[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

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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,756)

Search Parameters:
Keywords = HCT116 cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 2092 KiB  
Article
Enzymic Activity, Metabolites, and Hematological Responses Changes of Clinical Healthy High-Risk Beef Calves During Their First 56-Days from Arrival
by Octavio Carrillo-Muro, Pedro Hernández-Briano, Paola Isaira Correa-Aguado, Alejandro Rivera-Villegas, Oliver Yaotzin Sánchez-Barbosa, Rosalba Lazalde-Cruz, Alberto Barreras, Alejandro Plascencia and Daniel Rodríguez-Cordero
Animals 2025, 15(2), 133; https://doi.org/10.3390/ani15020133 - 8 Jan 2025
Viewed by 203
Abstract
The objective of this study was to evaluate the changes in enzymic activity, metabolites, and hematological responses during the first 56-d of arrival of newly received calves, which were qualified at reception as high-risk but diagnosed as clinically healthy. A total of 320 [...] Read more.
The objective of this study was to evaluate the changes in enzymic activity, metabolites, and hematological responses during the first 56-d of arrival of newly received calves, which were qualified at reception as high-risk but diagnosed as clinically healthy. A total of 320 blood samples were taken from 64 crossbred bull calves (average initial body weight = 148.3 ± 1.3 kg) at different times from arrival (d 0, 14, 28, 42, and 56 of received). Calves included in the study were received in June (n = 20), November (n = 24), and April (n = 20); thus, experimental treatments were arranged in a generalized complete block design (three blocks = month of arrival). The following parameters were determined: total white blood cells (WBC): lymphocytes (LYM), lymphocytes % (LYM%), monocytes (MON), monocytes % (MON%), granulocytes (GRA), granulocytes % (GRA%), platelets (PLT), and mean platelet volume (MPV); red blood cells (RBC): red blood cell distribution width test % (RDW%), hematocrit (HCT), and mean corpuscular volume (MCV); hemoglobin (HGB): mean corpuscular hemoglobin (MCH) and mean corpuscular hemoglobin concentration (MCHC). The enzymatic activity and metabolites analyzed were alkaline phosphatase (ALP), gamma glutamyltransferase (GGT), aspartate aminotransferase (AST), alanine aminotransferase (ALT), total protein (TP), albumin (ALB), globulin (GLO), ALB/GLO ratio, blood urea nitrogen (BUN), creatinine (CRE), total bilirubin (TBIL), total cholesterol (TCHO), triglycerides (TG); (4) calcium (Ca), glucose (GLU), sodium (Na+), potassium (K+), and chlorine (Cl). It was observed that ALP, ALT, TP, ALB, GLO, ALB/GLO ratio, TCHO, TG, Ca, and GLU increased as days from reception increased (linear effect, p ≤ 0.04), whereas CRE and TBIL were reduced (linear effect, p ≤ 0.02). A quadratic response (p ≤ 0.001) was observed to GGT and AST values being maximal on days 1 and 56 after arrival (p ≤ 0.001). Na+, K+, and Cl concentrations were not affected by prolonged days after arrival. Finally, blood cells of LYM, LYM%, PLT, RBC, HGB, HCT%, MCV, and MCH increased (linear effect, p ≤ 0.001) as the number of days after arrival increased. Whereas MON% was linearly decreased (p ≤ 0.05). It was concluded that even when all parameters were within the range of reference intervals (RIs) determined for healthy cattle, during the period of monitoring, as the days after arrival lengthened, blood serum parameters related to health and immunity increased, and metabolites related to tissue injury decreased. In contrast, plasmatic electrolytes (Na+, K+, and Cl) were slightly reduced as the day after arrival increased. Apparently, at least 42 d is the minimum period after arrival to permit calves to reach more adequate physiological and metabolic conditions before starting the fattening phase. Full article
Show Figures

Figure 1

Figure 1
<p>Received dates (3, block) and sampling schedule performed at 0, 14, 28, 42, and 56 d from reception to feedlot (five samplings).</p>
Full article ">Figure 2
<p>Effect of days after being received (0, 14, 28, 42, and 56 d) on metabolites of high-risk beef calves “clinically healthy”. <sup>a–c</sup> Means a row with different superscripts differ (<span class="html-italic">p</span> ≤ 0.05) according to Tukey’s test.</p>
Full article ">Figure 2 Cont.
<p>Effect of days after being received (0, 14, 28, 42, and 56 d) on metabolites of high-risk beef calves “clinically healthy”. <sup>a–c</sup> Means a row with different superscripts differ (<span class="html-italic">p</span> ≤ 0.05) according to Tukey’s test.</p>
Full article ">Figure 3
<p>Effect of days after being received (0, 14, 28, 42, and 56 d) on blood cells of high-risk beef calves “clinicaly healthy” <sup>a–c</sup> Means a row with different superscripts differ (<span class="html-italic">p</span> ≤ 0.05) according to Tukey’s test.</p>
Full article ">Figure 3 Cont.
<p>Effect of days after being received (0, 14, 28, 42, and 56 d) on blood cells of high-risk beef calves “clinicaly healthy” <sup>a–c</sup> Means a row with different superscripts differ (<span class="html-italic">p</span> ≤ 0.05) according to Tukey’s test.</p>
Full article ">
10 pages, 1053 KiB  
Article
Isolation and Structural Identification of New Diol Esters of Okadaic Acid and Dinophysistoxin-1 from the Cultured Prorocentrum lima
by Yeong Kwang Ji, Semin Moon, Sangbum Lee, Yun Na Kim, Eun Ju Jeong and Jung-Rae Rho
Toxins 2025, 17(1), 28; https://doi.org/10.3390/toxins17010028 - 7 Jan 2025
Viewed by 260
Abstract
Prorocentrum, a dinoflagellate responsible for producing diarrhetic shellfish poisoning (DSP) toxins, poses significant threats to marine ecosystems, aquaculture industries, and human health. DSP toxins, including okadaic acid (OA), dinophysis toxin (DTX), and their diverse derivatives, continue to be identified and characterized. In [...] Read more.
Prorocentrum, a dinoflagellate responsible for producing diarrhetic shellfish poisoning (DSP) toxins, poses significant threats to marine ecosystems, aquaculture industries, and human health. DSP toxins, including okadaic acid (OA), dinophysis toxin (DTX), and their diverse derivatives, continue to be identified and characterized. In this study, we report the isolation of four new diol esters of OA/DTX-1 from large-scale cultures of Prorocentrum lima. Their chemical structures were elucidated through comprehensive NMR and MS analyses, along with structural comparisons with the well-known OA. Notably, compound 1 featured an additional ester group within the diol unit, while compound 2 was revealed to be a C11 diol ester. The cytotoxicity of these newly isolated derivatives was evaluated against three cell lines: Neuro2a (mouse), HCT116 (human), and HepG2 (human). All diol esters exhibited cytotoxic effects, with compound 3 displaying toxicity comparable to OA. These results expand our understanding of DSP toxin diversity and provide valuable insight into the structural variations and biological activity of diol esters of OA/DTX-1. Full article
Show Figures

Figure 1

Figure 1
<p>Four new diol derivatives of OA/DTX-1 from the cultures <span class="html-italic">P. lima</span>.</p>
Full article ">Figure 2
<p>COSY (bold lines), key HMBC (arrows) correlations in the diol moiety of (<b>a</b>) compounds <b>1</b>, (<b>b</b>) <b>2</b>, (<b>c</b>) <b>3</b>, and (<b>d</b>) <b>4</b>.</p>
Full article ">Figure 3
<p>Cytotoxic effects of compounds <b>1</b>–<b>4</b> and okadaic acid on Neuro-2a (<b>a</b>), HCT116 (<b>b</b>), and HepG2 (<b>c</b>) cells.</p>
Full article ">Figure 3 Cont.
<p>Cytotoxic effects of compounds <b>1</b>–<b>4</b> and okadaic acid on Neuro-2a (<b>a</b>), HCT116 (<b>b</b>), and HepG2 (<b>c</b>) cells.</p>
Full article ">
17 pages, 4013 KiB  
Article
A Novel Bacteriophage with the Potential to Inhibit Fusobacterium nucleatum-Induced Proliferation of Colorectal Cancer Cells
by Ho Yin Pekkle Lam, Meng-Jiun Lai, Pin-Chun Wang, Wen-Jui Wu, Li-Kuang Chen, Hsiang-Wei Fan, Chun-Chieh Tseng, Shih-Yi Peng and Kai-Chih Chang
Antibiotics 2025, 14(1), 45; https://doi.org/10.3390/antibiotics14010045 - 7 Jan 2025
Viewed by 396
Abstract
Background: Increasing evidence shows that Fusobacterium nucleatum (F. nucleatum) largely affects colorectal cancer (CRC) growth and progression; therefore, the inhibition of intratumoral F. nucleatum may be one realistic approach to combat CRC. Although antibiotics are helpful in eliminating bacteria, the major [...] Read more.
Background: Increasing evidence shows that Fusobacterium nucleatum (F. nucleatum) largely affects colorectal cancer (CRC) growth and progression; therefore, the inhibition of intratumoral F. nucleatum may be one realistic approach to combat CRC. Although antibiotics are helpful in eliminating bacteria, the major problem remains the rise of potential antibiotic-resistant strains and antibiotic-associated adverse effects. Currently, bacteriophage therapy has gained interest because of its high selectivity to bacterial hosts and may become a realistic approach in treating bacteria-associated cancers. Methods: In this study, a new F. nucleatum bacteriophage, ØTCUFN3, was isolated and its biological characteristics were identified. In vitro and in vivo studies were performed to investigate the effect of ØTCUFN3 in combating F. nucleatum-induced CRC growth. Results: By applying ØTCUFN3 to F. nucleatum-induced CRC cell lines, p53+/+, and p53−/− isogenic HCT116 cells, our results revealed an inhibition of CRC proliferation and the expression of epithelial-to-mesenchymal transition (EMT) markers. ØTCUFN3 injection also reduced the growth of F. nucleatum-induced mouse xenografts. Conclusions: Our results demonstrated the use of F. nucleatum bacteriophage against CRC, laying the foundation for the future usage of bacteriophage in cancer treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Plaquing phenotype and morphology of ØTCUFN3. (<b>a</b>) Representative images of lytic plaques of ØTCUFN3 against <span class="html-italic">F. nucleatum</span> strain 34597 on a double-layer agar plate. (<b>b</b>) Representative transmission electron microscopic (TEM) image of ØTCUFN3, showing an icosahedron head with a noncontractile long tail attached to it.</p>
Full article ">Figure 2
<p>Genomic organization of ØTCUFN3. The bacteriophage contains 127,354 bp and 153 open reading frames (ORFs). ORFs predicted as hypothetical proteins are marked as grey; structural proteins as blue; DNA- and RNA-related proteins as red; endolysin-related proteins as green; unclassified functional proteins as purple. The figure was created using the SnapGene program, <a href="http://www.snapgene.com" target="_blank">http://www.snapgene.com</a> (accessed on 16 October 2024).</p>
Full article ">Figure 3
<p>Biological characteristics of ØTCUFN3. (<b>a</b>) The optimal multiplicity of infection (MOI) of ØTCUFN3 was determined by infecting <span class="html-italic">F. nucleatum</span> 34597. MOI = 10 was found to be the optimal MOI of ØTCUFN3. (<b>b</b>) Infection assay of ØTCUFN3 against <span class="html-italic">F. nucleatum</span> 34597. MOI = 10 and 100 show complete inhibition after 24 h. (<b>c</b>,<b>d</b>) Stability of ØTCUFN3 at different (<b>c</b>) temperatures and (<b>d</b>) pH. Phage incubated in BHI broth at 37 °C was served as a control. Results shown are mean ± S.D. from three independent tests. * <span class="html-italic">p</span>-value &lt; 0.005; ** <span class="html-italic">p</span>-value &lt; 0.001; **** <span class="html-italic">p</span>-value &lt; 0.0001 according to one-way ANOVA.</p>
Full article ">Figure 4
<p>ØTCUFN3 suppresses proliferation of <span class="html-italic">Fusobacterium nucleatum</span>-induced p53<sup>−/−</sup> HCT-116 cells. (<b>a</b>) p53 wild-type (p53<sup>+/+</sup>) and (<b>b</b>) p53 knockout (p53<sup>−/−</sup>) HCT-116 cells were treated with <span class="html-italic">F. nucleatum</span> (F. n.) at an MOI of 100, followed by treatment of ØTCUFN3 at MOIs that ranged from 0.1 to 100 against the bacteria. The cells were cultured for 24 h, and proliferation was measured by CCK-8 assay at a wavelength of 450 nm. Results shown are mean ± S.D. from three independent tests. * <span class="html-italic">p</span>-value &lt; 0.05 determined by one-way ANOVA.</p>
Full article ">Figure 5
<p>ØTCUFN3 potentially suppresses epithelial-to-mesenchymal transition (EMT) of <span class="html-italic">Fusobacterium nucleatum</span>-induced, p53-mutated HCT-116 cells. (<b>a</b>–<b>d</b>) p53 wild-type (p53<sup>+/+</sup>) and (<b>e</b>–<b>h</b>) p53 knockout (p53<sup>−/−</sup>) HCT-116 cells were treated with <span class="html-italic">F. nucleatum</span> (F. n.) at a MOI = 100 and ØTCUFN3 at a MOI = 10 against bacteria for 24 h. Gene expression of EMT markers, including N-cadherin (<b>a</b>,<b>e</b>), Snail (<b>b</b>,<b>f</b>), Vimentin (<b>c</b>,<b>g</b>), and E-cadherin (<b>d</b>,<b>h</b>) was measured by qRT-PCR. Results shown are mean ± S.D. from three independent tests. * <span class="html-italic">p</span>-value &lt; 0.05, ** <span class="html-italic">p</span>-value &lt; 0.01, and *** <span class="html-italic">p</span>-value &lt; 0.001 according to one-way ANOVA.</p>
Full article ">Figure 6
<p>ØTCUFN3 inhibited tumor growth in the HCT-116 p53<sup>−/−</sup> murine xenograft model. (<b>a</b>) Experimental scheme. Briefly, 1 × 10<sup>6</sup> CFU <span class="html-italic">F. nucleatum</span> was mixed with 1 × 10<sup>7</sup> suspended cells and injected subcutaneously into the right flanks of the mice. 1 × 10<sup>7</sup> PFU ØTCHFN3 was injected next to the xenograft at days 3, 5, and 7. Mice were sacrificed on day 8. (<b>b</b>) Body weight of the mice. (<b>c</b>) Tumor volume. For (<b>b</b>,<b>c</b>), the blue arrow represents the day of ØTCHFN3 injection. (<b>d</b>) Photograph of the excised xenograft tumor. (<b>e</b>) Tumor weight. (<b>f</b>) Abundance of <span class="html-italic">F. nucleatum</span> in the tumor. Results shown are mean ± S.D. with four mice in each group.* <span class="html-italic">p</span>-value &lt; 0.05 according to one-way ANOVA.</p>
Full article ">Figure 7
<p>ØTCUFN3 induces tumor necrosis in the HCT-116 p53<sup>−/−</sup> murine xenograft model. (<b>a</b>–<b>d</b>) Representative image of H&amp;E-stained sections. ØTCUFN3 treatment induces inflammatory infiltration and significant necrosis in xenografts infected or uninfected with <span class="html-italic">F. nucleatum</span>. (<b>e</b>–<b>h</b>) Representative image of Ki-67-stained sections. (<b>i</b>) Percentage of Ki-67 positivity. Scale bar, 100 μm. Results shown are mean ± S.D. with four mice in each group. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 according to one-way ANOVA.</p>
Full article ">Figure 8
<p>ØTCUFN3 activates immune responses but does not induce pathological injuries. (<b>a</b>) Experimental scheme. Vehicle (PBS) or ØTCUFN3 was injected intraperitoneally three times a week into healthy BALB/c mice for four weeks. (<b>b</b>) Body weight of the mice. (<b>c</b>) Spleen weight index. The calculation was performed by dividing the spleen weight by the body weight. (<b>d</b>) Total ØTCUFN3-specific IgG levels in serum every week. Comparison between the two groups was carried out by area under the curve (AUC). (<b>e</b>) Representative histology of the intestine, liver, and spleen. Immune activation is indicated by the red arrow. The black bracket highlights the magnified section (400× magnification). Scale bar, 100 μm. (<b>f</b>,<b>g</b>) Splenic gene expression of IL-1β (<b>f</b>) and IFN-γ (<b>g</b>). (<b>h</b>,<b>i</b>) Serum levels of IL-1β (<b>h</b>) and IFN-γ (<b>i</b>). Results shown are mean ± S.D. with five mice in each group. * <span class="html-italic">p</span> &lt; 0.05 according to <span class="html-italic">t</span>-test.</p>
Full article ">
25 pages, 6856 KiB  
Article
The Proapoptotic Effect of MB-653 Is Associated with the Modulation of Metastasis and Invasiveness-Related Signalling Pathways in Human Colorectal Cancer Cells
by Libor Sokoli, Peter Takáč, Mariana Budovská, Radka Michalková, Martin Kello, Natália Nosálová, Ľudmila Balážová, Šimon Salanci and Ján Mojžiš
Biomolecules 2025, 15(1), 72; https://doi.org/10.3390/biom15010072 - 6 Jan 2025
Viewed by 517
Abstract
Colorectal cancer is one of the most common cancers worldwide and has a high mortality rate. In this study, we investigated the cytotoxic, proapoptotic, and anti-invasive effects of the synthetic indole phytoalexin MB-653. The antiproliferative effect was determined using an MTT assay, showing [...] Read more.
Colorectal cancer is one of the most common cancers worldwide and has a high mortality rate. In this study, we investigated the cytotoxic, proapoptotic, and anti-invasive effects of the synthetic indole phytoalexin MB-653. The antiproliferative effect was determined using an MTT assay, showing IC50 values of 5.8 ± 0.3 μmol/L for HCT116 cells and 6.1 ± 2.1 μmol/L for Caco2 cells. Flow cytometry and Western blot analysis were employed to investigate the molecular mechanisms underlying cytotoxicity, proapoptotic action, and anti-invasion effects. The proapoptotic activity was evidenced by the activation of caspases 3 and 7, mitochondrial dysfunction, and an increased number of apoptotic cells, confirmed by annexin V/PI and AO/PI staining. Additionally, MB-653 induces dose-dependent G2/M phase cell cycle arrest, the cause of which could be cyclin B1/CDC2 complex dysfunction and/or a decrease in α-tubulin protein expression. Another important observation was that MB-653 modulated several signalling pathways associated with various cellular activities, including survival, proliferation, tumour invasiveness, metastasis, and epithelial–mesenchymal transition (EMT). We further demonstrated its safety for topical and parenteral application. To sum up, our results indicate the real potential of MB-653 in treating colorectal cancer. Full article
Show Figures

Figure 1

Figure 1
<p>MB-653(trans-(±)-N,N’-bis [1-(tert-butoxycarbonyl)-2-methoxy-spiro{indoline-3,5′-[4′,5′]dihydrotriazol-2′-yl}]benzene-1,4-diamine).</p>
Full article ">Figure 2
<p>Western blot analysis of cell cycle-associated proteins affected by MB-653 in HCT116 and Caco2 cells after 24, 48, and 72 h incubation. Representative figure.</p>
Full article ">Figure 3
<p>Densitometric analysis of phospho-Rb (<b>A</b>), phospho-cdc2 (<b>B</b>), Cyclin D1 (<b>C</b>), Cyclin B1 (<b>D</b>), phospho-Cyclin B1 (<b>E</b>) and α-Tubulin (<b>F</b>). Statistical significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 4
<p>Effect of MB-653 on the loss of mitochondrial membrane potential in HCT116 (<b>A</b>) and Caco 2 (<b>B</b>) cells after 24, 48, and 72 h exposure. Data were obtained from 3 independent acquisitions and are presented as the mean ± standard deviation. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 5
<p>Flow cytometric analysis of activated caspase-3/7 after 24, 48, and 72 h of MB-653 treatment in HCT116 (<b>A</b>) and Caco2 (<b>B</b>) cells. Data were obtained from 3 independent acquisitions and are presented as the mean ± standard deviation. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 6
<p>Fluorescence microscopic analysis of MB-653 induced apoptosis in HCT116 (<b>A</b>) and Caco2 (<b>B</b>) cell lines after 24, 48, and 72 h of treatment (10 µmol/L for HCT116 and Caco2) using AO/PI staining. Green shows live cells, yellow shows cells in the initial phase of apoptosis, orange shows cells in the advanced stage of apoptosis, and red indicates dead or necrotic cells. This image is a representative example from three separate experiments. Magnification is set at 100×.</p>
Full article ">Figure 7
<p>Western blot analysis of the effect on proliferation-associated proteins affected by MB-653 in HCT116 and Caco2 cells after 24, 48, and 72 h incubation. Representative figure. Data were obtained from 3 independent acquisitions.</p>
Full article ">Figure 8
<p>Densitometric analysis of NF-κ1 B p50 (<b>A</b>), NF-κ1 B p105 (<b>B</b>), Non-phospho (Active) β-Catenin (<b>C</b>), ß-Catenin (<b>D</b>), phospho-mTOR (<b>E</b>) and mTOR (<b>F</b>). Data were obtained from 3 independent acquisitions and are presented as the mean ± standard deviation. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 9
<p>Western blot analysis of proteins responsible for invasiveness and metastasis affected by MB-653 in HCT116 and Caco2 cells after 24, 48, and 72 h incubation. Representative figure. Data were obtained from 3 independent acquisitions.</p>
Full article ">Figure 10
<p>Densitometric analysis of Snail—1,2,3 (<b>A</b>), MMP-9 (<b>B</b>), N-Cadherin (<b>C</b>) and E-Cadherin (<b>D</b>). Data were obtained from 3 independent acquisitions and are presented as the mean ± standard deviation. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 11
<p>Substance MB-653 in concentration 50 µmol/L has no irritant activity at 30 s, 120 s, and 300 s after application. Data were obtained from 3 independent acquisitions.</p>
Full article ">Figure 12
<p>Measured angiogenesis parameters—vascular zone (<b>A</b>), number of branching points (<b>B</b>), total length (<b>C</b>), average thickness of vessels (<b>D</b>) after application of 10, 25, and 50 μM MB-653 according to software IKOSA. Px—pixels. Data were obtained from 3 independent acquisitions.</p>
Full article ">
19 pages, 1150 KiB  
Article
Limited Changes in Red Blood Cell Parameters After Probiotic Supplementation in Depressive Individuals: Insights from a Secondary Analysis of the PRO-DEMET Randomized Controlled Trial
by Agata Gajewska, Adam Wysokiński, Dominik Strzelecki and Oliwia Gawlik-Kotelnicka
J. Clin. Med. 2025, 14(1), 265; https://doi.org/10.3390/jcm14010265 - 5 Jan 2025
Viewed by 379
Abstract
Background: Depression often coexists with anemia, potentially sharing common pathways, highlighting the need for treatments addressing both conditions simultaneously. This study evaluated the effect of probiotics on red blood cell (RBC) parameters in adults with depressive disorder. We hypothesized that probiotics would [...] Read more.
Background: Depression often coexists with anemia, potentially sharing common pathways, highlighting the need for treatments addressing both conditions simultaneously. This study evaluated the effect of probiotics on red blood cell (RBC) parameters in adults with depressive disorder. We hypothesized that probiotics would positively influence RBC parameters, potentially modulated by baseline inflammation or dietary intake, with improved RBC function correlating with better antidepressant outcomes. Methods: This secondary analysis of a two-arm, randomized, double-blind, controlled trial involved 116 adults with depressive disorder. Participants received a probiotic formulation containing Lactobacillus helveticus Rosell®-52 and Bifidobacterium longum Rosell®-175 or a placebo for 60 days. Data from 97 subjects were analyzed for RBC parameters, including hemoglobin (HGB), RBC count, hematocrit (HCT), mean corpuscular volume (MCV), mean hemoglobin concentration (MCH), mean corpuscular hemoglobin concentration (MCHC), and RBC distribution width (RDW). Results: Probiotic supplementation did not result in significant changes in RBC parameters compared to the placebo. However, probiotics may help stabilize HGB, HCT, MCH, and MCHC levels, potentially preventing fluctuations observed in the placebo group. Conclusions: While probiotics showed potential benefits for depressive symptoms, significant changes in RBC parameters were not observed. Larger studies are needed to clarify the mechanisms and clinical implications. Full article
(This article belongs to the Special Issue Mood Disorders: Diagnosis, Management and Future Opportunities)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The study flow diagram. Abbreviations: PRO—probiotic; PLC—placebo; V2—the end of the intervention period.</p>
Full article ">
16 pages, 1769 KiB  
Article
The Role of Indoxyl Sulfate in Exacerbating Colorectal Cancer During Chronic Kidney Disease Progression: Insights into the Akt/β-Catenin/c-Myc and AhR/c-Myc Pathways in HCT-116 Colorectal Cancer Cells
by Yu Ichisaka, Chihiro Takei, Kazuma Naito, Manami Higa, Shozo Yano, Toshimitsu Niwa and Hidehisa Shimizu
Toxins 2025, 17(1), 17; https://doi.org/10.3390/toxins17010017 - 1 Jan 2025
Viewed by 553
Abstract
Epidemiological studies suggest an increased risk of colorectal cancer (CRC) aggravation in patients with chronic kidney disease (CKD). Our previous study demonstrated that indoxyl sulfate, a uremic toxin whose concentration increases with CKD progression, exacerbates CRC through activation of the AhR and Akt [...] Read more.
Epidemiological studies suggest an increased risk of colorectal cancer (CRC) aggravation in patients with chronic kidney disease (CKD). Our previous study demonstrated that indoxyl sulfate, a uremic toxin whose concentration increases with CKD progression, exacerbates CRC through activation of the AhR and Akt pathways. Consequently, indoxyl sulfate has been proposed to be a significant link between CKD progression and CRC aggravation. The present study aimed to investigate the roles of c-Myc and β-Catenin, which are hypothesized to be downstream factors of indoxyl sulfate-induced AhR and Akt activation, in CRC cell proliferation and EGF sensitivity in HCT-116 CRC cells. Indoxyl sulfate significantly induced CRC cell proliferation at concentrations exceeding 62.5 µM, a process suppressed by the c-Myc inhibitor 10058-F4. Indoxyl sulfate activated the Akt/β-Catenin/c-Myc pathway as evidenced by the Akt inhibitor MK2206, which decreased both β-Catenin and c-Myc protein levels, and the β-Catenin inhibitor XAV-939, which reduced c-Myc protein levels. The AhR antagonist CH223191 also inhibited c-Myc upregulation, indicating involvement of the AhR/c-Myc pathway. MK2206 partially attenuated the indoxyl sulfate-induced AhR transcriptional activity, suggesting that Akt activation influences the AhR/c-Myc pathway. MK2206, CH223191, and 10058-F4 suppressed the increase in EGFR protein levels induced by indoxyl sulfate, indicating that the Akt/β-Catenin/c-Myc and AhR/c-Myc pathways enhance the sensitivity of HCT-116 CRC cells to EGF. These findings indicate that the elevation of indoxyl sulfate levels in the blood, due to CKD progression, could worsen CRC by promoting the proliferation of CRC cells and enhancing EGF signaling. Therefore, indoxyl sulfate could potentially serve as a therapeutic target for CRC aggravation in patients with CKD. Full article
(This article belongs to the Special Issue The Role of Uremic Toxins in Comorbidities of Chronic Kidney Disease)
Show Figures

Figure 1

Figure 1
<p>Impact of skatole on proliferation of HCT-116 CRC cells. Quantification of cell proliferation. The results are presented as the mean values ± SE of quadruplicates from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, vs. 0 h. IS, indoxyl sulfate.</p>
Full article ">Figure 2
<p>Impact of indoxyl sulfate on c-Myc protein levels and subsequent proliferation of HCT-116 CRC cells. (<b>A</b>) The effects of indoxyl sulfate on c-Myc and β-actin protein levels. (<b>B</b>) The band intensity of c-Myc in response to indoxyl sulfate. The band intensity of c-Myc was normalized to that of β-actin. The results are presented as mean values ± SE from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, vs. 0 h. (<b>C</b>) The effect of 10058-F4 on indoxyl sulfate-induced cell proliferation. HCT-116 CRC cells were incubated with (+) or without (−) control (DMSO), indoxyl sulfate, or 10058-F4. The results are presented as the mean values ± SE of quadruplicates from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, vs. control solution (DMSO). <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, vs. indoxyl sulfate alone. IS, indoxyl sulfate.</p>
Full article ">Figure 3
<p>Impact of indoxyl sulfate on Akt, β-Catenin, and c-Myc signaling in HCT-116 CRC cells. HCT-116 CRC cells were incubated with (+) or without (−) control (DMSO), indoxyl sulfate, XAV-939, or MK2206. (<b>A</b>) The effects of XAV-939 on indoxyl sulfate-induced c-Myc and β-actin protein levels. (<b>B</b>) The band intensity of c-Myc in response to indoxyl sulfate alone and in combination with XAV-939. (<b>C</b>) The effects of MK2206 on indoxyl sulfate-induced phospho-GSK3β and β-actin protein levels. (<b>D</b>) The band intensity of phospho-GSK3β in response to indoxyl sulfate alone and in combination with MK2206. (<b>E</b>) The effects of MK2206 on indoxyl sulfate-induced β-Catenin and β-actin protein levels. (<b>F</b>) The band intensity of β-Catenin in response to indoxyl sulfate alone and in combination with MK2206. (<b>G</b>) The effects of MK2206 on indoxyl sulfate-induced c-Myc and β-actin protein levels. (<b>H</b>) The band intensity of c-Myc in response to indoxyl sulfate alone and in combination with MK2206. The band intensities of β-Catenin, phospho-GSK3β, and c-Myc were normalized to that of β-actin. The results are presented as mean values ± SE from three independent experiments for for (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>). * <span class="html-italic">p</span> &lt; 0.05, vs. indoxyl sulfate alone. IS, indoxyl sulfate; pGSK3β, phospho-GSK3β.</p>
Full article ">Figure 4
<p>Impact of AhR on c-Myc and β-Catenin protein levels triggered by indoxyl sulfate in HCT-116 CRC cells. HCT-116 CRC cells were incubated with (+) or without (−) control (DMSO), indoxyl sulfate, or CH223191. (<b>A</b>) The effects of CH223191 on indoxyl sulfate-induced c-Myc and β-actin protein levels. (<b>B</b>) The band intensity of c-Myc in response to indoxyl sulfate alone and in combination with CH223191. (<b>C</b>) The effects of CH223191 on indoxyl sulfate-induced β-Catenin and β-actin protein levels. (<b>D</b>) The band intensity of β-Catenin in response to indoxyl sulfate alone and in combination with CH223191. The band intensities of c-Myc and β-Catenin were normalized to that of β-actin. The results are presented as mean values ± SE from three independent experiments for (<b>B</b>,<b>D</b>). * <span class="html-italic">p</span> &lt; 0.05, vs. indoxyl sulfate alone. IS, indoxyl sulfate.</p>
Full article ">Figure 5
<p>Impact of Akt on AhR transactivation triggered by indoxyl sulfate in HCT-116 CRC cells. The effects of MK2206 on indoxyl sulfate-induced increases in AhR transcriptional activity. HCT-116 CRC cells were incubated with (+) or without (−) control (DMSO), indoxyl sulfate, or MK2206. The results are presented as mean values ± SE of duplicates from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, vs. the control solution (DMSO). <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, vs. indoxyl sulfate alone. IS, indoxyl sulfate.</p>
Full article ">Figure 6
<p>Impact of indoxyl sulfate on EGFR expression and enhanced EGF sensitivity in HCT-116 CRC cells. HCT-116 CRC cells were incubated with (+) or without (−) control (DMSO), indoxyl sulfate, EGF, MK2206 or CH223191. (<b>A</b>) The effects of MK2206 on indoxyl sulfate-induced EGFR and β-actin protein levels. (<b>B</b>) The band intensity of EGFR in response to indoxyl sulfate alone and in combination with MK2206. (<b>C</b>) The effects of CH223191 on indoxyl sulfate-induced EGFR and β-actin protein levels. (<b>D</b>) The band intensity of EGFR in response to indoxyl sulfate alone and in combination with CH223191. (<b>E</b>) The effects of indoxyl sulfate on EGF-induced Akt phosphorylation and β-actin protein levels. (<b>F</b>) The band intensity of phospho-Akt in response to EGF alone and EGF after pretreatment with indoxyl sulfate. (<b>G</b>) The effects of 10058-F4 on indoxyl sulfate-induced EGFR and β-actin protein levels. (<b>H</b>) The band intensity of EGFR in response to indoxyl sulfate alone and in combination with 10058-F4. The band intensities of EGFR and phospho-Akt were normalized to that of β-actin. The results are presented as mean values ± SE from three independent experiments for (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>). * <span class="html-italic">p</span> &lt; 0.05, vs. indoxyl sulfate alone. IS, indoxyl sulfate; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor.</p>
Full article ">Figure 7
<p>Schematic showing potential regulatory pathways by which indoxyl sulfate may exacerbate colorectal cancer (CRC) as chronic kidney disease (CKD) progresses. Indoxyl sulfate, a uremic toxin that accumulates in the blood of patients with CKD, induces proliferation of HCT-116 CRC cells and enhances their sensitivity to epidermal growth factor (EGF). This occurs through the activation of two distinct signaling pathways: the Akt/β-Catenin/c-Myc pathway and the AhR/c-Myc pathway. Activation of these pathways leads to the increased expression of c-Myc, a key oncogene involved in CRC development and progression. Furthermore, indoxyl sulfate increased EGFR protein levels via c-Myc-dependent mechanisms, thereby enhancing the responsiveness of CRC cells to EGF. These findings suggest that the accumulation of indoxyl sulfate in patients with CKD may contribute to the exacerbation of CRC by inducing CRC cell proliferation and augmenting EGF signaling. Targeting indoxyl sulfate may represent a potential therapeutic strategy for managing CRC in patients with concomitant CKD. AhR, aryl hydrocarbon receptor; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor.</p>
Full article ">
18 pages, 4796 KiB  
Article
Exploring the Chemopreventive Potential of Artemisia annua Methanolic Extract in Colorectal Cancer Induced by Azoxymethane in Mice
by Faris Alrumaihi
Pharmaceuticals 2025, 18(1), 34; https://doi.org/10.3390/ph18010034 - 31 Dec 2024
Viewed by 441
Abstract
Background/Objectives: Colorectal cancer (CRC) remains a major global health burden, necessitating innovative preventive approaches. Artemisia annua (A. annua), known for its extensive pharmacological properties, has shown potential in cancer therapy. This study investigates the chemopreventive efficacy of methanolic extract of A. [...] Read more.
Background/Objectives: Colorectal cancer (CRC) remains a major global health burden, necessitating innovative preventive approaches. Artemisia annua (A. annua), known for its extensive pharmacological properties, has shown potential in cancer therapy. This study investigates the chemopreventive efficacy of methanolic extract of A. annua (MEA) in an azoxymethane (AOM)-induced murine model of CRC, with a focus on its antioxidant, biomarker modulation, and pro-apoptotic activities. Methods: MEA was obtained via cold solvent extraction, yielding 39%, and demonstrated potent in vitro cytotoxicity against HCT116 and RKO colon cancer cell lines, with IC50 values of 20 µg/mL and 15 µg/mL, respectively. Swiss albino mice were treated with MEA beginning two weeks before AOM induction, with treatment continuing for 21 weeks. Survival was monitored for 40 weeks. Key outcomes included serum biomarker levels (ADA, GGT, CD73, LDH), antioxidant enzyme activities (SOD, CAT, GPx1, MDA), reactive oxygen species (ROS) modulation, apoptosis induction, and histopathological evaluation. Results: MEA significantly improved survival rates, reduced AOM-induced weight loss, and modulated cancer biomarkers, with marked reductions in ADA, GGT, CD73, and LDH levels. Antioxidant defenses were restored, as evidenced by increased SOD, CAT, and GPx1 activities and decreased MDA levels. ROS levels were significantly reduced, and apoptosis in colonic cells was effectively induced. Histopathological analysis revealed substantial mitigation of CRC-associated morphological abnormalities. Conclusions: MEA exhibits robust chemopreventive properties, demonstrating its potential to reduce oxidative stress, modulate key biomarkers, and induce apoptosis in CRC. These findings position MEA as a promising natural candidate for CRC prevention and therapy, warranting further exploration for clinical application. Full article
(This article belongs to the Special Issue Therapeutic Effects of Natural Products and Their Clinical Research)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chromatogram of bioactive compounds identified in the methanolic extract of <span class="html-italic">Artemisia annua</span> (MEA).</p>
Full article ">Figure 2
<p>Assessment of MEA’s concentration-dependent impact on cell proliferation. This figure illustrates the determination of the half-maximal inhibitory concentration (IC<sub>50</sub>) of MEA on the proliferation of HCT116 and RKO colon cancer cell lines. The evaluation was conducted using a cell cytotoxicity assay over 48 h, with varying concentrations of MEA.</p>
Full article ">Figure 3
<p>Analysis of MEA’s influence on carcinogen-induced effects. This figure illustrates (<b>A</b>) the changes in average body weight (ABW) and (<b>B</b>) the survival rates under the influence of MEA. Part (<b>A</b>) data represent the standard error of the mean (SEM) from a sample of five animals per group. For part (<b>B</b>), the survival data are based on a sample size of ten mice per group. ’ns’ denotes no significant difference. The symbol ‘*’ signifies a statistically significant difference between groups at a <span class="html-italic">p</span>-value less than 0.05, while ‘****’ represents a more pronounced statistical significance, indicating a <span class="html-italic">p</span>-value of less than 0.0001.</p>
Full article ">Figure 4
<p>Analysis of MEA’s impact on serum levels of AOM-induced cancer marker enzymes. This figure presents the effects of MEA on (<b>A</b>) adenosine deaminase (ADA), (<b>B</b>) aryl hydrocarbon hydroxylase (AHH), (<b>C</b>) gamma-glutamyl transferase (GGT), (<b>D</b>) 5′-nucleotidase (5-NT/CD73), and (<b>E</b>) lactate dehydrogenase (LDH) in serum. Data are expressed as the SEM derived from three independent experiments. ’ns’ indicates no significant difference within the treated groups. Symbols ‘*’, ‘**’, ‘***’, and ‘****’ denote statistically significant differences between groups with <span class="html-italic">p</span>-values less than 0.05, 0.01, 0.001, and 0.0001, respectively.</p>
Full article ">Figure 5
<p>Assessment of MEA’s influence on antioxidant enzymes in colon tissue. This figure depicts the effects of MEA on various antioxidant enzymes, including (<b>A</b>) superoxide dismutase (SOD), (<b>B</b>) catalase (CAT), (<b>C</b>) malondialdehyde (MDA), and (<b>D</b>) glutathione peroxidase 1 (GPx1). Data are expressed as the SEM based on results from three separate experiments. ‘ns’ denotes no significant differences within the groups. Symbols ‘**’, ‘***’, and ‘****’ indicate statistically significant differences within the groups, corresponding to <span class="html-italic">p</span>-values of less than 0.05, 0.01, 0.001, and 0.0001, respectively.</p>
Full article ">Figure 6
<p>Evaluation of MEA’s impact on cellular reactive oxygen species (ROS) in colon cells. This figure demonstrates the effects of MEA on cellular ROS levels, as measured by 2′,7′-dichlorofluorescein diacetate (DCFDA) assay and analyzed using flow cytometry. Data are presented as the standard error of the mean (SEM) based on three independent experiments. ‘ns’ indicates no significant difference within the groups. The ‘****’ symbol represents a statistically significant difference between the groups, with a <span class="html-italic">p</span>-value of less than 0.0001.</p>
Full article ">Figure 7
<p>Analysis of MEA’s effect on apoptosis induction in colon cells. This figure illustrates the impact of MEA on apoptosis in colon cells, as determined by annexin V-FITC and propidium iodide (PI) staining, with results analyzed using flow cytometry. Data are represented as the standard error of the mean (SEM) compiled from three experiments. ‘ns’ signifies no significant difference within the groups. The symbols ‘*’, ‘**’, and ‘****’ denote statistically significant differences between groups, with a <span class="html-italic">p</span>-value of less than 0.05, 0.01, and 0.0001, respectively.</p>
Full article ">Figure 8
<p>Histopathological analysis of MEA’s effect on AOM-induced colorectal carcinogenesis. Representative histopathological images of colon tissue showing the effects of MEA on AOM-induced carcinogenesis. Images are displayed at 100× magnification, with a scale bar indicating 100 µm.</p>
Full article ">Figure 9
<p>In vivo study design overview. Group 1 (control): Administered Phosphate-Buffered Saline (PBS) orally thrice weekly from week −2 to week 21. Group 2: Received azoxymethane (AOM) at a dose of 10 mg/kg body weight in 200 µL PBS, administered intraperitoneally three times weekly from week 0 to week 6. Group 3 (MEA Low Dose): Treated with MEA at 10 mg/kg in PBS following the schedule of Group 1, combined with AOM treatment as in Group 2. Group 4 (MEA High Dose): Received MEA at 20 mg/kg in PBS, with the Group 1 + AOM regimen as in Group 2. Group 5: Treated with a high dose of MEA (20 mg/kg) following the Group 1 protocol. PBS and MEA were given orally to all groups, while AOM was administered intraperitoneally.</p>
Full article ">
17 pages, 1676 KiB  
Article
Evolution of Nutritional Status in Patients Undergoing Autologous and Allogeneic Hematopoietic Cell Transplantation or CAR-T Therapy: A Retrospective Observational Study
by Roberto Regazzoni, Sergio Ferrante, Emanuela Morenghi, Diego Lopane, Manuela Pastore, Daniela Cattani, Simone Cosmai, Francesco Colotta, Elena Azzolini, Marco Sguanci, Giovanni Cangelosi, Luca Cozzaglio, Beatrice Mazzoleni and Stefano Mancin
Cancers 2025, 17(1), 79; https://doi.org/10.3390/cancers17010079 - 30 Dec 2024
Viewed by 358
Abstract
Background/Objectives: Hematopoietic cell transplantation (HCT) is a curative treatment for various hematological diseases but can lead to complications which increase malnutrition risk, particularly in allogeneic transplantation patients. This study evaluates the nutritional status evolution of patients undergoing HCT during hospitalization and follow-up. Methods: [...] Read more.
Background/Objectives: Hematopoietic cell transplantation (HCT) is a curative treatment for various hematological diseases but can lead to complications which increase malnutrition risk, particularly in allogeneic transplantation patients. This study evaluates the nutritional status evolution of patients undergoing HCT during hospitalization and follow-up. Methods: This retrospective observational study included 365 patients, divided into two groups: 134 underwent allogeneic HCT, while 231 underwent autologous transplantation or CAR-T therapy. Nutritional status was evaluated using Body Mass Index (BMI), Malnutrition Universal Screening Tool (MUST), and Global Leadership Initiative on Malnutrition (GLIM) criteria at four-time points: hospital admission, discharge, two-week follow-up, and one-month follow-up. Non-relapse-related complications were assessed based on hospital readmissions and reports during follow-up visits. Results: Patients experienced significant nutritional deterioration, with decreases in Body Mass Index (BMI) (p < 0.001) and increases in Malnutrition Universal Screening Tool (MUST) (p < 0.001) and Global Leadership Initiative on Malnutrition (GLIM) scores (p < 0.001), particularly among allogeneic transplant recipients (p = 0.025). Severe malnutrition or high malnutrition risk at discharge correlated with increased hospital readmissions during the follow-up (p = 0.024). Conclusions: The observed decline in nutritional status and its associated complications highlight the necessity of multidisciplinary interventions, such as nutritional prehabilitation programs and nutritional support protocols, to enhance clinical outcomes and reduce complications in HCT patients. Full article
Show Figures

Figure 1

Figure 1
<p>BMI variation over time. Legend: <span class="html-italic">p</span>: <span class="html-italic">p</span>-value; BMI: Body Mass Index; T0: upon hospital admission; T1: at hospital discharge; T2: two weeks post-discharge follow-up; T3: one-month post-discharge follow-up.</p>
Full article ">Figure 2
<p>Evolution of BMI over time for patients grouped into two cohorts. Legend: CAR-T: chimeric antigen receptor T cells; BMI: Body Mass Index; T0: upon hospital admission; T1: at hospital discharge; T2: two weeks post-discharge follow-up; T3: one-month post-discharge follow-up.</p>
Full article ">Figure 3
<p>Evolution of BMI over time for patients grouped by diabetes mellitus diagnosis. Legend: CAR-T: chimeric antigen receptor T cells; BMI: Body Mass Index; T0: upon hospital admission; T1: at hospital discharge; T2: two weeks post-discharge follow-up; T3: one-month post-discharge follow-up.</p>
Full article ">Figure 4
<p>Evolution of BMI over time for patients grouped by transplantation type. Legend: CAR-T: chimeric antigen receptor T cells; BMI: Body Mass Index; T0: upon hospital admission; T1: at hospital discharge; T2: two weeks post-discharge follow-up; T3: one-month post-discharge follow-up.</p>
Full article ">
15 pages, 5522 KiB  
Article
Cell Wall Protein 2 as a Vaccine Candidate Protects Mice Against Clostridioides difficile Infection
by Shaohui Wang, Joshua Heuler, Jessica Bullock, Junling Qin, Soumyadeep Chakraborty, Agbendeh Lubem Nathaniel, Shifeng Wang and Xingmin Sun
Vaccines 2025, 13(1), 21; https://doi.org/10.3390/vaccines13010021 - 30 Dec 2024
Viewed by 455
Abstract
Background/Objectives: Clostridioides difficile is a Gram-positive, spore-forming enteric pathogen that causes intestinal disorders, including inflammation and diarrhea, primarily through toxin production. Standard treatment options for C. difficile infection (CDI) involve a limited selection of antibiotics that are not fully effective, leading to high [...] Read more.
Background/Objectives: Clostridioides difficile is a Gram-positive, spore-forming enteric pathogen that causes intestinal disorders, including inflammation and diarrhea, primarily through toxin production. Standard treatment options for C. difficile infection (CDI) involve a limited selection of antibiotics that are not fully effective, leading to high recurrence rates. Vaccination presents a promising strategy for preventing both CDI and its recurrence. Cell wall protein 2 (Cwp2), a highly immunogenic and abundant surface-exposed C. difficile cell wall protein, plays an important role in the bacterium’s adherence in vitro. In this study, we aimed to analyze the homology and immunogenicity of Cwp2 and its protection efficacy as a vaccine candidate against CDI in mice. Methods: we conducted in silico analyses to assess the homology and immunogenicity of Cwp2, and we evaluated its potential as a vaccine candidate against CDI using a mouse model of immunization and infection. Results: Our in silico analyses predicted the immunogenic region (functional domain) of Cwp2 and revealed its high homology among various toxinotypes and ribotypes (R.T.s) or sequence types (S.T.s). Immunizations of mice with the Cwp2 functional domain (Cwp2_A) induced potent IgG/A antibody responses against Cwp2_A, protected mice from CDI, and reduced C. difficile spore and toxin levels in feces post-infection. Additionally, anti-Cwp2_A sera inhibited the binding of C. difficile vegetative cells to HCT8 cells. Conclusions: Our report demonstrates for the first time the potential of Cwp2_A as an effective vaccine candidate against CDI in mice. Full article
Show Figures

Figure 1

Figure 1
<p><b>Domain architecture of cell wall protein 2 (Cwp2) (WP_009891054.1) from <span class="html-italic">C. difficile</span> R20291.</b> The signal peptide (SP) is followed by the functional region, which includes domain 1 (D1), domain 2 (D2), and domain 3 (D3); D2 is connected to D3 via a strand of 13 aa in D1. The cell wall binding domain (CWB) has 3 repeated regions, CWB1, CWB2, and CWB3, as indicated in UniProt. The schematic representation of the domain architecture was developed in DOG 2.0.</p>
Full article ">Figure 2
<p><b>Predicted immunogenic regions (in yellow) of Cwp2.</b> B cell epitopes of Cwp2 were predicted using the BepiPred-2.0 server (<a href="https://www.iedb.org/" target="_blank">https://www.iedb.org/</a>; accessed on 6 May 2023). The residues with scores above the threshold (default value is 0.5) are predicted to be part of an epitope and are colored in yellow on the graph (where the Y-axis depicts residue scores and the X-axis depicts residue positions in the sequence).</p>
Full article ">Figure 3
<p><b>Cwp2 phylogeny.</b> Amino acid sequences of Cwp2 were aligned with the MUSCLE algorithm in MegaX before computing a maximum likelihood tree with 100 bootstrap replicates (bootstrap values &gt;50 are displayed). Scale bars indicate 0.010 substitutions per site. The ribotype (or sequence type) of each source strain is displayed adjacent to the strain name. Ribotypes with multiple representatives have multicolored labels, while black labels indicate ribotypes with only one representative on the tree. Toxinotypes for each strain are indicated to the right of the ribotype for each strain.</p>
Full article ">Figure 4
<p><b>Cwp2 homology.</b> Cwp2 sequences were aligned with MUSCLE and visualized with Jalview. The Jalview-calculated conservation scores are reported below the alignment from 0 (no conservation) to 11 (identical sequences). The ribotype of each source strain is displayed adjacent to the strain name and are color-coded for easier identification. The conserved amino acid sequences are highlighted in blue.</p>
Full article ">Figure 5
<p>(<b>A</b>) Expression and purification of Cwp2_A. Cwp2_A was cloned in <span class="html-italic">E. Coli</span> BL21, and the protein was purified and analyzed on SDS-PAGE. (<b>B</b>) Immunization with Cwp2_A via the intraperitoneal (i.p.) route elicited anti-Cwp2_A antibody responses. Groups of mice (<span class="html-italic">n</span> = 5–8) were immunized three times with 10 µg or 20 µg of Cwp2_A with aluminum. In (<b>C</b>–<b>E</b>), 20 µg of protein was used. Anti-Cwp2_A IgG/IgA titers in sera and feces were determined using an ELISA analysis. Data are presented as the mean ± SEM (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns, not significant; 2nd and 3rd IM vs. 1st IM in (<b>C</b>–<b>E</b>)).</p>
Full article ">Figure 6
<p><b>Immunization with Cwp2_A provides mice significant protection against infection with <span class="html-italic">C. difficile</span>.</b> Immunized mice or controls (non-immunized mice) (<span class="html-italic">n</span> = 10) were challenged with <span class="html-italic">C. difficile</span> R20291 spores (10<sup>6</sup>/mouse). Survivals (<b>A</b>), weight changes (<b>B</b>), and diarrhea percentages (<b>C</b>) are shown. Data are presented as the mean ± SEM (ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; in (B), immunization with 20 µg vs. control).</p>
Full article ">Figure 7
<p><b>Immunization of with Cwp2_A reduces <span class="html-italic">C. difficile</span> spore and toxin levels in feces of <span class="html-italic">C. difficile</span> R20291-infected mice.</b> <span class="html-italic">C. difficile</span> toxin (<b>A</b>,<b>B</b>) and spore (<b>C</b>) levels in feces were determined. Data are presented as the mean ± SEM. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus control).</p>
Full article ">Figure 8
<p><b>Anti-Cwp2_A serum inhibits adhesion of <span class="html-italic">C. difficile</span> to HCT8 cells.</b> The adhesion assay was performed as described in the methods. Experiments were independently repeated three times, and data are presented as the mean ± SEM (* <span class="html-italic">p</span> &lt; 0.05 vs. treatment with pre-immune serum).</p>
Full article ">Figure 9
<p><b>T cell responses.</b> Splenocytes from immunized (<span class="html-italic">n</span> = 3–4) and unimmunized (<span class="html-italic">n</span> = 4) mice were isolated 13 days after the second immunization with Cwp2_A and stimulated with Cwp2_A at 10 µg/mL for 72 h (<b>A</b>,<b>B</b>) or 6 h (<b>C</b>). The proliferative responses of CD4+ (<b>A</b>) and CD8+ (<b>B</b>) T cells were assayed by staining with appropriate antibodies and were analyzed by flow cytometry. (<b>C</b>) IL-17, IFN-γ, and TNF-α expression in the spleen cells was determined by qPCR processing. The y-axis value indicates the expression ratio relative to GAPDH. Data are presented as the mean ± SEM (N = 3, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns, not significant).</p>
Full article ">
22 pages, 1863 KiB  
Article
Antioxidant, Antiproliferative, Antibacterial, and Antimalarial Effects of Phenolic-Rich Green Tea Kombucha
by Udielle Vermelho Lacerda, Carolina Vargas Pereira da Costa, Rodrigo Rezende Cardoso, Carolina Thomaz dos Santos D’Almeida, Mariana Araújo Vieira do Carmo, Amanda dos Santos Lima, Laura da Silva Cruz, Amanda Bubula de Souza, Philipe Oliveira Fernandes, Vinícius Gonçalves Maltarollo, Mariana Simões Larraz Ferreira, Luciana Azevedo, Monique Renon Eller, Viviana Corich, Alessio Giacomini and Frederico Augusto Ribeiro de Barros
Beverages 2025, 11(1), 7; https://doi.org/10.3390/beverages11010007 - 30 Dec 2024
Viewed by 629
Abstract
Green tea kombucha, produced using a green tea (Camellia sinensis) grown in Brazil, was characterized and its in vitro bioactive properties were evaluated. Overall, 92 phenolic compounds were identified (70.7% flavonoids, 25% phenolic acids, 2.2% lignans, and 1.1% other polyphenols), contributing [...] Read more.
Green tea kombucha, produced using a green tea (Camellia sinensis) grown in Brazil, was characterized and its in vitro bioactive properties were evaluated. Overall, 92 phenolic compounds were identified (70.7% flavonoids, 25% phenolic acids, 2.2% lignans, and 1.1% other polyphenols), contributing to the observed high antioxidant capacity. The major phenolics identified were gallocatechin, catechin 5-O-gallate, and epicatechin. Green tea kombucha exhibited antibacterial activity against all tested bacteria, being more effective against Salmonella spp. In addition, green tea kombucha demonstrated antimalarial activity against both chloroquine-sensitive and chloroquine-resistant strains of Plasmodium falciparum, and antiproliferative activity against cancer cell lines A549, HCT8, HepG2, and HUVEC. Additionally, it presented antioxidant properties by effectively reducing the generation of reactive oxygen species (ROS) and provided protection to erythrocytes against AAPH-induced oxidative stress. Thus, green tea kombucha is abundant in antioxidants and possesses intriguing bioactive properties that can be investigated by both the food and pharmaceutical sectors. Full article
Show Figures

Figure 1

Figure 1
<p>Raw chromatogram of the ten most abundant phenolic compounds found in green tea kombucha. <b>1</b> (+)-gallocatechin 3-O-gallate/(-)-epigallocatechin 3-O-gallate; <b>2</b> (+)-gallocatechin; <b>3</b> Catechin 5-O-gallate; <b>4</b> (-)-epicatechin; <b>5</b> (-)-epigallocatechin; <b>6</b> 5-O-Galloylquinic acid; <b>7</b> catechin; <b>8</b> 5-p-coumaroylquinic acid; <b>9</b> quercetin 3-O-rutinoside; and <b>10</b> myricetin 3-O-glucoside.</p>
Full article ">Figure 2
<p>(<b>A</b>) IC<sub>50</sub> (50% cell viability inhibition); (<b>B</b>) GI<sub>50</sub> (50% growth inhibition); and (<b>C</b>) LC<sub>50</sub> (50% cell death) of green tea kombucha.</p>
Full article ">Figure 3
<p>Intracellular ROS measurement in HCT8 (<b>A</b>) and HUVEC (<b>B</b>) cells by spectrofluorimetry that received treatment with kombucha extract at 10, 50, and 100 μg/mL. Quantitative data are the mean ± standard deviation (n = 4). Different letters represent statistically significant differences (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 4
<p>(<b>A</b>) Effect of kombucha extract on blood hemolysis induced by 2′-azobis(2-amidinopropane) dihydrochloride (AAPH); (<b>B</b>) effect of kombucha extract on hemoglobin oxidation induced by AAPH; and (<b>C</b>) effect of kombucha extract on intracellular reactive oxygen species (ROS) induced by AAPH. Note: NC: negative control; AAPH: positive control; Q: quercetin. Quantitative data are the mean ± standard deviation (n = 4). Different letters represent statistically significant differences (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 5
<p>Antiplasmodial activity and cytotoxicity of kombucha phenolic extract, against chloroquine-resistant strain (W2) and chloroquine-sensitive strain (3D7).</p>
Full article ">
18 pages, 4974 KiB  
Article
MOMAST® Downregulates AQP3 Expression and Function in Human Colon Cells
by Ines Angelini, Mariangela Centrone, Giusy Rita Caponio, Annarita Di Mise, Andrea Gerbino, Marianna Ranieri, Giovanna Valenti and Grazia Tamma
Antioxidants 2025, 14(1), 26; https://doi.org/10.3390/antiox14010026 - 28 Dec 2024
Viewed by 371
Abstract
The water channel AQP3 is an aquaglyceroporin expressed in villus epithelial cells, and it plays a role in water transport across human colonic surface cells. Beyond water, AQP3 can mediate glycerol and H2O2 transport. Abnormal expression and function of AQP3 [...] Read more.
The water channel AQP3 is an aquaglyceroporin expressed in villus epithelial cells, and it plays a role in water transport across human colonic surface cells. Beyond water, AQP3 can mediate glycerol and H2O2 transport. Abnormal expression and function of AQP3 have been found in various diseases often characterized by altered cell growth and proliferation. Here, the beneficial effects of MOMAST® have been evaluated. MOMAST® is an antioxidant-patented natural phenolic complex obtained from olive wastewater (OWW) of the Coratina cultivar. Treatment of human colon HCT8 cells with MOMAST® reduced cell viability. Confocal studies and Western Blotting analysis demonstrated that treatment with MOMAST® significantly decreased the staining and the expression of AQP3. Importantly, functional studies revealed that the reduction of AQP3 abundance correlates with a significant decrease in glycerol and H2O2 uptake. Indeed, the H2O2 transport was partially but significantly reduced in the presence of MOMAST® or DFP00173, a selective inhibitor of AQP3. In addition, the MOMAST®-induced AQP3 decrease was associated with reduced epithelial-mesenchymal transition (EMT)-related proteins such as vimentin and β-catenin. Together, these findings propose MOMAST® as a potential adjuvant in colon diseases associated with abnormal cell growth by targeting AQP3. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>ROS content in HCT8 cells exposed to MOMAST<sup>®</sup>. ROS content was measured using dihydrorhodamine-123 fluorescence in cells left under basal conditions or treated with MOMAST<sup>®</sup> as described in Methods. As a positive control, cells were treated with the oxidant <span class="html-italic">tert</span>-Butyl hydroperoxide (tBHP). Untreated cells (CTR) are a blank control. Data are shown as means ± Standard Error Means (S.E.M.) of six independent experiments and analyzed with one-way ANOVA followed by Dunnett’s multiple comparisons test (<b>****</b> <span class="html-italic">p</span> &lt; 0.0001 vs. CTR; <b><sup>##</sup></b> <span class="html-italic">p</span> &lt; 0.01 vs. tBHP; <b><sup>####</sup></b> <span class="html-italic">p</span> &lt; 0.0001 vs. tBHP).</p>
Full article ">Figure 2
<p>Cell viability in HCT8 cells exposed to MOMAST<sup>®</sup>. Cells were left under basal conditions or exposed to increasing concentrations (250, 500, and 1000 µg/mL) of MOMAST<sup>®</sup> as described in Methods. As a positive internal control, cells were treated with 90% ethanol for 1 min (CTR+). Data are shown as means ± Standard Error Means (S.E.M.) of seven independent experiments and analyzed with one-way ANOVA followed by Dunnett’s multiple comparisons test (* <span class="html-italic">p</span> &lt; 0.05 vs. CTR; *** <span class="html-italic">p</span> &lt; 0.001 vs. CTR; **** <span class="html-italic">p</span> &lt; 0.0001 vs. CTR).</p>
Full article ">Figure 3
<p>(<b>a</b>) Real-time PCR analysis of AQP3 mRNA expression levels. MOMAST<sup>®</sup> treatment did not alter the AQP3 transcript levels in HCT8 cells. Data are shown as means ± Standard Error Means (S.E.M.) of three independent experiments and analyzed with one-way ANOVA followed by Dunnett’s multiple comparisons test. (<b>b</b>) Confocal immunofluorescence visualization of AQP3 in HCT8 cells. Cells were left under basal conditions or exposed to increasing concentrations (250, 500, and 1000 µg/mL) of MOMAST<sup>®</sup> as described in Methods. Confocal images show a slight but relevant reduction in AQP3 fluorescence signal.</p>
Full article ">Figure 4
<p>Effect of MOMAST<sup>®</sup> on AQP3 protein expression. Treatments with MOMAST<sup>®</sup> at increasing concentrations induced a significant reduction in the AQP3 protein expression. Data are shown as means ± Standard Error Means (S.E.M.) of 5 independent experiments and analyzed by one-way ANOVA followed by Dunnett’s multiple comparisons test (* <span class="html-italic">p</span> &lt; 0.05 vs. CTR; ** <span class="html-italic">p</span> &lt; 0.01 vs. CTR).</p>
Full article ">Figure 5
<p>Evaluation of the HyPer-3 sensitivity to H<sub>2</sub>O<sub>2</sub> exposure by analyzing the HyPer-3 fluorescence ratio. (<b>a</b>) A representative trace showing the HyPer-3 fluorescence ratio in transfected HCT8 cells stimulated with external H<sub>2</sub>O<sub>2</sub> at increasing concentrations (25, 50, and 100 μM). (<b>b</b>) A significant increase in normalized HyPer-3 fluorescence ratio intensity was recorded at 50 and 100 μM H<sub>2</sub>O<sub>2</sub> external stimuli. Data are shown as means ± Standard Error Means (S.E.M.) of six independent experiments and analyzed with one-way ANOVA followed by Dunnett’s multiple comparisons test (** <span class="html-italic">p</span> &lt; 0.01 vs. basal; **** <span class="html-italic">p</span> &lt; 0.0001 vs. basal).</p>
Full article ">Figure 6
<p>Evaluation of the HyPer-3 sensitivity to H<sub>2</sub>O<sub>2</sub> exposure by analyzing the HyPer-3 fluorescence ratio in the absence or in the presence of the AQP3 inhibitor, DFP00173. (<b>a</b>) A representative trace showing the HyPer-3 fluorescence ratio in HCT8 cells stimulated with external 50 μM H<sub>2</sub>O<sub>2</sub> in the absence and the presence of 5 μM DFP00173. (<b>b</b>) The histogram shows the normalized HyPer-3 fluorescence ratio. The normalized HyPer-3 fluorescence ratio was measured by calculating the fluorescence ratio at the peak compared to the starting fluorescence values. Data are shown as means ± Standard Error Means (S.E.M.) of five independent experiments and analyzed by paired <span class="html-italic">t</span> test (** <span class="html-italic">p</span> &lt; 0.01 vs. 50 µM H<sub>2</sub>O<sub>2</sub>).</p>
Full article ">Figure 7
<p>Live-cell imaging of changes in HyPer-3 fluorescence in HCT8 cells. (<b>a</b>) Representative traces showing the HyPer-3 fluorescence ratio in HCT8 cells treated with MOMAST<sup>®</sup>. (<b>b</b>) The histogram shows the normalized HyPer-3 fluorescence ratio where the normalized HyPer-3 ratio was calculated by evaluating the fluorescence ratio at the peak compared to the starting fluorescence values. Data are shown as means ± Standard Error Means (S.E.M.) of five independent experiments and analyzed by one-way ANOVA followed by Dunnett’s multiple comparisons test (* <span class="html-italic">p</span> &lt; 0.05 vs. 50 µM H<sub>2</sub>O<sub>2</sub>).</p>
Full article ">Figure 8
<p>Effect of MOMAST<sup>®</sup> on glycerol permeability. (<b>a</b>) Representative time courses of cell shrinking (water exit) followed by cell swelling indicative of the osmotic influx of water promoted by glycerol entry along its gradient (Δ 100 mOsm/L). (<b>b</b>) A bar plot showing the means ± Standard Error Means (S.E.M.) values of the cell swelling time constants (slope) reflecting glycerol entry into the cells. Data are obtained from 91 different measurements of 4 independent experiments. A one-way ANOVA and Dunnett’s Multiple Comparison test were performed (**** <span class="html-italic">p</span> &lt; 0.0001 vs. CTR).</p>
Full article ">Figure 9
<p>Effect of MOMAST<sup>®</sup> on EMT markers expression in HCT8 cells. Treatments with MOMAST<sup>®</sup> at increasing concentrations induced a significant reduction in the EMT proteins expression β-catenin (<b>a</b>) and vimentin (<b>b</b>) likely suggesting a possible role of MOMAST<sup>®</sup> in modulating the expression level of selective EMT protein markers. Data are shown as means ± Standard Error Means (S.E.M.) of four independent experiments and analyzed with one-way ANOVA followed by Dunnett’s multiple comparisons test (* <span class="html-italic">p</span> &lt; 0.05 vs. CTR; ** <span class="html-italic">p</span> &lt; 0.01 vs. CTR; *** <span class="html-italic">p</span> &lt; 0.001 vs. CTR).</p>
Full article ">Scheme 1
<p>Flowchart showing the experimental protocol applied in the present study.</p>
Full article ">Scheme 2
<p>Model depicting the effects of MOMAST<sup>®</sup> exerted in HCT8 cells. Specifically, MOMAST<sup>®</sup>-reduced AQP3 expression and function and affect the expression level of the EMT protein markers, vimentin and β-catenin. The downward arrow indicates a reduction.</p>
Full article ">
6 pages, 193 KiB  
Brief Report
Matched Unrelated Donor Hematopoietic Cell Transplantation: Increased Usage and Improvements in Clinical Outcomes in Canada
by Matthew D. Seftel, Grace Musto, David Allan, Oliver Bucher, Kevin Hay, Ivan Pasic, Tony Truong and Kristjan Paulson
Curr. Oncol. 2025, 32(1), 10; https://doi.org/10.3390/curroncol32010010 - 27 Dec 2024
Viewed by 306
Abstract
In allogeneic hematopoietic cell transplantation (HCT), a minority of patients have access to a suitable human leukocyte antigen (HLA)-matched related donor (MRD). To fill this gap, matched unrelated donors (MUDs) are an increasingly selected donor source. Usage and outcomes after MUD HCT for [...] Read more.
In allogeneic hematopoietic cell transplantation (HCT), a minority of patients have access to a suitable human leukocyte antigen (HLA)-matched related donor (MRD). To fill this gap, matched unrelated donors (MUDs) are an increasingly selected donor source. Usage and outcomes after MUD HCT for Canada are not described. We investigated temporal trends in MUD compared to MRD HCT from 2000 to 2019 using data reported to the Cell Therapy and Transplant Canada (CTTC) Registry. Of 7571 first allogeneic HCTs between 2000 and 2019, the proportion of MUD HCTs rose from 35.1% to 56.3% in the early (2000–2009) and later (2010–2019) eras, respectively. Comparing the two donor sources, the 5-year overall survival (OS) after MUD HCT for patients with malignant diseases was inferior to MRD HCT in the early era (p < 0.001). However, in the later era, OS was comparable for the two donor sources (p = 0.969). For patients with non-malignant diseases, the 5-year OS after MUD HCT was inferior to MRD in the early era (p < 0.001), but in the later era, the 5-year OS was similar between the two donor sources (p = 0.209). Improvements in OS after MUD HCT were accompanied by corresponding reductions in the 2-year non-relapse mortality after MUD HCT. We conclude that MUDs are the most common donor source in Canada, and key clinical outcomes after MUD have improved over time. Full article
(This article belongs to the Section Cell Therapy)
15 pages, 269 KiB  
Review
Vaccination After Haematopoietic Stem Cell Transplant: A Review of the Literature and Proposed Vaccination Protocol
by André Silva-Pinto, Isabel Abreu, António Martins, Juliana Bastos, Joana Araújo and Ricardo Pinto
Vaccines 2024, 12(12), 1449; https://doi.org/10.3390/vaccines12121449 - 23 Dec 2024
Viewed by 576
Abstract
Background/Objectives: Haematopoietic stem cell transplantation (HCT) induces profound immunosuppression, significantly increasing susceptibility to severe infections. This review examines vaccinations’ necessity, timing, and efficacy post-HCT to reduce infection-related morbidity and mortality. It aims to provide a structured protocol aligned with international and national recommendations. [...] Read more.
Background/Objectives: Haematopoietic stem cell transplantation (HCT) induces profound immunosuppression, significantly increasing susceptibility to severe infections. This review examines vaccinations’ necessity, timing, and efficacy post-HCT to reduce infection-related morbidity and mortality. It aims to provide a structured protocol aligned with international and national recommendations. Methods: A systematic review of current guidelines and studies was conducted to assess vaccination strategies in HCT recipients. The analysis included the timing of vaccine administration, factors influencing efficacy, and contraindications. Recommendations for pre- and post-transplant vaccination schedules were synthesised, specifically for graft-versus-host disease (GVHD), immunosuppressive therapy, and hypogammaglobulinemia. Results: Vaccination is essential as specific immunity is often lost after HCT. Inactivated vaccines are recommended to commence three months post-transplant, including influenza, COVID-19, and pneumococcal vaccines. Live attenuated vaccines remain contraindicated for at least two years post-transplant and in patients with ongoing GVHD or immunosuppressive therapy. Factors such as GVHD and immunosuppressive treatments significantly impact vaccine timing and efficacy. The review also underscores the importance of pre-transplant vaccinations and ensuring that patients’ close contacts are adequately immunised to reduce transmission risks. Conclusions: Implementing a structured vaccination protocol post-HCT is critical to improving patient outcomes. Timely and effective vaccination strategies can mitigate infection risks while addressing individual patient factors such as GVHD and immunosuppression. This review highlights the need for tailored vaccination approaches to optimize immune reconstitution in HCT recipients. Full article
(This article belongs to the Section Vaccination Optimization)
12 pages, 993 KiB  
Article
Mixed T-Cell Chimerism Following Hematopoietic Cell Transplantation for Non-Malignant Disorders Is Common, Facilitates Anti-Viral Immunity, and Is Not Associated with Graft Failure in Pediatric Patients
by Rubiya Nadaf, Helena Lee, Denise Bonney, Ramya Hanasoge-Nataraj, Srividhya Senthil, Claire Horgan, Malcolm Guiver, Kay Poulton and Robert Wynn
Cells 2024, 13(24), 2119; https://doi.org/10.3390/cells13242119 - 20 Dec 2024
Viewed by 463
Abstract
Myeloid chimerism better reflects donor stem cell engraftment than whole-blood chimerism in assessing graft function following allogeneic hematopoietic stem cell transplant (HCT). We describe our experience with 130 patients aged younger than 18 years, treated with allogeneic HCT using bone marrow or PBSC [...] Read more.
Myeloid chimerism better reflects donor stem cell engraftment than whole-blood chimerism in assessing graft function following allogeneic hematopoietic stem cell transplant (HCT). We describe our experience with 130 patients aged younger than 18 years, treated with allogeneic HCT using bone marrow or PBSC from HLA-matched donors for non-malignant diseases, whose pre-transplant conditioning therapy included alemtuzumab and who were monitored with lineage-specific chimerism after transplant. At 6 years post-transplant, overall survival (OS) was 91.1% and event-free survival (EFS) was 81.5%, with no grade III-IV acute GvHD or chronic GVHD observed. Recipient T-cells did not contribute to graft loss. Mixed T-cell chimerism (MC) did not affect EFS, and there was no connection between T-cell chimerism and myeloid chimerism in patients with MC or graft loss. MC significantly correlated with virus infection; more children with MC were CMV seropositive than those with complete chimerism (CC). Additionally, MC was more common in patients with CMV viramia post-transplant. CD8 T-cell reconstitution was affected by viral reactivation, including CMV, with CD8 T-cell counts higher in the MC group than in the CC group. Mixed T-cell chimerism is due to autologous, virus-specific, predominantly CD8, T-cell expansion, and is protective and not deleterious to the recipient. Full article
(This article belongs to the Special Issue State of the Art and Future Prospects in Stem Cell Transplantation)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Kaplan–Meier curves showing overall survival (OS) and event-free survival (EFS). An event was defined as graft failure, death, or chronic GVHD requiring immune suppression. (<b>B</b>) Kaplan–Meier curves showing event-free survival in two groups: T-cell chimerism less than 50%, <span class="html-italic">n</span> = 35; and T-cell chimerism more than 50% at 3 months, post-HCT, <span class="html-italic">n</span> = 95. Study cohort <span class="html-italic">n</span> = 130, Log-rank (Mantel–Cox) test, <span class="html-italic">p</span> = 0.75.</p>
Full article ">Figure 2
<p>(<b>A</b>) Comparison of last recorded lineage-specific chimerism data of the graft failure group; <span class="html-italic">n</span> = 12. <span class="html-italic">p</span>-value determined via unpaired two-tail <span class="html-italic">t</span>-test, <span class="html-italic">p</span> = 0.0001. (<b>B</b>) CD3+/CD15+ trends in chimerism values in the graft loss group over time.</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>) Comparison of last recorded lineage-specific chimerism data of the graft failure group; <span class="html-italic">n</span> = 12. <span class="html-italic">p</span>-value determined via unpaired two-tail <span class="html-italic">t</span>-test, <span class="html-italic">p</span> = 0.0001. (<b>B</b>) CD3+/CD15+ trends in chimerism values in the graft loss group over time.</p>
Full article ">Figure 3
<p>Box plots of comparison of lineage-specific chimerism in stable mixed chimerism groups.</p>
Full article ">Figure 4
<p>(<b>A</b>) Impact of CMV serology in relation to chimerism status at 3 and 6 months post-HCT. Fisher’s exact test was used to compute the group contingencies; n = 107. At 3 months, in the MC group, 64.2% (18/28) were CMV seropositive (R+) and 35.7% (10/28) were CMV seronegative (R−). At 3 months, in the CC group, 41.7% (33/79) were R+ and 58.2% (46/79) were R−; <span class="html-italic">p</span> = 0.049 via Chi-square test. At 6 months, in the MC group, 61.7% (21/34) were CMV seropositive (R+) and 38.2% (13/34) were CMV seronegative (R−). At 6 months, in the CC group, 41.1% (30/73) were R+ and 58.9% (43/73) were R−; <span class="html-italic">p</span> = 0.04 via Chi-square test. There is a statistically significant relationship. (<b>B</b>) Impact of CMV viremia in relation to chimerism status at 3 and 6 months post-HCT. At 3 months, in the MC group, 50% (14/28) had CMV viremia and 50% (14/28) were negative. At 3 months, in the CC group, 27.8% (22/79) had CMV viremia and 72.1% (57/79) were negative. At 6 months, in the MC group, 50% (17/34) had CMV viremia and 50% (17/34) were negative. At 6 months, in the CC group, 26% (19/73) had CMV viremia and 73.9% (54/73) were negative; <span class="html-italic">p</span> = 0.03 and 0.01 by Chi-square test.</p>
Full article ">Figure 5
<p>(<b>A</b>) Distribution of CD8 T-cell count in CMV-positive and -negative patients at 3 months post-HCT. Mean CD8 T-cell was 548 in the CMV viremia group and 77.7 in patients with no CMV infection; <span class="html-italic">p</span> = 0.0001. (<b>B</b>) Distribution of CD8 T-cell count in the mixed chimerism and complete chimerism groups at 3 months post-HCT.</p>
Full article ">Figure 6
<p>Summative figure showing autologous T-cell expansion with viral infection.</p>
Full article ">
22 pages, 5263 KiB  
Article
Determination of Heavy Metals and Hemato-Biochemical Profiling of Bagre marinus and Bagarius bagarius in Jhelum River
by Muneeba Shaheen, Sana Ullah, Muhammad Bilal, Ahmed Muneeb, Begum Yurdakok-Dikmen and Caterina Faggio
Water 2024, 16(24), 3603; https://doi.org/10.3390/w16243603 - 14 Dec 2024
Viewed by 493
Abstract
Heavy metals enter river basins through industrial effluents, agricultural wastes, surface run-offs, and other human activities, negatively impacting aquatic and terrestrial life by bioaccumulating in the food chain. This problem is on a continuous rise in under-developed and developing countries, such as in [...] Read more.
Heavy metals enter river basins through industrial effluents, agricultural wastes, surface run-offs, and other human activities, negatively impacting aquatic and terrestrial life by bioaccumulating in the food chain. This problem is on a continuous rise in under-developed and developing countries, such as in Pakistan. Therefore, the current study was aimed to determine concentrations of heavy metals, essential trace elements, and macrominerals (Zn, Pb, Ni, Mn, Mg, Fe, Cu, Cr, Co, Cd, Ca, and As) in the water, sediments, and tissues (gills, liver, and muscles) of Bagarius bagarius and Bagre marinus in the Jhelum River, Pakistan. The hematological and biochemical profiles of these fish across two sampling sites (Jhelum Bridge Khushab, upstream, and Langarwala Pull—downstream) were also evaluated. Results showed greater bioaccumulation of heavy metals in fish downstream, correlating with higher concentrations of these metals in water and sediments downstream. In the case of B. marinus, the highest concentration observed was 16.59 mg/g (Ca), and the lowest concentration was 9.51 mg/g (Fe). In the case of B. bagarius, the highest concentration observed was 17.47 mg/g (Ca), and the lowest concentration was 7.95 mg/g (Mg). Increased activities of alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and lactate dehydrogenase (LDH) were observed downstream. Hematological changes included increased white blood cells (WBCs) and decreased red blood cells (RBCs), lymphocytes, hemoglobin (Hb), platelets (Plt), and hematocrit (Hct). A significant correlation was observed among heavy metals across the water, sediment, and different tissues of B. marinus and B. bagarius. Moreover, principal component analysis (PCA) for both species along both sampling sites illustrated the relationship between fish tissues and metals. The current study concluded that the fish accumulated a significantly higher concentration of heavy metals downstream, which might be linked with dumping of the domestic wastes and industrial and agricultural runoff, adversely affecting both fish and human health. Full article
Show Figures

Figure 1

Figure 1
<p>Summary of the preparation of samples for analysis.</p>
Full article ">Figure 2
<p>Heavy metal concentrations in (<b>A</b>) water and (<b>B</b>) sediment; data presented as Mean ± SE; significant difference among sites is represented by asterisks (two-sample <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05 = *, <span class="html-italic">p</span> &lt; 0.01 = **, and <span class="html-italic">p</span> &lt; 0.001 = ***). Heavy metal concentrations in tissues of (<b>C</b>) <span class="html-italic">B. marinus</span> (upstream), (<b>D</b>) <span class="html-italic">B. marinus</span> (downstream), (<b>E</b>). <span class="html-italic">B. bagarius</span> (upstream), and (<b>F</b>) <span class="html-italic">B. bagarius</span> (downstream); data are given as Mean ± SE and superscripted letters on bars show significance difference at <span class="html-italic">p</span> &lt; 0.05 (ANOVA followed by LSD).</p>
Full article ">Figure 3
<p>Level of heavy metals in the tissues of <span class="html-italic">Bagre marinus</span> and <span class="html-italic">Bagarius bagarius</span>. (<b>A</b>) Liver of <span class="html-italic">B. marinus</span>, (<b>B</b>) liver of <span class="html-italic">B. bagarius</span>, (<b>C</b>) gills of <span class="html-italic">B. marinus</span>, (<b>D</b>) gills of <span class="html-italic">B. bagarius</span>, (<b>E</b>) muscles of <span class="html-italic">B. marinus</span>, and (<b>F</b>) muscles of <span class="html-italic">B. bagarius</span>. Data presented as Mean ± SE. Significant difference among sites is represented by an asterisks (two-sample <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05 = *, <span class="html-italic">p</span> &lt; 0.01 = **, and <span class="html-italic">p</span> &lt; 0.001 = ***).</p>
Full article ">Figure 4
<p>Hematological profile of <span class="html-italic">Bagre marinus</span> and <span class="html-italic">Bagarius Bagarius</span> from upstream (US) and downstream (DS) of Jhelum River in Khushab District. Data represented as Mean ± SE. Significant difference among sites is represented by asterisks (two-sample <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05 = *, <span class="html-italic">p</span> &lt; 0.01 = **, and <span class="html-italic">p</span> &lt; 0.001 = ***).</p>
Full article ">Figure 5
<p>Serum biochemical profile of <span class="html-italic">Bagre marinus</span> and <span class="html-italic">Bagarius bagarius</span> from upstream (US) and downstream (DS) of Jhelum River in Khushab. (<b>A</b>) Bilirubin for <span class="html-italic">B. marinus</span>, (<b>B</b>) bilirubin for <span class="html-italic">B. bagarius</span>, (<b>C</b>) protein for <span class="html-italic">B. marinus</span>, (<b>D</b>) protein for <span class="html-italic">B. bagarius</span>, (<b>E</b>) enzymes for <span class="html-italic">B. marinus</span>, (<b>F</b>) enzymes for <span class="html-italic">B. bagarius</span>, (<b>G</b>) nitrogenous wastes for <span class="html-italic">B. marinus</span>, (<b>H</b>) nitrogenous wastes for <span class="html-italic">B. bagarius</span>, and (<b>I</b>) LDH for both species. Data presented as Mean ± SE. Significant difference among sites is represented by asterisks (two-sample <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05 = *, <span class="html-italic">p</span> &lt; 0.01 = ** and <span class="html-italic">p</span> &lt; 0.001 = ***).</p>
Full article ">Figure 6
<p>PCA diagrams for <span class="html-italic">Bagre marinus</span> and <span class="html-italic">Bagarius bagarius</span> along upstream and downstream sites of Jhelum River in Khushab District. (<b>A</b>) shows <span class="html-italic">Bagre marinus</span> at upstream, (<b>B</b>) shows <span class="html-italic">Bagre marinus</span> downstream, (<b>C</b>) shows <span class="html-italic">Bagarius bagarius</span> upstream and (<b>D</b>) shows <span class="html-italic">Bagarius bagarius</span> downstream.</p>
Full article ">
Back to TopTop