>
Full article ">Figure 4
<p>Efficacy of R-399 and R-337 monotherapy on the growth of multiple metastatic-like lung CT26-HER2 tumors. (<b>A</b>) Treatment scheme with IV virus injections to treat multiple metastatic-like lung tumors. 3 × 10<sup>5</sup> CT26-HER2 cells were administered via an IV injection in BALB/c-TG mice to establish metastatic lung tumors. Eleven and 15 days later, mice received two IV injections of R-337 or R-399 (4 × 10<sup>7</sup> PFU/dose). Mice were sacrificed 25 days after tumor cell implantation; lungs were harvested, perfused with the staining solution, and fixed in Fekete’s solution. Lung metastatic nodules were counted under a microscope. (<b>B</b>) The number of tumor nodules scored in each mouse at lung surface. (<b>C</b>,<b>D</b>) Immune responses to CT26-wt and CT26-HER2 tumor cells in splenocytes (<b>C</b>) were detected as IFNγ secretion upon co-culture, and in sera (<b>D</b>), they were detected as mean fluorescence intensity (MFI) reactivity to the indicated cells. (<b>B</b>–<b>D</b>) Each symbol corresponds to an individual mouse; the horizontal line indicates the mean value. Vertical bars represent ± SD. Statistical analysis was performed with the nonparametric Kruskal–Wallis test with Dunn’s correction (non-normal distribution from Shapiro–Wilk test). Within each analysis, all possible comparisons between the study groups were considered; in (<b>C</b>,<b>D</b>), separate analyses were conducted for CT26-HER2 and CT26-wt samples. Results of the tests are reported in the graphs: * = <span class="html-italic">p</span>-value &lt; 0.05; ** = <span class="html-italic">p</span>-value &lt; 0.01; *** = <span class="html-italic">p</span>-value &lt; 0.001; “ns” = no significant difference. Mice treated with R-337 (<span class="html-italic">n</span> = 7) in blue, R-399 (<span class="html-italic">n</span> = 7) in orange, or vehicle (<span class="html-italic">n</span> = 6) in black.</p>
Full article ">Figure 5
<p>Efficacy of R-399 and R-337 monotherapy on the growth of subcutaneous CT26-HER2 tumors. (<b>A</b>) Treatment scheme with IT virus injections to treat the subcutaneous tumors. BALB/c-TG mice were implanted SC with 1 × 10<sup>6</sup> CT26-HER2 cells. Twenty days later, when the tumor volume averaged 70–100 mm<sup>3</sup>, mice received 3 IT injections of R-337 (<span class="html-italic">n</span> = 7), R-399 (<span class="html-italic">n</span> = 6), (3 × 10<sup>7</sup> PFU/dose), or vehicle (<span class="html-italic">n</span> = 7), administered at 3–4-day intervals. (<b>B</b>) Cumulative tumor growth curves for CT26-HER2 tumors. Each point represents the mean value relative to six-to-seven mice. (<b>C</b>) Tumor volumes at Day 35 after implantation. (<b>D</b>,<b>E</b>) Immune responses to CT26-wt and CT26-HER2 tumor cells in splenocytes (<b>D</b>) detected as IFNγ secretion upon co-culture, and in sera (<b>E</b>) detected as mean fluorescence intensity (MFI) reactivity to the indicated cells. (<b>C</b>–<b>E</b>) Each symbol corresponds to an individual mouse; the horizontal lines indicate the mean values; vertical bars indicate ± S.D. (<b>B</b>–<b>E</b>) Statistical analysis was performed with (<b>B</b>,<b>C</b>,<b>E</b>) the ordinary one-way ANOVA with Tukey’s correction (normal distribution, equality of variances from Shapiro–Wilk test and Bartlett’s test, respectively) or (<b>D</b>) the nonparametric Kruskal–Wallis test with Dunn’s correction (nonnormal distribution from Shapiro–Wilk test). (<b>B</b>–<b>E</b>) Within each analysis, all possible comparisons between the study groups were considered; in (<b>D</b>,<b>E</b>), separate analyses were conducted for CT26-HER2 and CT26-wt samples. (<b>B</b>) For each tumor-growth curve, the values of the area under the curve were calculated and compared in the statistical analysis. Results of the tests are reported in the graphs: * = <span class="html-italic">p</span>-value &lt; 0.05; ** = <span class="html-italic">p</span>-value &lt; 0.01; “ns” = no significant differences. Color code as in <a href="#cancers-16-01143-f004" class="html-fig">Figure 4</a>.</p>
Full article ">
16 pages, 4179 KiB  
Article
Elevated Levels of Mislocalised, Constitutive Ras Signalling Can Drive Quiescence by Uncoupling Cell-Cycle Regulation from Metabolic Homeostasis
by Elliot Piper-Brown, Fiona Dresel, Eman Badr and Campbell W. Gourlay
Biomolecules 2023, 13(11), 1619; https://doi.org/10.3390/biom13111619 - 6 Nov 2023
Cited by 1 | Viewed by 1652
Abstract
The small GTPase Ras plays an important role in connecting external and internal signalling cues to cell fate in eukaryotic cells. As such, the loss of RAS regulation, localisation, or expression level can drive changes in cell behaviour and fate. Post-translational modifications and [...] Read more.
The small GTPase Ras plays an important role in connecting external and internal signalling cues to cell fate in eukaryotic cells. As such, the loss of RAS regulation, localisation, or expression level can drive changes in cell behaviour and fate. Post-translational modifications and expression levels are crucial to ensure Ras localisation, regulation, function, and cell fate, exemplified by RAS mutations and gene duplications that are common in many cancers. Here, we reveal that excessive production of yeast Ras2, in which the phosphorylation-regulated serine at position 225 is replaced with alanine or glutamate, leads to its mislocalisation and constitutive activation. Rather than inducing cell death, as has been widely reported to be a consequence of constitutive Ras2 signalling in yeast, the overexpression of RAS2S225A or RAS2S225E alleles leads to slow growth, a loss of respiration, reduced stress response, and a state of quiescence. These effects are mediated via cAMP/PKA signalling and transcriptional changes, suggesting that quiescence is promoted by an uncoupling of cell-cycle regulation from metabolic homeostasis. The quiescent cell fate induced by the overexpression of RAS2S225A or RAS2S225E could be rescued by the deletion of CUP9, a suppressor of the dipeptide transporter Ptr2, or the addition of peptone, implying that a loss of metabolic control, or a failure to pass a metabolic checkpoint, is central to this altered cell fate. Our data suggest that the combination of an increased RAS2 copy number and a dominant active mutation that leads to its mislocalisation can result in growth arrest and add weight to the possibility that approaches to retarget RAS signalling could be employed to develop new therapies. Full article
Show Figures

Figure 1

Figure 1
<p>Cells expressing <span class="html-italic">RAS2</span>, <span class="html-italic">RAS2<sup>S225A</sup></span>, and <span class="html-italic">RAS2<sup>S225E</sup></span> from either a low-copy (CEN) or high-copy (2 µ) plasmid were grown in selective SD-URA minimal medium for 24 h at 30 °C before the total protein was extracted and probed by Western blotting with an anti-Ras2 and anti-Pgk1 antibody (loading control). Graphs represent the normalised relative band intensity from three biological replicates (<b>A</b>). Growth analysis of wild-type <span class="html-italic">S. cerevisiae</span> cells overexpressing <span class="html-italic">RAS2</span>, <span class="html-italic">RAS2<sup>S225A</sup></span>, <span class="html-italic">RAS2<sup>S225E</sup></span>, or an empty plasmid control (EV) from either a low-copy (CEN) (<b>B</b>) or high-copy (2 µ) (<b>C</b>) plasmid, representing an average of three biological replicates. Colony-forming unit assay of cells overexpressing <span class="html-italic">RAS2</span>, <span class="html-italic">RAS2<sup>S225A</sup></span>, <span class="html-italic">RAS2<sup>S225E</sup></span>, or an empty plasmid control grown in SD-URA medium (<b>D</b>). A one-way ANOVA using Dunnett’s multiple comparison test was used to determine statistical significance; * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001. Error bars represent standard deviations.</p>
Full article ">Figure 2
<p>Active Ras was visualised in cells overexpressing <span class="html-italic">RAS2</span>, <span class="html-italic">RAS2<sup>S225A</sup></span>, <span class="html-italic">RAS2<sup>S225E</sup></span>, or a control containing an empty plasmid using a 3xGFP-RBD probe during the logarithmic and stationary phases of growth. Cells were cultured in SD-URA/-LEU growth media. The experiment was repeated three times, and a representative dataset is shown. Scale bar—10 µm (<b>A</b>). Growth of wild-type cells overexpressing <span class="html-italic">RAS2</span>, <span class="html-italic">RAS2<sup>S225A</sup></span>, <span class="html-italic">RAS2<sup>S225E</sup></span>, or containing an empty plasmid control was carried out in SD-URA or SD-URA + 2 mM H<sub>2</sub>O<sub>2</sub> media; n = 3, error bars represent the standard deviation (<b>B</b>). Wild-type cells overexpressing <span class="html-italic">RAS2</span>, <span class="html-italic">RAS2<sup>S225A</sup></span>, <span class="html-italic">RAS2<sup>S225E</sup></span>, or containing an empty plasmid control were serially diluted from 2 × 10<sup>6</sup>/mL to 2 × 10<sup>3</sup>/mL and plated onto SD-URA plates supplemented with increasing concentrations of copper sulphate. This experiment was completed three times, and a representative result is shown (<b>C</b>).</p>
Full article ">Figure 3
<p>Wild-type cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> or an empty plasmid control were grown in SD-URA, and necrosis (PI uptake) (<b>A</b>) and ROS (DHE) (<b>B</b>) measurements were taken over a 12-day period of continuous incubation. The data displayed are the average of three technical repeats, and the error bars represent the standard deviation. A bar chart showing the routine, leak, ETS, and NMT O<sub>2</sub> flux values for wild-type cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> or <span class="html-italic">RAS2<sup>S225E</sup></span>, or containing an empty plasmid backbone control. The experiment was conducted in triplicate, and a representative dataset from one experiment is shown (<b>C</b>). A colony-forming unit assay of wild-type cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> or <span class="html-italic">RAS2<sup>S225E</sup></span>, or containing an empty plasmid control, grown in SD-URA medium for 24 h at 30 °C, was conducted and plated on either SD-URA or SD-URA + peptone (<b>D</b>). A colony-forming efficiency assay of wild-type cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span>, <span class="html-italic">RAS2<sup>S225E</sup></span>, or an empty plasmid backbone co-expressed with <span class="html-italic">PDE2</span> (<b>E</b>), or in a strain lacking <span class="html-italic">PDE2</span> (<b>F</b>). The data presented are the average of three biological replicates, and the error bars represent the standard deviation. A one-way ANOVA using Tukey’s multiple comparison test was used to determine statistical significance. Nonsignificant = NS, * = adjusted <span class="html-italic">p</span>-value ≤ 0.01, ** adjusted <span class="html-italic">p</span>-value of 0.05 and *** = adjusted <span class="html-italic">p</span> value ≤ 0.001.</p>
Full article ">Figure 4
<p>(<b>A</b>) Global gene expression changes in wild-type cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> when compared to a wild-type control. DESeq2 was used to compare gene expression in wild-type cells overexpressing RAS2S225A to a wild-type control; in total, there were 4133 significantly differentially expressed genes. Gene set cluster maps were created using the Cytoscape plugin, showing the most upregulated and downregulated gene sets, as determined by GSEA analysis, along with their cellular functions; circle size within a cluster represents the change in the expression level of a single gene. Representative gene sets shown to be upregulated (<b>B</b>) or downregulated (<b>C</b>) upon the overexpression of <span class="html-italic">RAS2<sup>S225A</sup></span> when compared to the wild-type control by GSEA. Vertical black lines represent individual genes in the significantly differentially expressed ranked gene list, from upregulated (<b>left</b>) to downregulated (<b>right</b>). An increase in the enrichment score is seen if there are many genes towards the beginning of the ranked list (upregulated) in the gene set.</p>
Full article ">Figure 5
<p>Growth analysis (<b>A</b>) and CFU assay (<b>B</b>) of wild-type and <span class="html-italic">Δcup9</span> cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> or <span class="html-italic">RAS2<sup>S225E</sup></span>, or containing an empty plasmid control. Routine, leak, ETS, and NMT O2 flux values for wild-type and <span class="html-italic">Δcup9</span> cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> or an empty plasmid backbone control. In each case, the data shown represent an average of three biological repeats, and the error bars indicate the standard deviation. A one-way ANOVA using Tukey’s multiple comparison test was used to determine statistical significance; Nonsignificant = NS, * <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001 (<b>C</b>). A representative GSEA gene set shown to be upregulated upon the overexpression of <span class="html-italic">RAS2<sup>S225A</sup></span> in a <span class="html-italic">Δcup9</span> background when compared to wild-type cells overexpressing <span class="html-italic">RAS2<sup>S225A</sup></span> (<b>D</b>).</p>
Full article ">Figure 6
<p>Model depicting a mechanism by which the overexpression of <span class="html-italic">RAS2<sup>S225A</sup></span> or <span class="html-italic">RAS2<sup>S225E</sup></span> promotes quiescence. The substitution of Ras2 at serine 225 for alanine or glutamate leads to constitutive activation at the nuclear envelope/ER when overexpressed. <span class="html-italic">RAS2<sup>S225A/E</sup></span>-driven cAMP/PKA signalling from the nuclear envelope/ER, in turn, promotes senescence under conditions of nutritional challenge by uncoupling the control of the expression of cell-cycle control for core metabolic processes. The addition of peptone or deletion of <span class="html-italic">CUP9</span>, which leads to the upregulation of a battery of metabolite transporters, can counteract quiescence driven by <span class="html-italic">RAS2<sup>S225A/E</sup></span> signalling, potentially by overcoming an essential metabolic checkpoint that is required to re-enter the cell cycle.</p>
Full article ">
20 pages, 4713 KiB  
Article
Efficacy of Systemically Administered Retargeted Oncolytic Herpes Simplex Viruses—Clearance and Biodistribution in Naïve and HSV-Preimmune Mice
by Andrea Vannini, Federico Parenti, Catia Barboni, Cristina Forghieri, Valerio Leoni, Mara Sanapo, Daniela Bressanin, Anna Zaghini, Gabriella Campadelli-Fiume and Tatiana Gianni
Cancers 2023, 15(16), 4042; https://doi.org/10.3390/cancers15164042 - 10 Aug 2023
Cited by 3 | Viewed by 1823
Abstract
We investigated the anticancer efficacy, blood clearance, and tissue biodistribution of systemically administered retargeted oncolytic herpes simplex viruses (ReHVs) in HSV-naïve and HSV-preimmunized (HSV-IMM) mice. Efficacy was tested against lung tumors formed upon intravenous administration of cancer cells, a model of metastatic disease, [...] Read more.
We investigated the anticancer efficacy, blood clearance, and tissue biodistribution of systemically administered retargeted oncolytic herpes simplex viruses (ReHVs) in HSV-naïve and HSV-preimmunized (HSV-IMM) mice. Efficacy was tested against lung tumors formed upon intravenous administration of cancer cells, a model of metastatic disease, and against subcutaneous distant tumors. In naïve mice, HER2- and hPSMA-retargeted viruses, both armed with mIL-12, were highly effective, even when administered to mice with well-developed tumors. Efficacy was higher for combination regimens with immune checkpoint inhibitors. A significant amount of infectious virus persisted in the blood for at least 1 h. Viral genomes, or fragments thereof, persisted in the blood and tissues for days. Remarkably, the only sites of viral replication were the lungs of tumor-positive mice and the subcutaneous tumors. No replication was detected in other tissues, strengthening the evidence of the high cancer specificity of ReHVs, a property that renders ReHVs suitable for systemic administration. In HSV-IMM mice, ReHVs administered at late times failed to exert anticancer efficacy, and the circulating virus was rapidly inactivated. Serum stability and in vivo whole blood stability assays highlighted neutralizing antibodies as the main factor in virus inactivation. Efforts to deplete mice of the neutralizing antibodies are ongoing. Full article
(This article belongs to the Collection Advances and Future Prospects in Oncolytic Virus Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Efficacy of IV monotherapy with R-337 on the growth of metastatic CT26-HER2 tumors. (<b>A</b>) Early treatment scheme with multiple IV injections. BALB/c-TG mice were injected IV with 1 × 10<sup>6</sup> CT26-HER2 cells to establish metastatic lung tumors and, 2 days later, received 4 IV injections of R-337 (1 × 10<sup>8</sup> PFU per injection) every 2–3 days. Mice were sacrificed on day 17 after tumor cell injection. At the time of sacrifice, lungs were harvested, perfused with the staining solution, fixed in Fekete’s solution, and lung metastatic nodules were counted under a microscope. (<b>B</b>) Representative images of stained lungs from groups treated with vehicle and R-337. Normal lung tissue is black, while metastases are white spots. (<b>C</b>) The number of tumor nodules counted on the surface of lungs from vehicle- and R-337-treated mice. (<b>D</b>,<b>E</b>) Immune response to CT26-wt and CT26-HER2 tumor cells in splenocytes (<b>D</b>) and sera (<b>E</b>) taken at the time of sacrifice from vehicle- and R-337-treated groups. (<b>F</b>) Scalar doses of R-337 in the early treatment schedule. BALB/c-TG mice were injected IV with 1 × 10<sup>6</sup> CT26-HER2 cells and, 3 days later, received 1 IV injections of R-337 (1 × 10<sup>7</sup> or 1 × 10<sup>8</sup> PFUs). Mice were sacrificed on day 17 after tumor cell injection. At the time of sacrifice, lungs were harvested and stained, and lung metastatic nodules were counted. (<b>G</b>) Representative images of the stained lungs of the vehicle-treated and R-337-treated groups. (<b>H</b>) The number of tumor nodules counted on the surface of the lungs. (<b>I</b>,<b>J</b>) Immune response to CT26-wt and CT26-HER2 tumor cells in splenocytes (<b>I</b>) and sera (<b>J</b>) collected at the time of sacrifice. Each circle corresponds to an individual mouse; the horizontal line indicates the mean value, and the vertical bars ± SD. (<b>C</b>–<b>E</b>,<b>H</b>–<b>J</b>) Statistical significance was calculated by t-test (<b>C</b>–<b>E</b>) or ANOVA test with Tukey’s correction (<b>H</b>–<b>J</b>) and expressed as * = <span class="html-italic">p</span>-value &lt; 0.05; ** = <span class="html-italic">p</span>-value &lt; 0.01; *** = <span class="html-italic">p</span>-value &lt; 0.001. Color code: mice treated with vehicle in black, R-337 (1 × 10<sup>8</sup> PFUs) in red, and R-337 (1 × 10<sup>7</sup> PFUs) in burgundy.</p>
Full article ">Figure 2
<p>Efficacy of IV therapy with R-337 in combination with anti-CTLA-4 antibody (αCTLA4) on the growth of metastatic CT26-HER2 tumors. (<b>A</b>) Scheme of early treatment with a single virus injection. BALB/c-TG mice were injected IV with 1 × 10<sup>6</sup> CT26-HER2 cells to establish metastatic lung tumors and, 3 days later, received 1 IV injection of R-337 (1 × 10<sup>7</sup> PFU per injection) and 3 i.p. injections of anti-CTLA-4 MAb (αCTLA4) at 3–4-day intervals. Mice were sacrificed on day 17 after tumor cell injection. At the time of sacrifice, lungs were harvested, perfused with the staining solution, fixed in Fekete’s solution, and lung metastatic nodules were counted under a microscope. (<b>B</b>) The number of tumor nodules counted on the surface of the lungs. (<b>C</b>,<b>D</b>) Immune response to CT26-wt and CT26-HER2 tumor cells in splenocytes (<b>C</b>) and sera (<b>D</b>) collected at the time of sacrifice. Each circle corresponds to an individual mouse; the horizontal line indicates the mean value, and the vertical bars ± SD. (<b>B</b>–<b>D</b>) Statistical significance was calculated by ANOVA test with Tukey’s correction and expressed as * = <span class="html-italic">p</span>-value &lt; 0.05; ** = <span class="html-italic">p</span>-value &lt; 0.01; *** = <span class="html-italic">p</span>-value &lt; 0.001; **** = <span class="html-italic">p</span>-value &lt; 0.0001. Color codes: mice treated with vehicle (black) or R-337 (red). Open circles indicate combination therapy with αCTLA4 antibody, while filled circles indicate no αCTLA4 antibody.</p>
Full article ">Figure 3
<p>Efficacy of IV therapy with R-405 in combination with anti-PD-1 antibody (αPD1) on the growth of subcutaneous LLC1-hPSMA tumors. (<b>A</b>) Scheme of treatments. C57BL/6 mice were implanted s.c. in the left flank with 1 × 10<sup>6</sup> LLC1-hPSMA cells. 9 days later, when the tumor volume averaged 70–100 mm<sup>3</sup>, mice received 2 IV injections of R-405 (1 × 10<sup>8</sup> PFU) or vehicle at 6-day intervals and 3 i.p. injections of anti-PD-1 MAb (αPD1) at 3–4-day intervals. (<b>B</b>) Replication of HSV in tumors and livers of R-405 treated and untreated mice. mRNA levels of viral glycoprotein gC were determined by qRT-PCR in tumor and liver samples. Normalized data on endogenous control Rpl13a were reported by setting the mean value of gC expression in tumors in the R-405-treated group to 1. (<b>C</b>,<b>D</b>) Tumor growth curves. The numbers shown in the panel indicate the number of mice that were completely cured of tumor (complete response, CR) or showed delayed/reduced tumor growth (partial response, PR). Mice were classified as PR when the tumor volume was 50% smaller than the mean tumor size in the vehicle group in at least 2 consecutive measurements. (<b>E</b>) Primary tumor volumes at day 19 after implantation. (<b>F</b>) Kaplan-Meier survival curves of the 2 groups of mice. (<b>G</b>) Immune response to LLC1-wt and LLC1-hPSMA tumor cells in splenocytes collected at the time of sacrifice. (<b>B</b>,<b>E</b>–<b>G</b>) Statistical significance was calculated by <span class="html-italic">t</span>-test (<b>B</b>,<b>E</b>,<b>G</b>) or Log-rank (Mantel-Cox) test (<b>F</b>) and expressed as * = <span class="html-italic">p</span>-value &lt; 0.05; *** = <span class="html-italic">p</span>-value &lt; 0.001. Color codes: mice treated with vehicle (black) or R-405 in combination with anti-PD-1 antibody (blue).</p>
Full article ">Figure 4
<p>Clearance and biodistribution of systemically administered R-337. (<b>A</b>) Treatment scheme. BALB/c-TG mice were injected IV with 1 × 10<sup>6</sup> CT26-HER2 cells to establish metastatic lung tumors (tumor-positive mice) or PBS (tumor-negative mice). 7 days later, both groups of mice received IV injections of R-337 (1 × 10<sup>7</sup> PFU) and were sacrificed after 10 and 30 min (both groups), 1, 6, and 24 h (tumor-negative mice only), 2, 5, and 7 days (both groups) and 21 days (tumor-negative mice only). At the time of sacrifice, blood and the indicated tissues were collected, homogenized, and DNA was purified. (<b>B</b>) Kinetic of R-337 biodistribution in the blood of tumor-positive and tumor-negative mice, measured as g.c and PFUs. Genome copies were quantified by qRT-PCR on the purified DNAs using a probe specific for HSV DNA polymerase and a standard curve to interpolate the results. Data were expressed as gc/100 µL of blood. The limit of quantification (LOQ, green dashed line) was estimated on the standard curve. Clearance of infectious virus in the bloodstream of tumor-negative mice was determined by titration of blood withdrawn from animals on SK-OV-3 cells and expressed as PFU/100 µL blood. (<b>C</b>–<b>H</b>) Kinetic of R-337 biodistribution (g.c.) in the lung (<b>C</b>), liver (<b>D</b>), spleen (<b>E</b>), kidney (<b>F</b>), heart (<b>G</b>), and brain (<b>H</b>) of tumor-positive and tumor-negative mice. See panel B for the details. (<b>I</b>) Replication of R-337 in the lungs of tumor-positive mice. mRNA levels of viral glycoprotein C were determined by qRT-PCR in lung samples of tumor-positive mice sacrificed 1, 2, 3, 4, or 5 days after IV injection of R-337. Normalized data on endogenous control Rpl13a were reported by setting the mean value of gC expression in R-337 mice sacrificed at day 1 to 1. (<b>J</b>) Replication of R-337 in the lungs of tumor-negative mice. (<b>K</b>,<b>L</b>) Replication of R-337 in the liver (<b>K</b>) and brain (<b>L</b>) of metastatic lung tumor-positive mice. (<b>M</b>) Detection of CT26-HER2 tumors in the indicated organs. Human HER2 was quantified by qRT-PCR on purified DNAs. Data were reported by setting the mean value of HER2 in the lungs of tumor-positive mice to 1. (<b>B</b>–<b>H</b>) Circles and squares indicate the mean values of 4–5 mice and vertical bars ± SD. (<b>I</b>–<b>M</b>) Each circle corresponds to an individual mouse; the horizontal line indicates the mean value, and the vertical bars ± SD. (<b>C</b>) Statistical significance was calculated by t-test and expressed as * = <span class="html-italic">p</span>-value &lt; 0.05. Color code: tumor-positive (red) and negative (black) mice treated with R-337. Untreated mice (blue).</p>
Full article ">Figure 4 Cont.
<p>Clearance and biodistribution of systemically administered R-337. (<b>A</b>) Treatment scheme. BALB/c-TG mice were injected IV with 1 × 10<sup>6</sup> CT26-HER2 cells to establish metastatic lung tumors (tumor-positive mice) or PBS (tumor-negative mice). 7 days later, both groups of mice received IV injections of R-337 (1 × 10<sup>7</sup> PFU) and were sacrificed after 10 and 30 min (both groups), 1, 6, and 24 h (tumor-negative mice only), 2, 5, and 7 days (both groups) and 21 days (tumor-negative mice only). At the time of sacrifice, blood and the indicated tissues were collected, homogenized, and DNA was purified. (<b>B</b>) Kinetic of R-337 biodistribution in the blood of tumor-positive and tumor-negative mice, measured as g.c and PFUs. Genome copies were quantified by qRT-PCR on the purified DNAs using a probe specific for HSV DNA polymerase and a standard curve to interpolate the results. Data were expressed as gc/100 µL of blood. The limit of quantification (LOQ, green dashed line) was estimated on the standard curve. Clearance of infectious virus in the bloodstream of tumor-negative mice was determined by titration of blood withdrawn from animals on SK-OV-3 cells and expressed as PFU/100 µL blood. (<b>C</b>–<b>H</b>) Kinetic of R-337 biodistribution (g.c.) in the lung (<b>C</b>), liver (<b>D</b>), spleen (<b>E</b>), kidney (<b>F</b>), heart (<b>G</b>), and brain (<b>H</b>) of tumor-positive and tumor-negative mice. See panel B for the details. (<b>I</b>) Replication of R-337 in the lungs of tumor-positive mice. mRNA levels of viral glycoprotein C were determined by qRT-PCR in lung samples of tumor-positive mice sacrificed 1, 2, 3, 4, or 5 days after IV injection of R-337. Normalized data on endogenous control Rpl13a were reported by setting the mean value of gC expression in R-337 mice sacrificed at day 1 to 1. (<b>J</b>) Replication of R-337 in the lungs of tumor-negative mice. (<b>K</b>,<b>L</b>) Replication of R-337 in the liver (<b>K</b>) and brain (<b>L</b>) of metastatic lung tumor-positive mice. (<b>M</b>) Detection of CT26-HER2 tumors in the indicated organs. Human HER2 was quantified by qRT-PCR on purified DNAs. Data were reported by setting the mean value of HER2 in the lungs of tumor-positive mice to 1. (<b>B</b>–<b>H</b>) Circles and squares indicate the mean values of 4–5 mice and vertical bars ± SD. (<b>I</b>–<b>M</b>) Each circle corresponds to an individual mouse; the horizontal line indicates the mean value, and the vertical bars ± SD. (<b>C</b>) Statistical significance was calculated by t-test and expressed as * = <span class="html-italic">p</span>-value &lt; 0.05. Color code: tumor-positive (red) and negative (black) mice treated with R-337. Untreated mice (blue).</p>
Full article ">Figure 5
<p>Efficacy of IV therapy with R-337 in combination with anti-CTLA-4 antibody (αCTLA4) on the growth of metastatic CT26-HER2 tumors in HSV-1-immune and naïve mice. (<b>A</b>) Late treatment scheme with multiple virus injections. BALB/c-TG mice were immunized twice with HSV-1 virions (<b>F</b>) 6 and 3 weeks before tumor cell injection. Immunized and naïve mice were injected IV with 3 × 10<sup>5</sup> CT26-HER2 cells to establish metastatic lung tumors, and blood samples were taken to verify HSV-1 immunization. 10 days later, mice received 2 IV injections of R-337 (1 × 10<sup>8</sup> PFUs per injection) every 4 days and 3 i.p. injections of αCTLA4 at 3-day intervals. Mice were sacrificed on day 21 after tumor cell injection. At the time of sacrifice, lungs were harvested, perfused with the staining solution, fixed in Fekete’s solution, and lung metastatic nodules were counted under a microscope. (<b>B</b>) Determination of immunization of mice to HSV-1. Sera obtained from the blood of mice (immunized and HSV-1-naïve), HSV-1-positive human patients and anti-HSV-1 MAb HD1 were diluted and reacted with RS cells previously infected with HSV-1, followed by incubation with anti-mouse peroxidase. The association of HSV-1-specific antibodies on the surface of infected cells was measured by the peroxidase reaction. (<b>C</b>) Determination of IC50 values of neutralizing antibodies in sera from mice (immunized and HSV-1 naïve), HSV-1-positive human patients, and the anti-HSV-1 MAb HD1. Sera and HD1 were serially diluted and used to neutralize infection with R-8102, an HSV-1-derived virus expressing the β-galactosidase reporter gene. The extent of infection in untreated (control) and serum-treated samples was determined by measuring the β-galactosidase activity. The curve of inhibition of infection versus dilution was used to calculate IC50 values. (<b>D</b>) The number of tumor nodules counted on the surface of the lungs. (<b>E</b>,<b>F</b>) Immune response to CT26-wt and CT26-HER2 tumor cells in splenocytes (<b>E</b>) and sera (<b>F</b>) collected at the time of sacrifice. Each circle corresponds to an individual mouse; the horizontal line indicates the mean value, and the vertical bars ± SD. (<b>B</b>–<b>D</b>) Statistical significance was calculated by t-test and expressed as ** = <span class="html-italic">p</span>-value &lt; 0.01; *** = <span class="html-italic">p</span>-value &lt; 0.001; **** = <span class="html-italic">p</span>-value &lt; 0.0001. Color codes: naïve mice treated with vehicle (black) or R-337 + αCTLA4 (red empty circle); HSV-1-immunized mice treated with vehicle (gray), αCTLA4 (gray empty circle), or R-337 + αCTLA4 combination (purple empty circle); HD1 (green); human sera (blue).</p>
Full article ">Figure 6
<p>Kinetic of R-337 biodistribution after IV injection in HSV-1-immune and naïve mice. (<b>A</b>) Treatment scheme. BALB/c-TG mice were immunized twice with HSV-1 virions (<b>F</b>) 6 and 3 weeks before tumor cell injection. Immunized and naïve mice were injected IV with 1 × 10<sup>6</sup> CT26-HER2 cells to establish metastatic lung tumors and, 12 days later, received IV injections of R-337 (1 × 10<sup>7</sup> PFU). Subgroups of mice were sacrificed after 10 min, 1, 2, 6 h, and 1 day. At the time of sacrifice, blood and the indicated tissues were collected, homogenized, and DNA was purified. (<b>B</b>) Kinetic of R-337 biodistribution (g.c.) in HSV-1-immunized and HSV-1-naïve mice. See <a href="#cancers-15-04042-f004" class="html-fig">Figure 4</a> for details (<b>C</b>) Clearance of infectious R-337 virus in the bloodstream of mice immunized and naïve to HSV-1. See <a href="#cancers-15-04042-f004" class="html-fig">Figure 4</a> for details. (<b>D</b>–<b>F</b>) Kinetic of R-337 biodistribution (g.c.) in the lung (<b>D</b>), liver (<b>E</b>), and brain (<b>F</b>) of HSV-1-immunized and HSV-1-naïve mice. Circles indicate the mean values of 4–5 mice and vertical bars ± SD. Color code: mice immunized with HSV-1 (purple) and naïve (red).</p>
Full article ">Figure 7
<p>Stability of R-337 in serum and in whole blood in vivo. (<b>A</b>) In vitro determination of the ability of HSV-1-immune and naïve sera to inactivate R-337 virions. Sera from the two types of mice were either heated to inactivate complement or treated with Sepharose Protein G to adsorb and reduce the antibodies, including NAbs. Treated and untreated sera were incubated with a fixed amount of R-337 for 30 min, and the amount of infectious virus was determined by titration on SK-OV-3 cells. Results are reported as the percentage of infectious viruses relative to the amount of PFU employed. (<b>B</b>) In vivo treatment scheme. BALB/c-TG mice were immunized twice with HSV-1 virions 6 and 3 weeks before R-337 injection. Naïve and HSV-IMM mice were treated twice with Cobra Venom Factor (CVF), 2 days and 1 h before R-337 injection. Other groups of immunized and naïve mice received the same treatment with CVF and the additional administration of cyclophosphamide (CPA) 2 days before the R-337 injection. Other mice were injected with UV-inactivated HSV-1 (NAb reduction) 1 day prior to virus injection. Another group of mice received CVF, CPA, and NAb reduction. After administration of R-337, blood samples were withdrawn after 2 and 30 min. (<b>C</b>) Clearance of R-337 PFUs in the bloodstream of HSV-1-immunized and naïve mice subjected to the different treatments or untreated. See <a href="#cancers-15-04042-f004" class="html-fig">Figure 4</a>B for details. At the 2-min time point, the amount of infectious virus in the bloodstream of naïve mice was similar irrespectively from the treatment. Hence its values were averaged to constitute the “start” point. The same approach was applied to the HSV-IMM group. Results are reported as the percentage of infectious viruses relative to the amount of PFU employed. (<b>D</b>) Determination of IC50 values of neutralizing antibodies in sera of HSV-1-immunized mice that received the UV-inactivated HSV-1 (NAb reduction). See <a href="#cancers-15-04042-f005" class="html-fig">Figure 5</a>C for details. (<b>A</b>,<b>C</b>) Each bar indicates the mean value of 3 mice (or the corresponding sera) and vertical bars ± SD.</p>
Full article ">
9 pages, 1418 KiB  
Review
SARS-CoV-2 as an Oncolytic Virus Following Reactivation of the Immune System: A Review
by Joao P. Bounassar-Filho, Laura Boeckler-Troncoso, Jocelyne Cajigas-Gonzalez and Maria G. Zavala-Cerna
Int. J. Mol. Sci. 2023, 24(3), 2326; https://doi.org/10.3390/ijms24032326 - 24 Jan 2023
Cited by 15 | Viewed by 4303
Abstract
The effects SARS-CoV-2 inflicts on human physiology, especially in patients who developed COVID-19, can range from flu-like symptoms to death, and although many lives have been lost during the pandemic, others have faced the resolution of aggressive neoplasms that once proclaimed a poor [...] Read more.
The effects SARS-CoV-2 inflicts on human physiology, especially in patients who developed COVID-19, can range from flu-like symptoms to death, and although many lives have been lost during the pandemic, others have faced the resolution of aggressive neoplasms that once proclaimed a poor prognosis following traditional treatments. The purpose of this review was to analyze several fortunate case reports and their associated biomolecular pathways to further explore new avenues that might provide oncological treatments in the future of medicine. We included papers that discussed cases in which patients affected by COVID-19 suffered beneficial changes in their cancer status. Multiple mechanisms which elicited a reactivation of the host’s immune system included cross-reactivity with viral antigens and downregulation of neoplastic cells. We were able to identify important cases presenting the resolution/remission of different aggressive neoplasms, for which most of the time, standard-of-care treatments offered little to no prospect towards a cure. The intricacy of the defense mechanisms humans have adopted against cancer cells through the millennia are still not well understood, but SARS-CoV-2 has demonstrated that the same ruinous cytokine storm which has taken so many lives can paradoxically be the answer we have been looking for to recalibrate the immunological system to retarget and vanquish malignancies. Full article
(This article belongs to the Special Issue Oncolytic Virotherapy 2.0)
Show Figures

Figure 1

Figure 1
<p>Inflammatory cytokines comprising IL-12, IL-15, IL-2, IFN-α, and INF-β produced by various cells including macrophages, dendritic, and epithelial lung cells in response to SARS-CoV-2 upregulate the expression of activating receptors (NKG2D) in NK cells, promoting a rapid response against tumor cells in the absence of APCs. The downregulation of MHC class I presentation by tumor cells as an evasion mechanism, triggers the activation of NK cells. Upon activation, NK cells release granzymes and perforins, which destroy the tumor. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Reduction of a tumor by the interaction between SARS-CoV-2 and ACE-2/NRP-1 receptors. ACE-2 receptors are present in alveolar pneumocytes and in other tissues, which allows the entry and attachment of the virus. The presence of the coreceptor neuropilin-1 promotes the interaction of the virus with the ACE-2 receptor, which leads to a direct immune response. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>SARS-CoV-2 infects both healthy and malignant NK cells and promotes the expression of the inhibitory molecule NKG2A, thus causing cell senescence. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
18 pages, 2730 KiB  
Article
Differentiable Constraints’ Encoding for Gradient-Based Analog Integrated Circuit Placement Optimization
by António Gusmão, Pedro Alves, Nuno Horta, Nuno Lourenço and Ricardo Martins
Electronics 2023, 12(1), 110; https://doi.org/10.3390/electronics12010110 - 27 Dec 2022
Viewed by 1737
Abstract
Analog IC design is characterized by non-systematic re-design iterations, often requiring partial or complete layout re-design. The layout task usually starts with device placement, where the several performance figures and constraints to be met escalate its complexity immensely, and, due to the inherent [...] Read more.
Analog IC design is characterized by non-systematic re-design iterations, often requiring partial or complete layout re-design. The layout task usually starts with device placement, where the several performance figures and constraints to be met escalate its complexity immensely, and, due to the inherent tradeoffs, an “optimal” floorplan solution does not usually exist. Deep learning models are now establishing for the automation of the placement task of analog integrated circuit layout design, promising to bypass the limitations of existing approaches based on: time-consuming optimization processes with several constraints; or placement retargeting from legacy designs/templates, which rely heavily on legacy layout data. However, as the complexity of analog design cases tackled by these methodologies increases, a broader set of topological constraints must be supported to cover the different layout styles and circuit classes. Here, model-independent differentiable encodings for regularity, boundary, proximity, and symmetry island constraints are formulated for the first time in the literature, and an unsupervised loss function is used for the artificial neural network model to learn how to generate placements that follow them. The use of a deep learning model makes push-button speed placement generation possible, additionally, as only sizing data are required for its training, it discards the need to acquire legacy layouts containing insights into this vast set of, often neglected, constraints. The model is ultimately used to produce floorplans from scratch at push-button speed for real state-of-the-art analog structures, including technology nodes not used for training. A case-study comparison with a floorplan design made by a human-expert presents improvements in the fulfillment of every constraint, reaching an overall improvement of around 70%, demonstrating the approach’s value in placement design. Full article
Show Figures

Figure 1

Figure 1
<p>ANN architecture used to solve the map from the physical dimensions and topological constraints to the placement coordinates. <span class="html-italic">N</span> represents the maximum number of circuit devices supported by the model.</p>
Full article ">Figure 2
<p>Three symmetric placements of the generic circuit: (<b>a</b>) Devices connected to the input/output ports are placed inside the circuit layout; (<b>b</b>) devices are placed within the circuit boundary and three horizontal regularity constraints are implemented; (<b>c</b>) two symmetry islands are formed.</p>
Full article ">Figure 3
<p>One-hot encoded matrices used to provide: (<b>a</b>) boundary constraints in the <math display="inline"><semantics> <mi>x</mi> </semantics></math> axis (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>b); (<b>b</b>) boundary constraints in the <math display="inline"><semantics> <mi>y</mi> </semantics></math> axis (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>b); (<b>c</b>) row regularity constraints (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>b); (<b>d</b>) symmetry relations (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>c); and (<b>e</b>) symmetry island grouping (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>c) to the deep model.</p>
Full article ">Figure 3 Cont.
<p>One-hot encoded matrices used to provide: (<b>a</b>) boundary constraints in the <math display="inline"><semantics> <mi>x</mi> </semantics></math> axis (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>b); (<b>b</b>) boundary constraints in the <math display="inline"><semantics> <mi>y</mi> </semantics></math> axis (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>b); (<b>c</b>) row regularity constraints (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>b); (<b>d</b>) symmetry relations (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>c); and (<b>e</b>) symmetry island grouping (<a href="#electronics-12-00110-f002" class="html-fig">Figure 2</a>c) to the deep model.</p>
Full article ">Figure 4
<p>Example placement where device <math display="inline"><semantics> <mrow> <msub> <mi>D</mi> <mn>1</mn> </msub> </mrow> </semantics></math> should be placed in the top boundary, resulting in three distinct errors <math display="inline"><semantics> <mrow> <msubsup> <mi>B</mi> <mrow> <mn>1</mn> <mo>,</mo> <mn>2</mn> </mrow> <mrow> <mi>t</mi> <mi>t</mi> </mrow> </msubsup> </mrow> </semantics></math>, <math display="inline"><semantics> <mrow> <msubsup> <mi>B</mi> <mrow> <mn>1</mn> <mo>,</mo> <mn>3</mn> </mrow> <mrow> <mi>t</mi> <mi>t</mi> </mrow> </msubsup> </mrow> </semantics></math>, and <math display="inline"><semantics> <mrow> <msubsup> <mi>B</mi> <mrow> <mn>1</mn> <mo>,</mo> <mn>2</mn> </mrow> <mi>h</mi> </msubsup> </mrow> </semantics></math>. Note that no error exists between <math display="inline"><semantics> <mrow> <msub> <mi>D</mi> <mn>1</mn> </msub> </mrow> </semantics></math> and <math display="inline"><semantics> <mrow> <msub> <mi>D</mi> <mn>4</mn> </msub> </mrow> </semantics></math> despite the existing horizontal overlap between them since <math display="inline"><semantics> <mrow> <msub> <mi>D</mi> <mn>4</mn> </msub> </mrow> </semantics></math> is not closer to the top boundary than <math display="inline"><semantics> <mrow> <msub> <mi>D</mi> <mn>1</mn> </msub> </mrow> </semantics></math>.</p>
Full article ">Figure 5
<p>Schematic of the VCOTA [<a href="#B25-electronics-12-00110" class="html-bibr">25</a>]. Current-paths highlighted in green and row regularity constraints highlighted in pink.</p>
Full article ">Figure 6
<p>Test set predictions: (<b>a</b>,<b>b</b>) FVC (tsmc65); (<b>c</b>,<b>d</b>) VCOTA.</p>
Full article ">Figure 6 Cont.
<p>Test set predictions: (<b>a</b>,<b>b</b>) FVC (tsmc65); (<b>c</b>,<b>d</b>) VCOTA.</p>
Full article ">Figure 7
<p>Placements generated for the VCOTA: (<b>a</b>) generated by this work, (<b>b</b>) designed by a human expert designer [<a href="#B25-electronics-12-00110" class="html-bibr">25</a>].</p>
Full article ">
23 pages, 2063 KiB  
Article
EEG-Based Emotion Recognition by Retargeted Semi-Supervised Regression with Robust Weights
by Ziyuan Chen, Shuzhe Duan and Yong Peng
Systems 2022, 10(6), 236; https://doi.org/10.3390/systems10060236 - 29 Nov 2022
Cited by 4 | Viewed by 2257
Abstract
The electroencephalogram (EEG) can objectively reflect the emotional state of human beings, and has attracted much attention in the academic circles in recent years. However, due to its weak, non-stationary, and low signal-to-noise properties, it is inclined to cause noise in the collected [...] Read more.
The electroencephalogram (EEG) can objectively reflect the emotional state of human beings, and has attracted much attention in the academic circles in recent years. However, due to its weak, non-stationary, and low signal-to-noise properties, it is inclined to cause noise in the collected EEG data. In addition, EEG features extracted from different frequency bands and channels usually exhibit different levels of emotional expression abilities in emotion recognition tasks. In this paper, we fully consider the characteristics of EEG and propose a new model RSRRW (retargeted semi-supervised regression with robust weights). The advantages of the new model can be listed as follows. (1) The probability weight is added to each sample so that it could help effectively search noisy samples in the dataset, and lower the effect of them at the same time. (2) The distance between samples from different categories is much wider than before by extending the ϵ-dragging method to a semi-supervised paradigm. (3) Automatically discover the EEG emotional activation mode by adaptively measuring the contribution of sample features through feature weights. In the three cross-session emotion recognition tasks, the average accuracy of the RSRRW model is 81.51%, which can be seen in the experimental results on the SEED-IV dataset. In addition, with the support of the Friedman test and Nemenyi test, the classification of RSRRW model is much more accurate than that of other models. Full article
(This article belongs to the Topic Human–Machine Interaction)
Show Figures

Figure 1

Figure 1
<p>Emotion models VA (<b>a</b>) and VAD (<b>b</b>). (<b>a</b>) The VA model consists of the dimensions named valence and arousal, (<b>b</b>) The VAD model consists of the dimensions named valence, arousal and dominance.</p>
Full article ">Figure 2
<p>The general framework of RSRRW model.</p>
Full article ">Figure 3
<p>Experimental protocol for SEED-IV [<a href="#B42-systems-10-00236" class="html-bibr">42</a>].</p>
Full article ">Figure 4
<p>Nemenyi test result.</p>
Full article ">Figure 5
<p>The average importance of EEG channels (<b>a</b>) and frequency bands (<b>b</b>) obtained by RSRRW.</p>
Full article ">Figure 6
<p>Top 10 EEG channels.</p>
Full article ">Figure 7
<p>Top 10 EEG channels.</p>
Full article ">Figure 8
<p>Examples to show the effectiveness of the <math display="inline"><semantics> <mi>ϵ</mi> </semantics></math>-dragging.</p>
Full article ">Figure 9
<p>Visualization of sample weights <math display="inline"><semantics> <mi mathvariant="bold">s</mi> </semantics></math>.</p>
Full article ">Figure 9 Cont.
<p>Visualization of sample weights <math display="inline"><semantics> <mi mathvariant="bold">s</mi> </semantics></math>.</p>
Full article ">
34 pages, 8144 KiB  
Article
3D Pose Estimation and Tracking in Handball Actions Using a Monocular Camera
by Romeo Šajina and Marina Ivašić-Kos
J. Imaging 2022, 8(11), 308; https://doi.org/10.3390/jimaging8110308 - 10 Nov 2022
Cited by 8 | Viewed by 6003
Abstract
Player pose estimation is particularly important for sports because it provides more accurate monitoring of athlete movements and performance, recognition of player actions, analysis of techniques, and evaluation of action execution accuracy. All of these tasks are extremely demanding and challenging in sports [...] Read more.
Player pose estimation is particularly important for sports because it provides more accurate monitoring of athlete movements and performance, recognition of player actions, analysis of techniques, and evaluation of action execution accuracy. All of these tasks are extremely demanding and challenging in sports that involve rapid movements of athletes with inconsistent speed and position changes, at varying distances from the camera with frequent occlusions, especially in team sports when there are more players on the field. A prerequisite for recognizing the player’s actions on the video footage and comparing their poses during the execution of an action is the detection of the player’s pose in each element of an action or technique. First, a 2D pose of the player is determined in each video frame, and converted into a 3D pose, then using the tracking method all the player poses are grouped into a sequence to construct a series of elements of a particular action. Considering that action recognition and comparison depend significantly on the accuracy of the methods used to estimate and track player pose in real-world conditions, the paper provides an overview and analysis of the methods that can be used for player pose estimation and tracking using a monocular camera, along with evaluation metrics on the example of handball scenarios. We have evaluated the applicability and robustness of 12 selected 2-stage deep learning methods for 3D pose estimation on a public and a custom dataset of handball jump shots for which they have not been trained and where never-before-seen poses may occur. Furthermore, this paper proposes methods for retargeting and smoothing the 3D sequence of poses that have experimentally shown a performance improvement for all tested models. Additionally, we evaluated the applicability and robustness of five state-of-the-art tracking methods on a public and a custom dataset of a handball training recorded with a monocular camera. The paper ends with a discussion apostrophizing the shortcomings of the pose estimation and tracking methods, reflected in the problems of locating key skeletal points and generating poses that do not follow possible human structures, which consequently reduces the overall accuracy of action recognition. Full article
(This article belongs to the Section Computer Vision and Pattern Recognition)
Show Figures

Figure 1

Figure 1
<p>Creating a sequence of poses using human pose estimation to produce human skeleton keypoints and object tracking for grouping collected poses across frames (<span class="html-italic">t</span>) into a single sequence of poses.</p>
Full article ">Figure 2
<p>Standard 18-person keypoints in pose estimation.</p>
Full article ">Figure 3
<p>Taxonomy of pose estimation approaches based on Ref. [<a href="#B2-jimaging-08-00308" class="html-bibr">2</a>].</p>
Full article ">Figure 4
<p>Heatmap poses estimation. It starts by creating heatmaps of all keypoints within the image, and then additional methods are used to construct the final stick figure.</p>
Full article ">Figure 5
<p>The top-down pipeline in multi-person approach for pose estimation. It starts by detecting all persons within an image and producing bounding boxes, on which a single-person approach is applied. The result are keypoints for each detected person, after which the pipeline may involve additional post-processing steps and improving the final results.</p>
Full article ">Figure 6
<p>The bottom-up pipeline in multi-person approach for pose estimation. It starts by detecting all the keypoints in the image, which are then associated with human instances.</p>
Full article ">Figure 7
<p>An example of pose retargeting where the predicted pose is retargeted based on the target skeleton. Retargeting will translate the joint angles from the predicted pose to a standardized skeleton, thus ensuring that a pose has the same lengths of limbs.</p>
Full article ">Figure 8
<p>Examples from the Human3.6 dataset (<b>a</b>) and the RI-HJS dataset (<b>b</b>) for 3D pose estimation.</p>
Full article ">Figure 9
<p>Taxonomy of the tracking methods.</p>
Full article ">Figure 10
<p>Two frames of a tracked player executing a jump shop where poses are estimated and performed necessary transformation. Blue bounding boxes visualize the detectors’ outputs, while white bounding boxes visualize the tracking algorithm bounding box prediction. To standardize pose sizes because players can be further away or closer to the camera, we perform transformations to the pose (i.e., standardization).</p>
Full article ">Figure 11
<p>A 3D plot visualization of the 2D sequence joints in space and time when executing a jump shot, showing a side and top view of the plot.</p>
Full article ">Figure 12
<p>Examples from the tracking datasets DanceTrack (<b>a</b>), SportsMOT (<b>b</b>), MOT17 (<b>c</b>), and RI-HB-PT (<b>d</b>).</p>
Full article ">Figure 13
<p>Comparison of the 3D pose estimation model results in terms of PA-PCK on Human 3.6M and custom RI-HJS datasets (higher is better). All models experienced a significant drop in performance on the RI-HJS dataset, except the two-step model UDP-Pose + EvoSkeleton, which retained high accuracy, showing robustness in an unseen environment. It is interesting to note that all two-step models that use VideoPose3D experienced the largest performance drop compared to other models.</p>
Full article ">Figure 14
<p>Comparison of the 3D pose estimation model results in terms of PA-MPJPE on Human 3.6M and custom RI-HJS datasets (lower is better). The comparison shows a significant drop in performance on the RI-HJS dataset, which is not surprising given that the models have never seen uncommon poses such as the handball jump-shot from the RI-HJS dataset. Two-step models that use VideoPose3D are more prone to errors due to unseen data, as they have the largest performance drop.</p>
Full article ">Figure 15
<p>The robustness of the tested 3D models trained on the Human3.6M dataset shown as a difference of obtained results and performance drops between PA-PCK pose estimation results on Human 3.6M and custom RI-HJS datasets.</p>
Full article ">Figure 16
<p>Comparison of pose sequence estimation in terms of PA-PCK on custom RI-HJS datasets (higher score is better). Two-step models that use EvoSkeleton show a significant improvement when using smoothing on the sequence of poses, showing the lack of consistency in the process of “lifting” 2D keypoints to 3D space. When using retargeting on the ground truth and smoothed predicted sequence, the results are significantly improved, indicating that all models lack an understanding of the human skeleton structure, which is especially true in the case of VideoPose3D.</p>
Full article ">Figure 17
<p>Comparison of pose sequence estimation in terms of PA-MPJPE measure on Human3.6M datasets (lower is better). All models show a slight improvement when using smoothing on the sequence of poses, showing the lack of consistency in the detection location of keypoints and “lifting” 2D keypoints to the 3D space. An exception to this conclusion is the VideoPose3D model, which constructed a smooth sequence of poses by utilizing temporal information. When using retargeting on the ground truth and smoothed predicted sequence, results are significantly improved, which indicates that all models lack an understanding of the human skeleton structure.</p>
Full article ">Figure 18
<p>Examples of poor detection of keypoint location that happens mostly because the true keypoint location is occluded or less clear. The right side of the player is coloured purple while the left side of the person is coloured blue. In the first row, where the left elbow and hand are not visible, methods PoseRegression and ArtTrack incorrectly assume the location, while Mask R-CNN and UDP-Pose placed the left elbow and hand on the right elbow and hand of the player. The second row shows a situation where parts are visible but less clear, where all methods fail to detect the left hand, which is close to the head, while methods ArtTrack and Mask R-CNN miss the right foot. The third row shows situations where methods ArtTrack and Mask R-CNN produced invalid human structures by detecting the right foot on the location of the left foot, while the UDP-Pose almost correctly detected the keypoints. PoseRegression generally did not perform well on uncommon poses such as the handball jump-shot.</p>
Full article ">Figure 19
<p>Examples of wrong player side keypoint detection, with an unclear reason for this occurrence. The right side of the player is coloured purple while the left side of the person is coloured blue. While all methods detected almost all keypoints correctly, all methods switched sides of the player, producing an invalid pose. Occurrences of this problem can also be observed on a few keypoints in <a href="#jimaging-08-00308-f018" class="html-fig">Figure 18</a>.</p>
Full article ">Figure 20
<p>Comparison of pose sequence estimation in terms of PA-PCK on custom RI-HJS datasets before and after additional training of the Mask R-CNN and UDP-Pose models on training part on RI-HJS dataset (higher score is better).</p>
Full article ">Figure 21
<p>Examples of detection after training on the 227 images of the RI-HJS dataset. The right side of the player is coloured purple while the left side of the person is coloured blue. Untrained models missed detection when the left hand was hidden or less clear, as shown in <a href="#jimaging-08-00308-f018" class="html-fig">Figure 18</a>. After training, UDP-Pose successfully detected the left hand on the second row, while on the first row, it made a reasonable guess of the hand position. Mask R-CNN performed worse on both examples after training, wrongly detecting the right knee location on the left knee.</p>
Full article ">Figure 22
<p>Examples of detection after training on the 227 images of the RI-HJS dataset. The right side of the player is coloured purple while the left side of the person is coloured blue. Untrained models made a mistake and switched the players’ sides, shown in <a href="#jimaging-08-00308-f019" class="html-fig">Figure 19</a>. After training, UDP-Pose successfully detected keypoints on the correct sides, while Mask R-CNN did not manage to detect all keypoint sides correctly.</p>
Full article ">
21 pages, 2439 KiB  
Article
Adapter-Mediated Transduction with Lentiviral Vectors: A Novel Tool for Cell-Type-Specific Gene Transfer
by Nicole Cordes, Nora Winter, Carolin Kolbe, Bettina Kotter, Joerg Mittelstaet, Mario Assenmacher, Toni Cathomen, Andrew Kaiser and Thomas Schaser
Viruses 2022, 14(10), 2157; https://doi.org/10.3390/v14102157 - 30 Sep 2022
Cited by 3 | Viewed by 6414
Abstract
Selective gene delivery to a cell type of interest utilizing targeted lentiviral vectors (LVs) is an efficient and safe strategy for cell and gene therapy applications, including chimeric antigen receptor (CAR)-T cell therapy. LVs pseudotyped with measles virus envelope proteins (MV-LVs) have been [...] Read more.
Selective gene delivery to a cell type of interest utilizing targeted lentiviral vectors (LVs) is an efficient and safe strategy for cell and gene therapy applications, including chimeric antigen receptor (CAR)-T cell therapy. LVs pseudotyped with measles virus envelope proteins (MV-LVs) have been retargeted by ablating binding to natural receptors while fusing to a single-chain antibody specific for the antigen of choice. However, the broad application of MV-LVs is hampered by the laborious LV engineering required for every new target. Here, we report the first versatile targeting system for MV-LVs that solely requires mixing with biotinylated adapter molecules to enable selective gene transfer. The analysis of the selectivity in mixed cell populations revealed transduction efficiencies below the detection limit in the absence of an adapter and up to 5000-fold on-to-off-target ratios. Flexibility was confirmed by transducing cell lines and primary cells applying seven different adapter specificities in total. Furthermore, adapter mixtures were applied to generate CAR-T cells with varying CD4/CD8-ratios in a single transduction step. In summary, a selective and flexible targeting system was established that may serve to improve the safety and efficacy of cellular therapies. Compatibility with a wide range of readily available biotinylated molecules provides an ideal technology for a variety of applications. Full article
(This article belongs to the Special Issue Structure and Cell Biology of Viral Infection)
Show Figures

Figure 1

Figure 1
<p>Concept of LLE adapter-mediated transduction with lentiviral vectors. (<b>A</b>) For directly targeted LVs, a binding ligand (e.g., scFv) is fused to the H protein of MV that is mutated to block natural receptor interactions. This allows specific entry to cells expressing the respective antigen (left). The adapter-mediated transduction is based on an scFv fused to the H protein of MV pseudotyped LVs, which binds to biotin on a specific adapter molecule. In the absence of adapter molecules, transduction is abolished (middle). Only in the presence of a biotinylated adapter molecule transduction with the Adapter-LV (Ad-LV) is mediated (right). (<b>B</b>) Here, antibodies or antibody fragments were used as adapter molecules. The specific linker chemistry used for biotinylation generates a linker moiety and a label moiety (biotin). The scFv used in this study interacts with the linked biotin (linker–label epitope, LLE) thereby avoiding recognition of free biotin.</p>
Full article ">Figure 2
<p>Establishing the Ad-LV system. (<b>A</b>,<b>B</b>) Flow cytometric analysis of the surface expression of the H protein fused to the original scFv or a mutant variant (Opt) on HEK293T cells by either staining for the C-terminal HIS-tag (<b>A</b>) of the H protein or with fluorescently labeled biotin (<b>B</b>) or untreated cells (w/o). Data are represented as mean ± SD of 4 independent experiments. (<b>C</b>) LV productivity for concentrated LV supernatant was determined on biotinylated cells. Data are represented as mean ± SD of 3 independent experiments. (<b>D</b>) The optimal order of combination of the three components (LVs, cells, and adapter) required for transduction was evaluated via transduction of Raji cells with a GFP-encoding Ad-LV (0.05 TU/cell) using an LLE-CD20-mAB (clone: LT20, 1000 ng/mL). While cells with vector were preincubated for 30 min at 37 °C, cells with adapter or vector with adapter were preincubated for 30 min at 4 °C. Afterwards, the respective third component was added. Transduction without the addition of any adapter (w/o) was used to confirm specificity. MV-LV directly targeting CD20 (α-CD20-LV) was used as positive control. Transduction efficiency was analyzed 3 days post transduction via quantification of the GFP-positive cells using flow cytometry. Data are represented as mean ± SD of 3 technical replicates. Statistic ordinary one-way ANOVA using Tukey’s multiple comparisons test; ns, non-significant; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Flexibility of Ad-LV towards adapter specificity and format. The flexibility of the adapter system was evaluated by transduction of cell lines using different adapter specificities. SupT1 (CD4 + CD8 + CD46+) (<b>A</b>) and Raji cells (CD19 + CD20 + CD46+) (<b>B</b>) were left untreated (w/o) or were transduced with GFP-encoding Ad-LV with 0.2 TU/cell using LLE-CD4 (clone: MT-466), LLE-CD8 (clone: BW135/80), LLE-CD19 (clone: LT19), LLE-CD20 (clone: LT20), and LLE-CD46 (clone: REA312) mABs (+) in concentrations ranging from 1 to 1000 ng/mL. Non-biotinylated mABs (-) used in the highest concentration and transduction in the absence of any adapter (-) were used to confirm specificity. VSV-G LV was used as reference. Transduction efficiency was analyzed 3 days (Raji) or 4 days (SupT1) post transduction via quantification of GFP-positive cells using flow cytometry. (<b>C</b>) Transduction efficiency of SupT1 comparing fab and f(ab)<sub>2</sub> adapter formats of the same LLE-CD4 or LLE-CD8 clone with adapter concentrations ranging from 0.1 to 1 μg/mL using a GFP-encoding Ad-LV (0.1 TU/cell). VSV-G LV was used as reference. Data are represented as mean ± SD of 3 technical replicates.</p>
Full article ">Figure 4
<p>Selectivity of Ad-LVs in co-culture assay using different transduction protocols. To confirm selectivity of Ad-LVs, target cells were transduced with 0.2 TU/cell in a co-culture with non-target cells. Cell-trace-dye-labeled SupT1 cells (CD4+, CD8+, and CD46+) were co-cultivated in a 1:1 ratio with Raji cells (CD19, CD20+). (<b>A</b>) Representative data are shown using spinoculation. The co-culture was left untreated (-) or was transduced with Ad-LVs in the presence of non-biotinylated mAB (-) or LLE-CD4 mAB (clone: MT-466, 50 ng/mL), LLE-CD8 mAB (clone: BW135/80, 10 ng/mL) or LLE-CD20 mAB (clone: LT20, 500 ng/mL). (<b>B</b>) Three transduction conditions were compared using no enhancer, using the transduction enhancer Vectofusin<sup>®</sup>-1, or using spinoculation. Transduction efficiency was evaluated 4 days post transduction via gating on the cell-trace-dye-labeled SupT1 and the CD19-expressing Raji cells. Data are represented as mean ± SD of 3 technical replicates.</p>
Full article ">Figure 5
<p>Selective transduction of human T and B cells within activated PBMCs using different transduction protocols. Activated PBMCs were transduced with Ad-LVs with 0.1 TU/cell in the absence of adapter (w/o) and in the presence of CD4-mAB (clone:MT-466, 100 ng/mL), CD8-mAB (clone: BW135/80, 10 ng/mL), or CD19-mAB (clone: LT19, 100 ng/mL) with (+) or without LLE-tag (-). (<b>A</b>) Representative data are shown using LLE-CD8 mAB or CD8-mAB w/o enhancer. (<b>B</b>) Three transduction conditions were compared: using no enhancers, using spinoculation, or the transduction enhancer Vectofusin<sup>®</sup>-1. VSV-G LVs were used as positive control. Transduction efficiency was evaluated 10 days post transduction gated on the subpopulations (CD4+ T cells, CD8+ T cells, and B cells). Data are represented as mean ± SD of 3 different donors and 2 independent experiments.</p>
Full article ">Figure 6
<p>Selective transduction of activated murine T cells. Murine T cells were isolated from murine spleens, activated overnight, and transduced using Ad-LVs at a dose of 5 TU/cell. (<b>A</b>) Representative data are shown of CD4+ T cells transduced in the absence of adapter or in the presence of LLE-CD4 fab. (<b>B</b>) Isolated murine T cells from spleen were either left untreated (-) or transduced with Ad-LVs in the absence of adapter (-) or in the presence of LLE-CD4 fab (clone: GK1.5) and LLE-CD8 fab (clone: 53–6.7)-specific adapter molecules (500 ng/mL). VSV-G LVs were used as positive controls. Data are represented as mean ± SD of 3 different mice.</p>
Full article ">Figure 7
<p>Generation of CD20-CAR-T cells with Ad-LVs. CAR-T cells were generated from activated Pan T cells using Ad-LV (0.5 TU/cell). To generate CAR-T cells with varying CD4/CD8 ratios, either LLE-CD4 f(ab)<sub>2</sub> or LLE-CD8 f(ab)<sub>2</sub> adapters alone or a mixture of both adapters in 1:1 or 1:5 ratio was used. (<b>A</b>) Representative data of T cells transduced with Ad-LVs using LLE-CD4 f(ab)<sub>2</sub> (clone: MT-466) or LLE-CD8 f(ab)<sub>2</sub> (clone: BW135/80) alone or in a mixture are shown. (<b>B</b>) The CD4/CD8 ratio of the CAR-T cells is shown. To analyze if the generated CAR-T cells are functional co-cultures with CD20, + GFP+ Raji cells were set up in an E:T ratio of 1:1. (<b>C</b>) Specific lysis of target cells was analyzed via flow cytometry 24 h post setup of the co-culture. (<b>D</b>) Secretion of specific cytokines was analyzed using the MACS Plex Assay Kit. (<b>E</b>) Activation/exhaustion marker expression was analyzed 6 days post setup of the co-culture assay. Data are represented as mean ± SD of 4 different donors. Statistics: repeated measure one-way ANOVA using Dunnett’s or Tukey’s multiple comparison test; ns = non-significant * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
Back to TopTop