Nothing Special   »   [go: up one dir, main page]

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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (748)

Search Parameters:
Keywords = VEGFA

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 4423 KiB  
Article
A Versatile Skin-Derived Extracellular Matrix Hydrogel-Based Platform to Investigate the Function of a Mechanically Isolated Adipose Tissue Stromal Vascular Fraction
by Xue Zhang, Jan Aart M. Schipper, Rutger H. Schepers, Johan Jansma, Fred K. L. Spijkervet and Martin C. Harmsen
Biomolecules 2024, 14(12), 1493; https://doi.org/10.3390/biom14121493 - 23 Nov 2024
Viewed by 280
Abstract
Introduction: To ccelerate cutaneous wound healing and prevent scarring, regenerative approaches such as injecting a mechanically derived tissue stromal vascular fraction (tSVF) are currently under clinical and laboratory investigations. The aim of our study was to investigate a platform to assess the interaction [...] Read more.
Introduction: To ccelerate cutaneous wound healing and prevent scarring, regenerative approaches such as injecting a mechanically derived tissue stromal vascular fraction (tSVF) are currently under clinical and laboratory investigations. The aim of our study was to investigate a platform to assess the interaction between skin-derived extracellular matrix (ECM) hydrogels and tSVF and their effects on their microenvironment in the first ten days of culture. Material and Methods: A tSVF mixed with ECM hydrogel was cultured for ten days. After 0, 3, 5, and 10 days of culture viability, histology, immunohistochemistry, gene expression, and collagen alignment and organization were assessed. Results: The viability analysis showed that tSVF remained viable during 10 days of culture and seemed to remain within their constitutive ECM. The fiber analysis demonstrated that collagen alignment and organization were not altered. No outgrowth of capillaries was observed in (immuno)histochemical staining. The gene expression analysis revealed that paracrine factors TGFB1 and VEGFA did not change and yet were constitutively expressed. Pro-inflammatory factors IL1B and IL6 were downregulated. Matrix remodeling gene MMP1 was upregulated from day three on, while MMP14 was upregulated at day three and ten. Interestingly, MMP14 was downregulated at day five compared to day three while MMP2 was downregulated after day zero. Conclusions: Skin-derived ECM hydrogels appear to be a versatile platform for investigating the function of a mechanically isolated adipose tissue stromal vascular fraction. In vitro tSVF remained viable for 10 days and sustained the expression of pro-regenerative factors, but is in need of additional triggers to induce vascularization or show signs of remodeling of the surrounding ECM. In the future, ECM-encapsulated tSVF may show promise for clinical administration to improve wound healing. Full article
(This article belongs to the Special Issue Recent Developments in Mesenchymal Stem Cells)
14 pages, 1716 KiB  
Article
Angiogenesis as a Survival Mechanism in Heartworm Disease: The Role of Fructose-Bisphosphate Aldolase and Actin from Dirofilaria immitis in an In Vitro Endothelial Model
by Manuel Collado-Cuadrado, Claudia Alarcón-Torrecillas, Alfonso Balmori-de la Puente, Iván Rodríguez-Escolar, Elena Infante González-Mohino, Miguel Pericacho and Rodrigo Morchón
Animals 2024, 14(23), 3371; https://doi.org/10.3390/ani14233371 - 22 Nov 2024
Viewed by 317
Abstract
Heartworm disease, caused by Dirofilaria immitis, is a vector-borne zoonotic disease, (mainly affecting canids and felids) causing chronic vascular and pulmonary pathology in its early stages, which worsens with parasite load and/or death of adult worms in the pulmonary artery or right heart [...] Read more.
Heartworm disease, caused by Dirofilaria immitis, is a vector-borne zoonotic disease, (mainly affecting canids and felids) causing chronic vascular and pulmonary pathology in its early stages, which worsens with parasite load and/or death of adult worms in the pulmonary artery or right heart cavity, and can be fatal to the host. Angiogenesis is a mechanism by which new blood vessels are formed from existing ones. The aim of this work was to study the effect of two molecules of the D. immitis excretory/secretory antigen (DiES) on the angiogenic process, taking into account that this antigen is able to interact with this process and use it as a survival mechanism. For this purpose, an in vitro model of endothelial cells was used and treated with two recombinant proteins, i.e., actin (Act) and fructose-bisphosphate aldolase (FBAL) proteins belonging to DiES, and both pro- and antiangiogenic molecules were analyzed, as well as the cellular processes of cell proliferation, migration, and pseudocapillary formation. Act and FBAL proteins, together with vascular endothelial growth factor (VEGF-A), as an angiogenic precursor, are able to stimulate the production of proangiogenic factors as well as cellular processes of proliferation, migration, and pseudocapillary formation. This implies that these molecules could be produced by D. immitis to facilitate its survival, and the relationship between parasite and canine host would be further elaborated. Full article
(This article belongs to the Section Companion Animals)
23 pages, 1653 KiB  
Review
The Emerging Role of Long Noncoding RNAs in Sorafenib Resistance Within Hepatocellular Carcinoma
by Puneet Vij, Mohammad Shabir Hussain, Sanjaya K. Satapathy, Everardo Cobos and Manish K. Tripathi
Cancers 2024, 16(23), 3904; https://doi.org/10.3390/cancers16233904 - 21 Nov 2024
Viewed by 275
Abstract
Hepatocellular carcinoma (HCC), a liver cancer originating from hepatocytes, is a major health concern and among the most common malignancies worldwide. Sorafenib, approved by the U.S. F.D.A., is the primary first-line treatment for patients with advanced HCC. While the preferred first-line systemic regimen [...] Read more.
Hepatocellular carcinoma (HCC), a liver cancer originating from hepatocytes, is a major health concern and among the most common malignancies worldwide. Sorafenib, approved by the U.S. F.D.A., is the primary first-line treatment for patients with advanced HCC. While the preferred first-line systemic regimen for HCC is immunotherapy with Atezolizumab plus bevacizumab or Tremelimumab-actl + durvalumab, Sorafenib is still an alternative recommended regimen. While some patients with advanced HCC may benefit from Sorafenib treatment, most eventually develop resistance, leading to poor prognosis. Long noncoding RNAs (lncRNAs) have been found to play a critical role in tumorigenesis and the development of HCC, as well as other cancers. They are also key players in tumor drug resistance, though the mechanisms of lncRNAs in Sorafenib resistance in HCC remain poorly understood. This review summarizes the molecular mechanisms contributing to Sorafenib resistance in HCC with their potential correlation with lncRNAs, including the roles of transporters, receptors, cell death regulation, and other influencing factors. Full article
(This article belongs to the Special Issue Advances in Cancer Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Sorafenib, an anticancer drug, is transported into the cell through SLC proteins (OCT1 and FKSG16) and undergoes metabolism via CYP3A4 (phase 1) and phase 2 U.D.P. glucuronosyltransferase 1A9 (UGT1A9) to form the M1-M8 metabolites. Among the metabolites of sorafenib, M2, M4 (demethylation), and M5 (oxidative metabolite) were found to inhibit vascular endothelial growth factor receptors (VEGFRs). Sorafenib resistance: On the left side of the figure, sorafenib, an anticancer drug, is taken up into the cell primarily through the solute carrier (SLC proteins) and the organic cation transporter (OCT1). Once inside, sorafenib undergoes metabolism via CYP3A4 (phase 1) and phase 2 U.D.P. glucuronosyltransferase 1A9 (UGT1A9) to form the M1-M6 metabolites, including sorafenib glucuronide. These metabolites, particularly M2, can undergo glucuronidation to further reduce their activity. Sorafenib and its metabolites are also effluxed out of the cell by ABC transporters, including BCRP and MRP2, which contribute to drug re-sistance.</p>
Full article ">Figure 2
<p>Sorafenib induces the AMPK (AMP-activated protein kinase) pathway by increasing the ATP, initiating autophagy via AMPK, and through mTORC1 inhibition or ULK1 activation. Sorafenib can inhibit mTORC1 via the ERK pathway by inhibiting PTEN (Phosphatase and Tensin Homolog), but it can also activate mTORC1 through Akt activation (key regulators of P13K/AKT/mTOR pathway); it also inhibits ERK, further reducing mTOR activity. In addition, miRNAs are involved in sorafenib-mediated autophagy regulation. Sorafenib increases miR-21 and 25 expression, suppressing autophagy and decreasing FBXW7 protein expression, downregulating PTEN expression, and subsequently activating Akt.</p>
Full article ">
15 pages, 5367 KiB  
Article
Prolonged Extracorporeal Circulation Leads to Inflammation and Higher Expression of Mediators of Vascular Permeability Through Activation of STAT3 Signaling Pathway in Macrophages
by Jana Luecht, Camila Pauli, Raphael Seiler, Alexa-Leona Herre, Liliya Brankova, Felix Berger, Katharina R. L. Schmitt and Giang Tong
Int. J. Mol. Sci. 2024, 25(22), 12398; https://doi.org/10.3390/ijms252212398 - 19 Nov 2024
Viewed by 240
Abstract
Congenital heart defects (CHDs) are one of the most common congenital malformations and often require heart surgery with cardiopulmonary bypass (CPB). Children undergoing cardiac surgery with CPB are especially at greater risk of post-operative complications due to a systemic inflammatory response caused by [...] Read more.
Congenital heart defects (CHDs) are one of the most common congenital malformations and often require heart surgery with cardiopulmonary bypass (CPB). Children undergoing cardiac surgery with CPB are especially at greater risk of post-operative complications due to a systemic inflammatory response caused by innate inflammatory mediators. However, the pathophysiological response is not fully understood and warrants further investigation. Therefore, we investigated the inflammatory response in macrophages initiated by peri-operative serum samples obtained from patients with CHD undergoing CPB cardiac surgery. Human differentiated THP-1 macrophages were pretreated with Stattic, a STAT3 (Tyr705) inhibitor, before stimulation with serum samples. STAT3 and NF-κB activation were investigated via a Western blot, IL-1β, TNFα, IL-10, mediators for vascular permeability (VEGF-A, ICAM), and SOCS3 gene expressions via RT-qPCR. CPB induced an inflammatory response in macrophages via the activation of the STAT3 but not NF-κB signaling pathway. Longer duration on the CPB correlated with increased cytokine, VEGF, and ICAM expressions, relative to individual pre-operation levels. Patients that did not require CPB showed no significant immune response. Pretreatment with Stattic significantly attenuated all inflammatory mediators investigated except for TNFα in the macrophages. CPB induces an increased expression of cytokines and mediators of vascular permeability via the activation of STAT3 by IL-6 and IL-8 in the serum samples. Stattic attenuates all mediators investigated but promotes TNFα expression. Full article
(This article belongs to the Special Issue Molecular Pharmacology and Interventions in Cardiovascular Disease)
Show Figures

Figure 1

Figure 1
<p>Patients’ blood samples were obtained via the central venous line pre-operatively after the induction of anesthesia (T0), post-operatively upon arrival in the pediatric intensive care unit (PICU) (T1), and both 6 h (T2) and 24 h (T3) after the operation.</p>
Full article ">Figure 2
<p>(<b>a</b>) CPB induces the activation of STAT3 at phosphorylation site Tyr705 that was attenuated by pretreatment with 5 µM Stattic in THP-1 macrophages in vitro. LPS had no effect on STAT3 phosphorylation. (<b>b</b>) CPB had no effect on the NF-κB p65 signaling pathway in THP-1 macrophages in vitro. Pretreatment with Stattic attenuated LPS-induced NF-κB p65 phosphorylation. Data from five separate experiments (<span class="html-italic">n</span> = 5) are represented as box plots. Statistical test analysis: RM one-way ANOVA with Dunnett’s multiple comparisons post-test relative to individual pre-operative control (T0), a paired t-test relative to an untreated sample of the same time point, or an unpaired <span class="html-italic">t</span>-test relative to control; * <span class="html-italic">p</span> ≤ 0.05 was considered significant.</p>
Full article ">Figure 3
<p>A prolonged duration of CPB correlates with expressions of serum-induced cytokines and mediators of vascular permeability in THP-1 macrophages in vitro, including (<b>a</b>) IL-1β, (<b>b</b>) TNFα, (<b>c</b>) IL-10, (<b>d</b>) SOCS3, (<b>e</b>) ICAM, and (<b>f</b>) VEGF. Data from 56 patients are represented as line graphs. Statistical test analysis: RM one-way ANOVA with Dunnett’s multiple comparisons post-test relative to patients not undergoing CPB (0 h); * <span class="html-italic">p</span> ≤ 0.05 was considered significant.</p>
Full article ">Figure 4
<p>Stattic attenuates CPB-induced IL-1β and IL-10 expressions but augments TNFα expression in THP-1 macrophages in vitro. Serum samples from patients undergoing cardiac surgery without and with CPB for 1–3 h (<span class="html-italic">n</span> = 32) and 4–6 h (<span class="html-italic">n</span> = 13) induced significantly higher (<b>a</b>) IL-1β and (<b>c</b>) IL-10 expressions that could be attenuated by pretreatment with Stattic. No significant increases were observed in patients who did not require CPB (0 h, <span class="html-italic">n</span> = 5) nor in patients with an extremely long duration of CPB (7–9 h, <span class="html-italic">n</span> = 6). CPB for 1–3 h also elicited a significant increase in (<b>b</b>) TNFα expression that was augmented by pretreatment with Stattic. Data from 56 patients are represented as violin plots. Statistical test analysis: RM one-way ANOVA with Dunnett’s multiple comparisons post-test relative to individual pre-operative control (T0) or a paired <span class="html-italic">t</span>-test relative to an untreated sample of the same time point; * <span class="html-italic">p</span> ≤ 0.05 was considered significant.</p>
Full article ">Figure 4 Cont.
<p>Stattic attenuates CPB-induced IL-1β and IL-10 expressions but augments TNFα expression in THP-1 macrophages in vitro. Serum samples from patients undergoing cardiac surgery without and with CPB for 1–3 h (<span class="html-italic">n</span> = 32) and 4–6 h (<span class="html-italic">n</span> = 13) induced significantly higher (<b>a</b>) IL-1β and (<b>c</b>) IL-10 expressions that could be attenuated by pretreatment with Stattic. No significant increases were observed in patients who did not require CPB (0 h, <span class="html-italic">n</span> = 5) nor in patients with an extremely long duration of CPB (7–9 h, <span class="html-italic">n</span> = 6). CPB for 1–3 h also elicited a significant increase in (<b>b</b>) TNFα expression that was augmented by pretreatment with Stattic. Data from 56 patients are represented as violin plots. Statistical test analysis: RM one-way ANOVA with Dunnett’s multiple comparisons post-test relative to individual pre-operative control (T0) or a paired <span class="html-italic">t</span>-test relative to an untreated sample of the same time point; * <span class="html-italic">p</span> ≤ 0.05 was considered significant.</p>
Full article ">Figure 5
<p>Effects of Stattic on expressions of mediators of vascular permeability, (<b>a</b>) ICAM, (<b>b</b>) VEGF, and (<b>c</b>) the inhibitor of STAT3 activation (SOCS3) in THP-1 macrophages in vitro, stimulated with serum samples from patients undergoing cardiac surgery without and with CPB. The patient cohort was classified into four distinct groups based on the duration of CPB as follows: 0 h (<span class="html-italic">n</span> = 5), 1–3 h (<span class="html-italic">n</span> = 32), 4–6 h (<span class="html-italic">n</span> = 13), and 7–9 h (<span class="html-italic">n</span> = 6). Statistical test analysis: RM one-way ANOVA with Dunnett’s multiple comparisons post-test relative to individual pre-operative control (T0) or a paired <span class="html-italic">t</span>-test relative to an untreated sample of the same time point; * <span class="html-italic">p</span> ≤ 0.05 was considered significant.</p>
Full article ">Figure 5 Cont.
<p>Effects of Stattic on expressions of mediators of vascular permeability, (<b>a</b>) ICAM, (<b>b</b>) VEGF, and (<b>c</b>) the inhibitor of STAT3 activation (SOCS3) in THP-1 macrophages in vitro, stimulated with serum samples from patients undergoing cardiac surgery without and with CPB. The patient cohort was classified into four distinct groups based on the duration of CPB as follows: 0 h (<span class="html-italic">n</span> = 5), 1–3 h (<span class="html-italic">n</span> = 32), 4–6 h (<span class="html-italic">n</span> = 13), and 7–9 h (<span class="html-italic">n</span> = 6). Statistical test analysis: RM one-way ANOVA with Dunnett’s multiple comparisons post-test relative to individual pre-operative control (T0) or a paired <span class="html-italic">t</span>-test relative to an untreated sample of the same time point; * <span class="html-italic">p</span> ≤ 0.05 was considered significant.</p>
Full article ">
12 pages, 601 KiB  
Article
First-Line Combination of R-CHOP with the PDE4 Inhibitor Roflumilast for High-Risk DLBCL
by Adolfo E. Diaz Duque, Pedro S. S. M. Ferrari, Purushoth Ethiraj, Carine Jaafar, Zhijun Qiu, Kenneth Holder, Mathew J. Butler, Gabriela Huelgas-Morales, Anand Karnad, Patricia L. M. Dahia and Ricardo C. T. Aguiar
Cancers 2024, 16(22), 3857; https://doi.org/10.3390/cancers16223857 - 18 Nov 2024
Viewed by 527
Abstract
Background: Diffuse large B-cell lymphoma (DLBCL) is a common and often fatal malignancy. The standard-of-care immunochemotherapy, R-CHOP, cures only about 60% of DLBCL patients. Improving this cure rate will likely require the effective translation of basic biology knowledge into clinical activities. We previously [...] Read more.
Background: Diffuse large B-cell lymphoma (DLBCL) is a common and often fatal malignancy. The standard-of-care immunochemotherapy, R-CHOP, cures only about 60% of DLBCL patients. Improving this cure rate will likely require the effective translation of basic biology knowledge into clinical activities. We previously identified the cyclic-AMP/phosphodiesterase 4 (PDE4) axis as an important modulator of lymphomagenic processes. We also showed that the FDA-approved PDE4 inhibitor roflumilast can suppress B-cell receptor (BCR) signals, phosphoinositide 3-kinase (PI3K) activity and angiogenesis. These data suggested that combining roflumilast with R-CHOP may be beneficial in DLBCL. Methods: We conducted a single-center, single-arm, open-label, phase 1 study of roflumilast in combination with the standard of care, R-CHOP (Ro+R-CHOP), in pathologically proven, treatment-naïve, high-risk DLBCL patients. Results: Ro+R-CHOP was safe, and at a median follow-up time of 44 months, 70% of patients were alive and disease free (median OS not reached, PFS 44% (95% CI, 21–92). In this pilot series, we found that the addition of roflumilast suppressed PI3K activity in peripheral blood mononuclear cells, and VEGF-A secretion in the urine. We also encountered preliminary evidence to suggest that the Ro+R-CHOP combination may be particularly beneficial to patients diagnosed with high-risk genetic subtypes of DLBCL, namely MCD and A53. Conclusions: These initial findings suggest that roflumilast may be an alternative agent able to inhibit BCR/PI3K activity and angiogenesis in DLBCL, and that the testing of Ro+R-CHOP in a larger series of genetically characterized tumors is warranted. This study was registered at ClinicalTrials.gov, number NCT03458546. Full article
(This article belongs to the Special Issue PI3K Pathway in Cancer)
Show Figures

Figure 1

Figure 1
<p>VEGF-A quantification and PI3K activity in DLBCL patients treated with SOC R-CHOP or Ro+R-CHOP. (<b>A</b>) Left to right, fold change in urinary VEGF-A levels from baseline (before cycle 1—C1) to after the completion of two or five treatment cycles (at the start of cycle 3 or cycle 6, C3 or C6); nine Ro+R-CHOP- and six or seven R-CHOP-treated patients were included in these analyses. (<b>B</b>) Left to right, fold change in urinary VEGF-A levels from baseline (before cycle 1—C1) to after the completion of two or five treatment cycles (at the start of cycle 3 or cycle, C3 or C6); only non-GCB DLBCL patients and complete responders (CRs) were included in these analyses, with five Ro+R-CHOP- and three R-CHOP-treated patients. (<b>C</b>) Fold change in PI3K activity measured in the PBMCs of patients treated with Ro+R-CHOP, comparing the baseline (before cycle 1—C1) to after the completion of two treatment cycles (at the start of cycle 3—C3). Three patients with PR-R/R (partial responder–relapsed/refractory) who died of their disease are compared to four patients who achieved CR and remain free of disease (materials were not available for UPN#1, a complete responder and long-term survivor). (<b>D</b>) Fold change in PI3K activity measured in the PBMCs of patients treated with SOC R-CHOP, comparing the baseline (before cycle 1—C1) to after the completion of two treatment cycles (at the start of cycle 3—C3). Two patients in this series did not reach CR, and materials were not available for one of them. Data are mean ± SD. All data points quantified are shown (three replicates for each patient); <span class="html-italic">p</span> values are from two-sided Student’s <span class="html-italic">t</span>-test, * &lt; 0.05, *** &lt; 0.001, **** &lt; 0.0001.</p>
Full article ">
21 pages, 5560 KiB  
Article
Hypoxia Preconditioned Serum Hydrogel (HPS-H) Accelerates Dermal Regeneration in a Porcine Wound Model
by Jun Jiang, Tanita Man, Manuela Kirsch, Samuel Knoedler, Kirstin Andersen, Judith Reiser, Julia Werner, Benjamin Trautz, Xiaobin Cong, Selma Forster, Sarah Alageel, Ulf Dornseifer, Arndt F. Schilling, Hans-Günther Machens, Haydar Kükrek and Philipp Moog
Gels 2024, 10(11), 748; https://doi.org/10.3390/gels10110748 - 17 Nov 2024
Viewed by 428
Abstract
Harnessing the body’s intrinsic resources for wound healing is becoming a rapidly advancing field in regenerative medicine research. This study investigates the effects of the topical application of a novel porcine Hypoxia Preconditioned Serum Hydrogel (HPS-H) on wound healing using a minipig model [...] Read more.
Harnessing the body’s intrinsic resources for wound healing is becoming a rapidly advancing field in regenerative medicine research. This study investigates the effects of the topical application of a novel porcine Hypoxia Preconditioned Serum Hydrogel (HPS-H) on wound healing using a minipig model over a 21-day period. Porcine HPS exhibited up to 2.8× elevated levels of key angiogenic growth factors (VEGF-A, PDGF-BB, and bFGF) and demonstrated a superior angiogenic effect in a tube formation assay with human umbilical endothelial cells (HUVECs) in comparison to porcine normal serum (NS). Incorporating HPS into a hydrogel carrier matrix (HPS-H) facilitated the sustained release of growth factors for up to 5 days. In the in vivo experiment, wounds treated with HPS-H were compared to those treated with normal serum hydrogel (NS-H), hydrogel only (H), and no treatment (NT). At day 10 post-wounding, the HPS-H group was observed to promote up to 1.7× faster wound closure as a result of accelerated epithelialization and wound contraction. Hyperspectral imaging revealed up to 12.9% higher superficial tissue oxygenation and deep perfusion in HPS-H-treated wounds at day 10. The immunohistochemical staining of wound biopsies detected increased formation of blood vessels (CD31), lymphatic vessels (LYVE-1), and myofibroblasts (alpha-SMA) in the HPS-H group. These findings suggest that the topical application of HPS-H can significantly accelerate dermal wound healing in an autologous porcine model. Full article
(This article belongs to the Special Issue Gel-Based Novel Wound Dressing)
Show Figures

Figure 1

Figure 1
<p>In vivo experimental setup. (<b>A</b>) Timeline: At day −18, the animals arrived and were housed in groups for acclimatization. At day −4, Hypoxia Preconditioned Serum Hydrogel (HPS-H) and normal serum hydrogel (NS-H) were produced for each pig and were stored at −20 °C for later use. At day 0, wounding of the pigs was proceeded according to (<b>B</b>), and the topical autologous applications (HPS-H and NS-H) and hydrogel only (H) were performed. In the no treatment (NT) group, only saline was applied. Wound analysis included digital photographs and hyperspectral imaging. On days 5, 10, and 14, the dressings were changed, wound analysis was conducted, and, additionally, the respective rows of the indicated days were biopsied for immunohistochemical analysis (see (<b>B</b>)). Topical applications were performed for the rest of the wounds. On day 21, final analysis and biopsies were conducted. A total of three independent experiments were conducted, each with two animal pairs.</p>
Full article ">Figure 2
<p>Analysis of growth factor concentration in porcine Hypoxia Preconditioned Serum (HPS) and its release from HPS Hydrogel (HPS-H). (<b>A</b>) Quantitative measurements of pro- (VEGF-A, PDGF-BB, and bFGF) and (<b>B</b>) anti-angiogenic (PF-4 and TSP-1) growth factors in HPS in comparison to NS. Paired <span class="html-italic">t</span>-test. Data presented as mean ± SEM, porcine blood donors: <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. (<b>C</b>) Quantitative analysis of growth factor release in HPS-H over a 5-day period. Data were provided as cumulative concentrations. Data points are means ± SEM. Porcine blood donors: <span class="html-italic">n</span> = 4.</p>
Full article ">Figure 3
<p>Effect of porcine HPS on the tube formation of HUVECs. (<b>A</b>) Representative microscopic photographs of the tube formation assay (6 h) of HUVECs, carried out in the presence of the blood-derived secretomes compared to positive/negative control. Scale bar = 100 μm. (<b>B</b>–<b>E</b>) Image analysis of the digital photographs depicted in (<b>A</b>). One-way ANOVA with Tukey’s multiple comparison test. Data points are means ± SEM. Porcine blood donors: <span class="html-italic">n</span> = 3. Capital letters over plots indicate statistical significance of data pairs with different letters. For all pair comparisons, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Wound closure of excisional wounds treated with autologous porcine HPS-H in comparison to NS-H, H, and NT. (<b>A</b>) Representative standardized photographs of full-thickness wounds in the excisional porcine skin model on postoperative days 0, 5, 10, 14, and 21. Scale bar = 0.5 cm. (<b>B</b>) Plot showing total wound closure in % of initial wound surface area (1.5 cm × 1.5 cm). (<b>C</b>) Plot showing wound epithelization in % of the contracted wound area. (<b>D</b>) Plot showing wound contraction in % of initial wound surface area. (<b>B</b>–<b>D</b>) Data points represent means ± SEM from three independent animal experiments. Two-way ANOVA with Tukey’s multiple comparisons test. For pair comparisons HPS-H vs. NS-H: * = <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; 0.0001; HPS-H vs. H: # = <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, #### = <span class="html-italic">p</span> &lt; 0.0001; HPS-H vs. NT: + = <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, ++++ = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Hyperspectral images of excisional wounds treated with autologous porcine HPS-H in comparison to NS-H, H, and NT. Representative hyperspectral image of (<b>A</b>) stO2 (%), (<b>C</b>) NIR index, and (<b>E</b>) TWI measurements on days 0, 5, 10, 14, and 21. Scale bar = 0.5 cm. The wound edges were analyzed from each day and were plotted in (<b>B</b>) for stO2 (%), (<b>D</b>) for NIR index, and (<b>F</b>) for TWI. (<b>B</b>,<b>D</b>,<b>F</b>) Data points represent means ± SEM from three independent animal experiments. Two-way ANOVA with Tukey’s multiple comparisons test. For pair comparisons HPS-H vs. NT: * = <span class="html-italic">p</span> &lt; 0.05; HPS-H vs. H: # = <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Immunohistochemical diaminobenzidine (DAB)-staining of wounds treated with autologous porcine HPS-H in comparison to NS-H, H, and NT. Representative immunohistochemical staining of (<b>A</b>) CD31, (<b>C</b>) LYVE-1, and (<b>E</b>) alpha-SMA on days 5, 10, 14, and 21. (<b>A</b>,<b>C</b>) Scale bar = 100 μm and (<b>E</b>) scale bar = 33 μm. The area of the DAB-positive staining (μm<sup>2</sup>) was analyzed from each day and were plotted in (<b>B</b>) for CD31, (<b>D</b>) for LYVE-1, and (<b>F</b>) for alpha-SMA. (<b>B</b>,<b>D</b>,<b>F</b>) Data points represent means ± SEM from three independent animal experiments. Two-way ANOVA with Tukey’s multiple comparisons test. * <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, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
13 pages, 1531 KiB  
Article
Effect of Subconjunctival Injection of Canine Adipose-Derived Mesenchymal Stem Cells on Canine Spontaneous Corneal Epithelial Defects
by Pechchalee Kengkla, Yaowalak Panyasing, Aree Thayananuphat and Nalinee Tuntivanich
Animals 2024, 14(22), 3270; https://doi.org/10.3390/ani14223270 - 13 Nov 2024
Viewed by 453
Abstract
Spontaneous chronic corneal epithelial defects (SCCEDs) are characterized by nonadherent corneal epithelium leading to poor attachment to the corneal stroma. The objective of this study was to characterize corneal outcomes concurrently with the quantification of tumor necrosis factor-alpha (TNF-α) and vascular endothelial growth [...] Read more.
Spontaneous chronic corneal epithelial defects (SCCEDs) are characterized by nonadherent corneal epithelium leading to poor attachment to the corneal stroma. The objective of this study was to characterize corneal outcomes concurrently with the quantification of tumor necrosis factor-alpha (TNF-α) and vascular endothelial growth factor-A (VEGF-A) in tear fluid after the subconjunctival injection of canine adipose-derived mesenchymal stem cells (cAD-MSCs) in canine SCCEDs. Ten eyes with SCCEDs, which were nonresponsive to two rounds of diamond burr debridement, were included in this study. All eyes received a single subconjunctival injection of 1 × 106 cAD-MSCs. Ophthalmic examinations were performed before treatment and on day 7, 14, and 21 after treatment. Tear samples were collected for the quantification of TNF-α and VEGF-A concentrations by a canine multiplex immunoassay. Nine out of ten eyes revealed complete healing by day 21. The mean healing time was 10.89 ± 1.7 days. All eyes showed a decreased degree of ocular discomfort, in accordance with the degree of corneal characteristics. The concentrations of VEGF-A significantly reduced from pre-treatment (4334.91 ± 1275.92 pg/mL) to day 21 post-treatment (3064.61 ± 1028.66 pg/mL). No significant difference in TNF-α concentration was observed before/after treatment. In conclusion, the single use of a subconjunctival injection of cAD-MSCs could be used as an alternative treatment for canine SCCEDs. Full article
(This article belongs to the Section Companion Animals)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Corneal characteristics of 10 canine eyes with SCCEDs before and after treatment with the subconjunctival injection of cAD-MSCs.</p>
Full article ">Figure 2
<p>Bar graph illustrating percentage of corneal epithelial defect (<b>a</b>), corneal neovascularization (<b>b</b>), and corneal opacification area (<b>c</b>) before and after subconjunctival injection of cAD-MSCs; * <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.</p>
Full article ">Figure 3
<p>Bar graph illustrating concentrations of TNF-α (<b>a</b>) and VEGF-A (<b>b</b>) (pg/mL) in tear fluid before and after subconjunctival injection of cAD-MSCs; * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
12 pages, 320 KiB  
Article
Is Serum VEGF-A Level an Indicator of Early-Onset Poststroke Depression?
by Emine Yildirim Uslu and Sevler Yildiz
Medicina 2024, 60(11), 1828; https://doi.org/10.3390/medicina60111828 - 7 Nov 2024
Viewed by 421
Abstract
Background and Objectives: Poststroke depression (PSD) is a psychiatric complication occurring after a stroke, and is known to negatively impact quality of life. In the present study, the possible relationship between serum vascular endothelial growth factor (VEGF-A) levels and early-onset PSD, as [...] Read more.
Background and Objectives: Poststroke depression (PSD) is a psychiatric complication occurring after a stroke, and is known to negatively impact quality of life. In the present study, the possible relationship between serum vascular endothelial growth factor (VEGF-A) levels and early-onset PSD, as well as the predictive value of serum VEGF-A levels for early-onset PSD, were investigated. Materials and Methods: The study included 88 individuals diagnosed with acute ischemic stroke (AIS). Demographic data, clinical characteristics, and serum VEGF-A levels were recorded, and radiological images were examined to determine the lesion locations. The National Institutes of Health Stroke Scale (NIHSS), Montreal Cognitive Assessment (MoCA), and Hamilton depression scale (HAMD-17) were administered to the patients. Furthermore, serum VEGF-A levels were measured in all participants. Results: Although the body mass index (BMI) and VEGF-A levels were similar between the groups, MoCA scores were lower [(19.2 ± 4.4) vs. (22.3 ± 3), p = 0.001] and NIHSS scores were higher [18 (8–28) vs. 14 (3–24), p = 0.006] in individuals with PSD than in those without it. When the patients with PSD were categorized into three groups, patients with severe PSD had higher NIHSS scores [26 (23–27) vs. 15 (8–23), p = 0.006] and lower MoCA scores [(14.3 ± 1) vs. (20.9 ± 3.8), p = 0.005] than those with mild PSD. Moreover, VEGF-A levels and lesion localization were similar between mild, moderate, and severe PSD groups (p = 0.130). The MoCA score was negatively (r = −0.498, p < 0.001) correlated and the NIHSS score was positively correlated (r = 0.497, p < 0.001) with the HAMD-17 score. Conclusions: Our findings suggest that longitudinal studies in large cohorts including healthy control groups are needed to examine the possibility of using serum VEGF-A level as a marker for predicting early-onset PSD. Full article
(This article belongs to the Special Issue Recent Advances in Stroke Screening)
19 pages, 2569 KiB  
Article
A Prospective Cohort Study on the Effects of Repeated Acute Stress on Cortisol Awakening Response and Immune Function in Military Medical Students
by Madison A. Propp, Dean Paz, Sukhrob Makhkamov, Mark E. Payton, Qamrul Choudhury, Melodie Nutter and Rebecca Ryznar
Biomedicines 2024, 12(11), 2519; https://doi.org/10.3390/biomedicines12112519 - 4 Nov 2024
Viewed by 541
Abstract
Background: The cortisol awakening response (CAR) is a pivotal component of the body’s stress response, yet its dynamics under repeated acute stress and its interplay with immune biomarkers remain inadequately understood. Methods: This study examined 80 second-year military medical students undergoing a 5-day [...] Read more.
Background: The cortisol awakening response (CAR) is a pivotal component of the body’s stress response, yet its dynamics under repeated acute stress and its interplay with immune biomarkers remain inadequately understood. Methods: This study examined 80 second-year military medical students undergoing a 5-day intensive surgical simulation designed to elicit stress responses. Salivary samples were collected daily upon waking and 30 min thereafter to measure cortisol and a panel of cytokines using bead-based multiplex ELISA. Results: Analysis revealed a significant blunting of the CAR on the third day of training (p = 0.00006), followed by a recovery on the fourth day (p = 0.0005). Concurrently, specific cytokines such as CXCL1 (r = 0.2, p = 0.0005), IL-6 (r = 0.13, p = 0.02), IL-10 (r = 0.14, p = 0.02), and VEGF-A (r = 0.17, p = 0.003) displayed patterns correlating with the CAR, with increased strength of associations observed when assessing cytokine levels against the CAR of the preceding day (CXCL1 r = 0.41, p = 0.0002. IL-6 r = 0.38, p = 0.0006. IL-10 r = 0.3, p = 0.008. VEGF-A r = 0.41, p = 0.0002). Conclusions: These results suggest a temporal relationship between stress-induced cortisol dynamics and immune regulation. The CAR pattern demonstrated in this study may represent induction of and recovery from psychological burnout. Moreover, the observed cytokine associations provide insight into the mechanisms by which stress can influence immune function. The results may have broader implications for managing stress in high-performance environments, such as military and medical professions, and for identifying individuals at risk of stress-related immune suppression. Full article
(This article belongs to the Special Issue The Role of Cytokines in Health and Disease: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Intensive Surgical Skills Week schedule. Samples were collected at wake and 30 min post waking. Students go through hyper-realistic mass casualty and operating room simulations, going through each simulation two times over the duration of training.</p>
Full article ">Figure 2
<p>Daily mean percent change in AM salivary cortisol from waking to 30 min later (CAR) across all participants (n = 79) during repeated acute stress. Error bars represent the standard error of the mean (SEM). Results of pairwise comparisons between daily means are shown by the compact letter display. Two means with the same letter are not significantly different at the 0.05 level. Day 3 CAR is significantly reduced compared to all other days (D1, D3 <span class="html-italic">p</span> = 0.014), (D2, D3 <span class="html-italic">p</span> = 0.00006), (D4, D3 <span class="html-italic">p</span> = 0.0005). Day 1 average CAR = 67.28%, Day 2 = 84.45%, Day 3 = 21.30%, Day 4 = 78.83%. Typically, a healthy CAR is expected to be between 50 and 80%.</p>
Full article ">Figure 3
<p>Panels A-H represent daily average salivary interleukin levels in pg/mL at Time 1 (waking) and Time 2 (30 min post waking) during repeated acute stress. Error bars represent the SEM. Results of pairwise comparisons between average cytokine values at each time point are shown by the compact letter display. Two means with the same letter are not significantly different at the 0.05 level. All interleukins exhibited significant decreases in average salivary concentration from Time 1 to Time 2 in all days of the sampling period. IL-1a, IL-8 Il-15, IL-18, and IL-1RA, <span class="html-italic">p</span> &lt; 0.0001. IL-6 Day 1, <span class="html-italic">p</span> = 0.0014; Day 2, <span class="html-italic">p</span> = 0.0124; Day 3, <span class="html-italic">p</span> = 0.0003; Day 4, <span class="html-italic">p</span> &lt; 0.0001. IL-10 Day 1, <span class="html-italic">p</span> = 0.003; Day 2, <span class="html-italic">p</span> = 0.0056; Day 3, <span class="html-italic">p</span> = 0.0086; Day 4, <span class="html-italic">p</span> = 0.0098. IL-1B Day 1, <span class="html-italic">p</span> = 0.0001; Days 2–4, <span class="html-italic">p</span> &lt; 0.0001. (<b>a</b>) IL-1a (n = 79), (<b>b</b>) IL-6 (n = 79), (<b>c</b>) IL-8 (n = 79), (<b>d</b>) IL-10 (n = 79), (<b>e</b>) IL-15 (n = 79), (<b>f</b>) IL-18 (n = 79), (<b>g</b>) IL-1β (n = 40), (<b>h</b>) IL-1RA (n = 79).</p>
Full article ">Figure 4
<p>Panels A-E represent daily average salivary growth factor levels in pg/mL at Time 1 (waking) and Time 2 (30 min post waking) during repeated acute stress. Error bars represent SEM. Results of pairwise comparisons between average cytokine values at each time point are shown by the compact letter display. Two means with the same letter are not significantly different at the 0.05 level. VEGF-A showed significant increases in the post wake period on all 4 days (<span class="html-italic">p</span> = 0.0059, <span class="html-italic">p</span> = 0.0105, <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">p</span> = 0.0155). PDGF-AA showed significant increases in the post wake period on Days 3 and 4 (<span class="html-italic">p</span> = 0.0001, <span class="html-italic">p</span> = 0.0005). EGF, showed significant decreases in the post wake period on all days (<span class="html-italic">p</span> &lt; 0.0001). FGF-2 showed significant decreases on Days 1–3 (<span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">p</span> = 0.0003, <span class="html-italic">p</span> = 0.008). TGFα showed a significant decrease on Day 3 (<span class="html-italic">p</span> = 0.011). (<b>a</b>) EGF (n = 79), (<b>b</b>) FGF-2 (n = 79), (<b>c</b>) VEGF-A (n = 79), (<b>d</b>) TGFα (n = 79), (<b>e</b>) PDGF-AA (n = 79).</p>
Full article ">Figure 5
<p>Panels A-F represent daily average salivary chemokine and miscellaneous cytokine levels in pg/mL at Time 1 (waking) and Time 2 (30 min post waking) during repeated acute stress. Error bars represent SEM. Results of pairwise comparisons between average cytokine values at each time point are shown by the compact letter display. Two means with the same letter are not significantly different at the 0.05 level. MCP-1 showed significant decreases in the post wake period on all 4 days (<span class="html-italic">p</span> &lt; 0.0001). CX3CL1 showed decreases in salivary concentration in the post wake period on all days but only the decrease on Day 2 reached statistical significance (<span class="html-italic">p</span> = 0.0198). TNFα showed significant decreases in salivary concentration from Time 1 to Time 2 on all days (<span class="html-italic">p</span> = 0.0021, <span class="html-italic">p</span> = 0.0055, <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">p</span> = 0.0028). G-CSF levels do not show significant changes from Time 1 to Time 2 on any day; however, overall levels are significantly decreased between Day 3 and Day 4 (T1 <span class="html-italic">p</span> = 0.0028, T2 <span class="html-italic">p</span> = 0.0001). (<b>a</b>) CXCL1 (n = 79), (<b>b</b>) CX1CL3 (n = 40), (<b>c</b>) MCP-1 (n = 79), (<b>d</b>) IFNγ (n = 40), (<b>e</b>) TNFα (n = 40), (<b>f</b>) G-CSF (n = 40).</p>
Full article ">
12 pages, 10244 KiB  
Article
Angiopoietin 1 Attenuates Dysregulated Angiogenesis in the Gastrocnemius of DMD Mice
by Andrew McClennan and Lisa Hoffman
Int. J. Mol. Sci. 2024, 25(21), 11824; https://doi.org/10.3390/ijms252111824 - 4 Nov 2024
Viewed by 712
Abstract
Duchenne muscular dystrophy (DMD) is a degenerative neuromuscular disease caused by a lack of functional dystrophin. Ang 1 paracrine signalling maintains the endothelial barrier of blood vessels, preventing plasma leakage. Chronic inflammation, a consequence of DMD, causes endothelial barrier dysfunction in skeletal muscle. [...] Read more.
Duchenne muscular dystrophy (DMD) is a degenerative neuromuscular disease caused by a lack of functional dystrophin. Ang 1 paracrine signalling maintains the endothelial barrier of blood vessels, preventing plasma leakage. Chronic inflammation, a consequence of DMD, causes endothelial barrier dysfunction in skeletal muscle. We aim to elucidate changes in the DMD mouse’s gastrocnemius microvascular niche following local administration of Ang 1. Gastrocnemii were collected from eight-week-old mdx/utrn+/− and healthy mice. Additional DMD cohort received an intramuscular injection of Ang 1 to gastrocnemius and contralateral control. Gastrocnemii were collected for analysis after two weeks. Using immunohistochemistry and real-time quantitative reverse transcription, we demonstrated an abundance of endothelial cells in DMD mouse’s gastrocnemius, but morphology and gene expression were altered. Myofiber perimeters were shorter in DMD mice. Following Ang 1 treatment, fewer endothelial cells were present, and microvessels were more circular. Vegfr1, Vegfr2, and Vegfa expression in Ang 1-treated gastronemii increased, while myofiber size distribution was consistent with vehicle-only gastrocnemii. These results suggest robust angiogenesis in DMD mice, but essential genes were underexpressed—furthermore, exogenous Ang 1 attenuated angiogenesis. Consequentially, gene expression increased. The impact must be investigated further, as Ang 1 therapy may be pivotal in restoring the skeletal muscle microvascular niche. Full article
Show Figures

Figure 1

Figure 1
<p>Histology of anti-CD31-stained endothelial cells and anti-laminin-stained myofibers in Healthy (<b>a</b>–<b>f</b>), Diseased (<b>g</b>–<b>l</b>), Vehicle-only (<b>m</b>–<b>r</b>), and Ang 1-Treated (<b>s</b>–<b>x</b>) transverse gastrocnemius samples. Scale bar = 50 µm (<b>a</b>,<b>c</b>,<b>e</b>,<b>g</b>,<b>i</b>,<b>k</b>,<b>m</b>,<b>o</b>,<b>q</b>,<b>s</b>,<b>u</b>,<b>w</b>), 10 µm (<b>b</b>,<b>d</b>,<b>f</b>,<b>h</b>,<b>j</b>,<b>l</b>,<b>n</b>,<b>p</b>,<b>r</b>,<b>t</b>,<b>v</b>,<b>x</b>).</p>
Full article ">Figure 2
<p>Microvessel morphometry in Healthy, Diseased, Vehicle-only, and Ang 1-Treated gastrocnemius samples: (<b>a</b>,<b>b</b>) percent area CD31 positively stained tissue, (<b>c</b>,<b>d</b>) capillary-to-fibre perimeter exchange (CFPE) index, (<b>e</b>,<b>f</b>) microvessel area, (<b>g</b>,<b>h</b>) microvessel density, and (<b>i</b>,<b>j</b>) microvessel circularity. Data are mean ± SEM. <span class="html-italic">n</span> = 6–7. * <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>Analysis of endothelial cell-related gene expression by Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR). Gene expression was measured in the gastrocnemius of (<b>a</b>) Diseased relative to Healthy and (<b>b</b>) Ang 1-Treated relative to Vehicle-only. Data are mean ± SEM. <span class="html-italic">n</span> = 6. * <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, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p><span class="html-italic">Ang 1</span>:<span class="html-italic">Ang 2</span> relative fold change ratio of (<b>a</b>) Healthy and Diseased and (<b>b</b>) Vehicle-only and Ang 1-Treated gastrocnemii. Data are mean ± SEM. <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Distribution of myofiber area of (<b>a</b>) Healthy, Diseased <span class="html-italic">p</span> = 0.0642, (<b>b</b>) Vehicle-only, and Ang 1-Treated <span class="html-italic">p</span> = 0.0792 gastrocnemius samples. <span class="html-italic">n</span> = 6.</p>
Full article ">Figure 6
<p>Representative histology of Masson’s Trichrome staining. Blue arrows indicate collagen deposition in (<b>a</b>) Healthy, (<b>b</b>) Diseased, (<b>c</b>) Vehicle-only, and (<b>d</b>) Ang 1-Treated gastrocnemius with scale bar = 150 μm. (<b>e</b>) Healthy, (<b>f</b>) Diseased, (<b>g</b>) Vehicle-only, and (<b>h</b>) Ang 1-Treated gastrocnemius with scale bar = 35 μm.</p>
Full article ">Figure 7
<p>Quantitative analysis of collagen deposition in (<b>a</b>) Healthy and Diseased, and (<b>b</b>) Vehicle-only, and Ang 1-Treated gastrocnemius. <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
17 pages, 8149 KiB  
Article
Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling
by Yuqing Long, Yuxi Dan, Yao Jiang, Jing Ma, Tao Zhou, Liaoqiong Fang and Zhibiao Wang
Biology 2024, 13(11), 873; https://doi.org/10.3390/biology13110873 - 27 Oct 2024
Viewed by 708
Abstract
Background: Angiogenesis plays a crucial role in the growth of colorectal cancer (CRC). Recent studies have identified extracellular vesicles (EVs) in the tumor microenvironment as important mediators of cell-to-cell communication. However, the specific role and mechanisms of CRC-derived EVs in regulating tumor angiogenesis [...] Read more.
Background: Angiogenesis plays a crucial role in the growth of colorectal cancer (CRC). Recent studies have identified extracellular vesicles (EVs) in the tumor microenvironment as important mediators of cell-to-cell communication. However, the specific role and mechanisms of CRC-derived EVs in regulating tumor angiogenesis remain to be further investigated. Methods: EVs were isolated from the conditioned medium of the CRC cells using ultracentrifugation. We investigated the effects of HT-29-derived EVs on tumor growth and angiogenesis in a subcutaneous HT-29 CRC tumor model in mice. Additionally, we evaluated the impact of HT-29-derived EVs on the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs). Subsequently, bioinformatics analysis was performed to identify relevant signaling pathways, and pathway inhibitors were used to block the activation of these pathways, aiming to elucidate their roles in angiogenesis. Results: We found that HT-29-derived EVs can promote tumor growth and angiogenesis in vivo, as well as significantly enhance the proliferation, migration, and tube formation of HUVECs. Bioinformatics analysis revealed that HT-29-derived EVs may regulate angiogenesis through the JAK/STAT3 signaling pathway. Specifically, we observed that CRC-derived EVs promoted the phosphorylation of STAT3 (p-STAT3) and the expression of VEGFA in the nucleus of HUVECs. Treatment with the STAT3 inhibitor Stattic reduced the nuclear expression of p-STAT3, which impaired its function as a transcription factor, thereby inhibiting VEGFA expression and the pro-angiogenic effects of CRC-derived EVs. Conclusions: EVs derived from CRC cells promote CRC tumor angiogenesis by regulating VEGFA through the JAK/STAT3 pathway in endothelial cells. Full article
(This article belongs to the Special Issue Extracellular Vesicles and Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>Isolation and characterization of EVs derived from HT-29 colorectal cancer cells (HT29-EVs). (<b>A</b>) Flowchart of the extracellular vesicles isolation procedure. (<b>B</b>) Nanoparticle tracking analysis of the size distribution of the extracellular vesicles (<a href="#app1-biology-13-00873" class="html-app">Figure S9</a>). (<b>C</b>) Transmission electron microscopy image of the extracellular vesicles (<a href="#app1-biology-13-00873" class="html-app">Figure S10</a>). (<b>D</b>) Western blotting analysis of the positive (CD63 and TSG101) and negative (calnexin) markers of extracellular vesicles (<a href="#app1-biology-13-00873" class="html-app">Figure S11</a>).</p>
Full article ">Figure 2
<p>HT-29-derived EVs promote HT-29 colorectal cancer tumor growth and angiogenesis in vivo. (<b>A</b>) Model diagram of mouse tumorigenesis model. (<b>B</b>) Tumor image of each group. (<b>C</b>) The tumor growth curve shows the tumor size measured every 2 days. (<b>D</b>) The weight of tumors in each group. (<b>E</b>) CD31 staining of tumors and quantification of vessel area (200×: scale bar = 100 μm, 400×: scale bar = 50 μm). (<b>F</b>) VEGFA staining of tumors and quantification of the average optical density (200×: scale bar = 100 μm, 400×: scale bar = 50 μm). Data were analyzed by <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, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>HT29-EVs promote angiogenesis in vitro. (<b>A</b>) HUVECs were treated with control PBS or HT29-EVs that had been labeled with PKH67 (green) for 6 h and stained with DAPI (blue) (scale bar = 13 μm). HT29-EVs uptake by HUVECs was observed. (<b>B</b>) The EdU assay indicated the effect of different concentrations of HT29-EVs on the proliferation rate of HUVECs (scale bar = 65 μm). (<b>C</b>) Immunofluorescence analysis of PCNA protein expression in HUVECs treated with different concentrations of HT29-EVs and fluorescence intensity statistics (scale bar = 25 μm). (<b>D</b>) The transwell assay was performed to detect the impact of different concentrations of HT29-EVs on the migration of HUVECs. (<b>E</b>) The scratch wound healing assay was performed to detect the impact of different concentrations of HT29-EVs on the migration of HUVECs (scale bar = 520 μm). (<b>F</b>) The FITC-phalloidin assay was performed to detect the effect of different concentrations of HT29-EVs on F-actin protein expression in HUVECs (scale bar = 25 μm). (<b>G</b>) The tube formation assay was used to detect the angiogenesis ability of HUVECs (scale bar = 200 μm). (<b>H</b>) Quantitative analysis of nodes, total tube length, and total loops (<a href="#app1-biology-13-00873" class="html-app">Figure S12</a>). (<b>I</b>) Western blotting analysis of VEGFA in HUVECs incubated with different concentrations of HT29-EVs. (<b>J</b>) Quantitative analysis of VEGFA expression in HUVECs treated with different concentrations of HT29-EVs. Data were analyzed by one-way ANOVA, * <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.</p>
Full article ">Figure 3 Cont.
<p>HT29-EVs promote angiogenesis in vitro. (<b>A</b>) HUVECs were treated with control PBS or HT29-EVs that had been labeled with PKH67 (green) for 6 h and stained with DAPI (blue) (scale bar = 13 μm). HT29-EVs uptake by HUVECs was observed. (<b>B</b>) The EdU assay indicated the effect of different concentrations of HT29-EVs on the proliferation rate of HUVECs (scale bar = 65 μm). (<b>C</b>) Immunofluorescence analysis of PCNA protein expression in HUVECs treated with different concentrations of HT29-EVs and fluorescence intensity statistics (scale bar = 25 μm). (<b>D</b>) The transwell assay was performed to detect the impact of different concentrations of HT29-EVs on the migration of HUVECs. (<b>E</b>) The scratch wound healing assay was performed to detect the impact of different concentrations of HT29-EVs on the migration of HUVECs (scale bar = 520 μm). (<b>F</b>) The FITC-phalloidin assay was performed to detect the effect of different concentrations of HT29-EVs on F-actin protein expression in HUVECs (scale bar = 25 μm). (<b>G</b>) The tube formation assay was used to detect the angiogenesis ability of HUVECs (scale bar = 200 μm). (<b>H</b>) Quantitative analysis of nodes, total tube length, and total loops (<a href="#app1-biology-13-00873" class="html-app">Figure S12</a>). (<b>I</b>) Western blotting analysis of VEGFA in HUVECs incubated with different concentrations of HT29-EVs. (<b>J</b>) Quantitative analysis of VEGFA expression in HUVECs treated with different concentrations of HT29-EVs. Data were analyzed by one-way ANOVA, * <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.</p>
Full article ">Figure 4
<p>HT29-EVs regulate angiogenesis possibly through the JAK/STAT3 pathway. (<b>A</b>) Differential expression profile of miRNAs in EVs derived from HT-29 and FHC cell lines from the GSE40247 dataset. (<b>B</b>) The top 30 hub genes for the target genes of DEMs between HT29-EVs and FHC-EVs. (<b>C</b>) GO enrichment analysis of the hub genes of target genes for DEMs between HT29-EVs and FHC-EVs. (<b>D</b>) KEGG pathway analysis of the hub genes of target genes for DEMs between HT29-EVs and FHC-EVs.</p>
Full article ">Figure 5
<p>Stattic inhibits the pro-angiogenic effect of HT29-EVs. (<b>A</b>) Western blotting analysis of the protein expressions of STAT3, p-STAT3, and VEGFA in HUVECs (<a href="#app1-biology-13-00873" class="html-app">Figure S13</a>). (<b>B</b>) Immunofluorescence analysis of p-STAT3 and VEGFA protein expression in HUVECs (scale bar = 50 μm). (<b>C</b>) The EdU assay was carried out to measure HUVECs proliferation (scale bar = 65 μm). (<b>D</b>) Immunofluorescence analysis of PCNA protein expression in HUVECs and fluorescence intensity statistics (scale bar = 25 μm). (<b>E</b>) The transwell assay was performed to detect HUVECs migration. (<b>F</b>) The scratch wound healing assay was performed to detect HUVECs migration (scale bar = 250 μm). (<b>G</b>) The FITC-phalloidin assay was performed to detect F-actin protein expression in HUVECs (F-actin (green) and nuclei (DAPI, blue)). (<b>H</b>) The tube formation assay was used to detect the angiogenesis ability of HUVECs (scale bar = 520 μm) and quantitative analysis of nodes, total tube length, and total loops. Data were analyzed by <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, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>Stattic inhibits the pro-angiogenic effect of HT29-EVs. (<b>A</b>) Western blotting analysis of the protein expressions of STAT3, p-STAT3, and VEGFA in HUVECs (<a href="#app1-biology-13-00873" class="html-app">Figure S13</a>). (<b>B</b>) Immunofluorescence analysis of p-STAT3 and VEGFA protein expression in HUVECs (scale bar = 50 μm). (<b>C</b>) The EdU assay was carried out to measure HUVECs proliferation (scale bar = 65 μm). (<b>D</b>) Immunofluorescence analysis of PCNA protein expression in HUVECs and fluorescence intensity statistics (scale bar = 25 μm). (<b>E</b>) The transwell assay was performed to detect HUVECs migration. (<b>F</b>) The scratch wound healing assay was performed to detect HUVECs migration (scale bar = 250 μm). (<b>G</b>) The FITC-phalloidin assay was performed to detect F-actin protein expression in HUVECs (F-actin (green) and nuclei (DAPI, blue)). (<b>H</b>) The tube formation assay was used to detect the angiogenesis ability of HUVECs (scale bar = 520 μm) and quantitative analysis of nodes, total tube length, and total loops. Data were analyzed by <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, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
18 pages, 5164 KiB  
Article
Identification of Specific microRNAs in Adipose Tissue Affected by Lipedema
by Erika Cione, Sandro Michelini, Diana Marisol Abrego-Guandique, Nicola Vaia, Serena Michelini, Valeria Puleo, Matteo Bertelli, Maria Cristina Caroleo and Roberto Cannataro
Curr. Issues Mol. Biol. 2024, 46(11), 11957-11974; https://doi.org/10.3390/cimb46110710 - 25 Oct 2024
Viewed by 1167
Abstract
Lipedema is a chronic disorder affecting women with a 10% incidence worldwide. It is often confused with obesity. This study was undertaken to study microRNAs in lipedema tissue assessed by direct hybridization using the robust n-counter flex DX CE-IVD platform. The mean age [...] Read more.
Lipedema is a chronic disorder affecting women with a 10% incidence worldwide. It is often confused with obesity. This study was undertaken to study microRNAs in lipedema tissue assessed by direct hybridization using the robust n-counter flex DX CE-IVD platform. The mean age of the subjects participating in the study was 40.29 (±12.17). The mean body weight and BMI were 67.37 (±10.02) and 25.75 (±4.10), respectively. The lipedema stages included were I and II. The differential expressed human (hsa)-miRNAs were determined according to a log2 fold-change (LFC) of 0.5 and p value < 0.05. To these, increased expression of hsa-let-7g-5p was evident, as well as reduced levels of hsa-miR-371a-5p, -4454+7975, -365a+b-3p, -205-5p, -196a-5p, -4488, -2116-5p, -141-3p, -208a-3p, -302b-3p, 374a-5p, and -1297. Then, several bioinformatics tools were used to analyze microarray data focusing on validated target genes in silico. KEGG and Gene Ontology (GO) pathway enrichment analysis was conducted. Furthermore, the protein–protein interaction and co-expression network were analyzed using STRING and Cytoscape, respectively. The most upregulated miRNA mainly affected genes related to cell cycle, oocyte meiosis, and inflammatory bowel disease. The downregulated microRNAs were related to endocrine resistance, insulin resistance, hypersensitivity to AGE-RAGEs, and focal adhesion. Finally, we validated by RT-PCR the upregulated hsa-let-7g-5p and two down-regulated ones, hsa-miR-205-5p and hsa-miR-302b-3p, confirming microarray results. In addition, three mRNA target miRNAs were monitored, SMAD2, the target of the hsa-let-7g-5p, and ESR1 and VEGFA, the target of hsa-miR-205-5p and hsa-miR-302b-3p, respectively. Our results open a new direction for comprehending biochemical mechanisms related with the pathogenesis of lipedema, shedding light on this intricate pathophysiological condition that could bring to light possible biomarkers in the future. Full article
(This article belongs to the Section Bioinformatics and Systems Biology)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Volcano plot of lipedema versus healthy tissue. Blue spots represent downregulated miRNAs (hsa-miR-371a-5p, -4454+7975, -365a+b-3p, -205-5p, -196a-5p, -4488, -2116-5p, -141-3p, -208a-3p, -302b-3p, 374a-5p, and -1297), and the pink spot represent the upregulated miRNA (hsa-let-7g-5p). Gray spots represent miRNAs that did not show significant changes between the two groups. (<b>B</b>) Heatmap of differentially expressed miRNAs (<span class="html-italic">n</span> = 7 of lipedema tissue; <span class="html-italic">n</span> = 5 of healthy tissue).</p>
Full article ">Figure 2
<p>Gene Ontology analysis of target genes of miRNAs differentially expressed. (<b>A</b>–<b>C</b>) (left) correspond to the upregulated miRNA targets, and (<b>D</b>–<b>F</b>) (right) to the downregulated miRNA targets. The panels show the top 10 most represented Biological Processes (<b>A</b>,<b>D</b>), Cellular Components (<b>B</b>,<b>E</b>), and Molecular Functions (<b>C</b>,<b>F</b>) among the DE miRNA targets. The color of each dot represents the <span class="html-italic">p</span> value of each term involved in the analysis. The size of each dot represents the counts of overlapped genes between the input genes and the total gene list on the GO pathway.</p>
Full article ">Figure 3
<p>KEGG pathways analysis of target genes by miRNAs. (<b>A</b>) upregulated and (<b>B</b>) downregulated miRNAs of inflamed vs. healthy tissue. The color of each dot represents the <span class="html-italic">p</span> value of each term involved in the analysis. The size of each dot represents the counts of overlapped genes between the input genes and the total gene list on the KEGG pathway.</p>
Full article ">Figure 4
<p>Protein–protein interaction network. Each node is a protein, and an edge is an interaction between two proteins. The green circle indicates the protein, and the node size represents the degree of value. The node size represents the degree value.</p>
Full article ">Figure 5
<p>Network and Sub-network Protein–protein interaction of downregulated miRNAs. (<b>A</b>) Whole network protein–protein interaction. (<b>B</b>) Sub-network PTEN, ESR1, and SOX2. (<b>C</b>) Sub-network EGFR, VEGFA, and IGF1R. Each node is a protein, and an edge is an interaction between two proteins. The node size represents the degree value.</p>
Full article ">Figure 6
<p><b>RT-PCR analysis of miRNAs expression</b>. Relative mRNA levels in lipedema compared to healthy tissue: (<b>A</b>) hsa-let-7g-5p, (<b>B</b>) hsa-miR-205-5p, (<b>C</b>) hsa-miR-302b-3p. MiRNAs are normalized to the RNU48 RNA of each sample. Statistical analysis with <span class="html-italic">test-t</span> was as follows: * <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> ≤ 0.001. Data are mean ± SEM (<span class="html-italic">n</span> = 7 of lipedema tissue; <span class="html-italic">n</span> = 5 of healthy tissue).</p>
Full article ">Figure 7
<p><b>RT-PCR analysis of mRNAs</b>. Relative expression of mRNA levels in lipedema compared to healthy tissue: (<b>A</b>) SMAD2, (<b>B</b>) ESR1, (<b>C</b>) VEGFA; miRNAs are normalized to 18S RNA of each sample. Statistical analysis with <span class="html-italic">t</span>-test was as follows: * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> ≤ 0.0001. Data are mean ± SEM (<span class="html-italic">n</span> = 7 of lipedema tissue; <span class="html-italic">n</span> = 5 of healthy tissue).</p>
Full article ">
29 pages, 10148 KiB  
Article
Development of a Custom Fluid Flow Chamber for Investigating the Effects of Shear Stress on Periodontal Ligament Cells
by Mustafa Nile, Matthias Folwaczny, Andreas Kessler, Andrea Wichelhaus, Mila Janjic Rankovic and Uwe Baumert
Cells 2024, 13(21), 1751; https://doi.org/10.3390/cells13211751 - 23 Oct 2024
Viewed by 788
Abstract
The periodontal ligament (PDL) is crucial for maintaining the integrity and functionality of tooth-supporting structures. Mechanical forces applied to the tooth during orthodontic tooth movement generate pore pressure gradients, leading to interstitial fluid movement within the PDL. The generated fluid shear stress (FSS) [...] Read more.
The periodontal ligament (PDL) is crucial for maintaining the integrity and functionality of tooth-supporting structures. Mechanical forces applied to the tooth during orthodontic tooth movement generate pore pressure gradients, leading to interstitial fluid movement within the PDL. The generated fluid shear stress (FSS) stimulates the remodeling of PDL and alveolar bone. Herein, we present the construction of a parallel fluid-flow apparatus to determine the effect of FSS on PDL cells. The chamber was designed and optimized using computer-aided and computational fluid dynamics software. The chamber was formed by PDMS using a negative molding technique. hPDLCs from two donors were seeded on microscopic slides and exposed to FSS of 6 dyn/cm2 for 1 h. The effect of FSS on gene and protein expression was determined using RT-qPCR and Western blot. FSS upregulated genes responsible for mechanosensing (FOS), tissue formation (RUNX2, VEGFA), and inflammation (PTGS2/COX2, CXCL8/IL8, IL6) in both donors, with donor 2 showing higher gene upregulation. Protein expression of PTGS2/COX2 was higher in donor 2 but not in donor 1. RUNX2 protein was not expressed in either donor after FSS. In summary, FSS is crucial in regulating gene expression linked to PDL remodeling and inflammation, with donor variability potentially affecting outcomes. Full article
(This article belongs to the Special Issue Modelling Tissue Microenvironments in Development and Disease)
Show Figures

Figure 1

Figure 1
<p>Chamber design and chamber production. (<b>A</b>) Inner chamber dimension. Dimensions of the master chamber model (identical to the inner part of the chamber). (<b>B</b>–<b>D</b>) Chamber production using the negative molding technique. (<b>B</b>) The master model of the inner chamber (1) was glued to the 3D-printed flask consisting of a base and frame (2) using modeling wax. Threaded nozzles (3) were placed onto the chamber’s inlet and outlet and degassed PDMS was poured into the flask. (<b>C</b>) The final chamber is made from PDMS with threaded fittings. (<b>D</b>) Exploded 3D image of the final chamber with all parts including chamber closing frame (4), chamber closing lid (5), and polyurethane nano tape (6). The parts not made from PDMS were 3D printed using an SLA printer.</p>
Full article ">Figure 2
<p>Computational fluid flow shear stress simulation of the parallel flow chamber using Autodesk CFD (Autodesk, San Rafael, CA, USA). (<b>A</b>) The shear stress magnitude was determined to confirm the mathematical calculations and distribution of wall shear stress at 6 dyn/cm<sup>2</sup> across the desired chamber cell seeding area. Arrows represent the flow direction. (<b>B</b>) The velocity was simulated to determine undesirable phenomena such as turbulence. Streamline visualization of the flow field shows no flow turbulence at the seeding area of the flow chamber. Turbulence lines near the inlet and outlet are shown. (<b>C</b>) The region of consistent FSS was identified by fluid flow simulation (Autodesk CFD; Autodesk, San Rafael, CA, USA). The graphs depict FSS along the length (left) and across the middle of the chamber (right). A custom-made gasket was designed using Autodesk Inventor (Autodesk, San Rafael, CA, USA) (see <a href="#cells-13-01751-f003" class="html-fig">Figure 3</a> for details).</p>
Full article ">Figure 3
<p>Workflow of the experimental setup. (<b>A</b>) Custom-made culture well gaskets. The gasket was constructed by blocking the desired area of the glass slide using modeling wax and molded with 1:10 PDMS in cell culture dishes. (<b>B</b>) First, the gaskets were placed onto microscopic slides and coated with collagen. Second, cells were seeded in the gasket well at a density of 3 × 10<sup>5</sup> cells/cm<sup>2</sup> and incubated overnight. Third, the slides were loaded into a parallel flow chamber and secured using clamps, after which the two chambers were stimulated in parallel. The complete setup consists of (1) a water bath used to keep the culturing medium temperature at ~37 °C; (2) culturing medium reservoir; (3) a peristaltic pump; (4) a pulse damper; (5) a bubble trap composed of a T-connector and a valve; (6) parallel flow chamber; (7) clamps; (8) silicon tubing (black: chamber 1; red: chamber 2).</p>
Full article ">Figure 4
<p>Calibration of the temperature in the parallel flow chamber. The temperature within the chamber was calibrated with the water heating bath using a digital thermometer implanted within the parallel flow chamber using a fluid flow rate of 166.67 mL/min.</p>
Full article ">Figure 5
<p>Cell attachment of human periodontal ligament cells (hPDLCs) and human osteosarcoma cell line (SaOS-2) was assessed by microscopy before and after applying FSS. Microscopic images of cells growing in the corners and center of the seeding area of the microscopic slide are shown. (Scale bar: 1000 μm).</p>
Full article ">Figure 6
<p>Cell viability of human periodontal ligament cells (hPDLCs) and human osteosarcoma cell line (SaOS-2) was assessed by live/dead cell staining. Microscopic images of cells growing in the center of the seeding area of the microscopic slide are shown. Live cells are indicated by calcein AM staining (green), and dead cells are indicated by ethidium homodimer-1 (EthD-1) staining (red arrows). (Scale bar: 400 μm).</p>
Full article ">Figure 7
<p>Reference gene primer stability was assessed using RefFinder [<a href="#B25-cells-13-01751" class="html-bibr">25</a>]. (<b>a</b>) Descriptive statistics of the Cq values of the reference gene panel (FSS: 1 h FSS) (n = 4); Control: negative control (n = 4); All: FSS and control groups combined (n = 8). (<b>b</b>) The result from the comprehensive analysis of gene stability for the reference gene panel from RefFinder. Lower values in this analysis correspond to higher gene stability.</p>
Full article ">Figure 8
<p>Gene expression of the early mechanosensitive responder <span class="html-italic">FOS</span> after 1 h fluid shear stress. Each test group is represented by the mean (━), with error bars that indicate the standard deviation (SD). The 2<sup>−ΔΔCq</sup> technique was used, with <span class="html-italic">RPL0</span> and <span class="html-italic">RPL22</span> as reference genes. The differences between the test and control groups were evaluated using the Mann-Whitney U Test. Groups with significant differences are highlighted as follows: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 9
<p>Gene expression of inflammation-related genes after 1 h fluid shear stress: (<b>A</b>) <span class="html-italic">PTGS2</span>, (<b>B</b>) <span class="html-italic">CXCL8</span> (<span class="html-italic">IL8)</span>, and (<b>C</b>) <span class="html-italic">IL6</span>. Each test group is represented by the mean (━), with error bars that indicate the standard deviation (SD). The 2<sup>−ΔΔCq</sup> technique was used, with <span class="html-italic">RPL0</span> and <span class="html-italic">RPL22</span> as reference genes. The differences between the test and control groups were evaluated using the Mann-Whitney U Test. Groups with significant differences are highlighted as follows: * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 10
<p>Gene expression of osteogenic differentiation-related genes after 1 h fluid shear stress: (<b>A</b>) <span class="html-italic">RUNX2</span>, (<b>B</b>) <span class="html-italic">VEGFA</span>, (<b>C</b>) <span class="html-italic">TNFRSF11B</span>, and (<b>D</b>) <span class="html-italic">SP7</span>. Each test group is represented by the mean (━), with error bars that indicate the standard deviation (SD). The 2<sup>−ΔΔCq</sup> technique was used, with <span class="html-italic">RPL0</span> and <span class="html-italic">RPL22</span> as reference genes. The differences between the test and control groups were evaluated using the Mann-Whitney U Test. Groups with significant differences are highlighted as follows: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 11
<p>Western blot analysis of PTGS2/COX2 and RUNX2 proteins after 1 h fluid shear stress. FSS induced the expression of PTGS2 but not RUNX2. Lysates from donor 1 (<b>A</b>) and donor 2 (<b>B</b>) from 1 h FSS, the corresponding control (Ctrl), and the positive controls for GAPDH (HeLa), COX2, and RUNX2 (both expressed in baculovirus-insect cells) were separated by PAGE on a 14% SDS gel and transferred onto a PVDF membrane.</p>
Full article ">Figure 12
<p>Force-related tooth displacement alters PDL dynamics (pore size, pore pressure, flow rate, and FSS). (<b>A</b>) The amount of displacement was presented in a table. As illustrated, the greatest displacement happens during the first hour using a force of 1 N [modified from [<a href="#B18-cells-13-01751" class="html-bibr">18</a>]. (<b>B</b>,<b>C</b>) Conceptual representation of FSS magnitude during low and high orthodontic force. By applying low orthodontic force, the duration of fluid flow will be longer with a lower FSS magnitude over time and vice versa.</p>
Full article ">
9 pages, 233 KiB  
Communication
The Effects of VEGF-A and GSTM1/GSTT1 Variants in the Susceptibility to the Chronic Rhinosinusitis with Nasal Polyposis: A Pilot Genetic Study
by Leandro Azevedo Camargo, Angela Adamski da Silva Reis, Stela Oliveira Rodrigues, Rodrigo da Silva Santos and Melissa Ameloti Gomes Avelino
Biomedicines 2024, 12(10), 2383; https://doi.org/10.3390/biomedicines12102383 - 18 Oct 2024
Viewed by 587
Abstract
Nasal polyps (NPs) are usually part of chronic rhinosinusitis with nasal polyposis (CRSwNP). However, the exact etiology of CRSwNP is still unknown. In addition, the suggested etiological causes are infection, allergy, and immunological disorders, among others, such as genetic predisposition. Moreover, it is [...] Read more.
Nasal polyps (NPs) are usually part of chronic rhinosinusitis with nasal polyposis (CRSwNP). However, the exact etiology of CRSwNP is still unknown. In addition, the suggested etiological causes are infection, allergy, and immunological disorders, among others, such as genetic predisposition. Moreover, it is also suggested that oxygen-free radicals play a vital role in the pathogenesis of nasal polyposis, and inflammatory cells produce free radicals during phagocytosis, which is the primary source of ROS, controlled by the glutathione S-transferase (GST) system. Although, vascular endothelial growth factor (VEGF) plays an important role in angiogenesis, it is closely interwoven with the mobilization of inflammatory cells. This pilot study evaluated the association between genetic variant VEGF-A (rs28357093) and GSTM1/GSTT1 deletion polymorphism in susceptibility to CRSwNP. A case–control study was conducted with 61 individuals diagnosed with CRSwNP and 100 healthy subjects. VEGF-A (rs28357093) and GSTM1/GSTT1 deletion polymorphisms were genotyped by RFLP-PCR and SYBR Green real-time PCR, respectively. Individuals with allergic rhinitis carriers with AC genotype (rs28357093) presented a 4-fold increased risk to CRSwNP (OR = 4.20, 95% CI = 1.31 to 13.50; p = 0.015). This evidence shows that the increased vascular permeability probably causes an inflamed nasal area leading to extensive edema and polyp growth. On the other hand, no association was verified for each genetic variant by inheritance models. Interestingly, the GSTT1 present genotype showed a protective effect on CRSwNP. In conclusion, additional studies that have larger groups in different geographic localizations may be useful to verify and assess the association between genetic variants and CRSwNP. Full article
(This article belongs to the Section Immunology and Immunotherapy)
12 pages, 5452 KiB  
Article
Enhancing Bone Formation Through bFGF-Loaded Mesenchymal Stromal Cell Spheroids During Fracture Healing in Mice
by Kugo Takeda, Hiroki Saito, Shintaro Shoji, Hiroyuki Sekiguchi, Mitsuyoshi Matsumoto, Masanobu Ujihira, Masayuki Miyagi, Gen Inoue, Masashi Takaso and Kentaro Uchida
Bioengineering 2024, 11(10), 1041; https://doi.org/10.3390/bioengineering11101041 - 18 Oct 2024
Viewed by 688
Abstract
This study aimed to evaluate the osteogenic potential of mesenchymal stromal cell (MSC) spheroids combined with the basic fibroblast growth factor (bFGF) in a mouse femur fracture model. To begin, MSC spheroids were generated, and the expression of key trophic factors (bFGF Bmp2 [...] Read more.
This study aimed to evaluate the osteogenic potential of mesenchymal stromal cell (MSC) spheroids combined with the basic fibroblast growth factor (bFGF) in a mouse femur fracture model. To begin, MSC spheroids were generated, and the expression of key trophic factors (bFGF Bmp2, and Vegfa) was assessed using quantitative PCR (qPCR). A binding assay confirmed the interaction between the bFGF and the spheroids’ extracellular matrix. The spheroid cultures significantly upregulated bFGF, Bmp2, and Vegfa expression compared to the monolayers (p < 0.001), and the binding assay demonstrated effective bFGF binding to the MSC spheroids. Following these in vitro assessments, the mice were divided into five groups for the in vivo study: (1) no treatment (control), (2) spheroids alone, (3) bFGF alone, (4) bFGF-loaded spheroids (bFGF-spheroids), and (5) non-viable (frozen) bFGF-loaded spheroids (bFGF-dSpheroids). Bone formation was analyzed by a micro-CT, measuring the bone volume (BV) and bone mineral content (BMC) of the mice four weeks post-fracture. A high dose of the bFGF (10 µg) significantly promoted bone formation regardless of the presence of spheroids, as evidenced by the increases in BV (bFGF, p = 0.010; bFGF-spheroids, p = 0.006; bFGF-dSpheroids, p = 0.032) and BMC (bFGF, p = 0.023; bFGF-spheroids, p = 0.004; bFGF-dSpheroids, p = 0.014), compared to the controls. In contrast, a low dose of the bFGF (1 µg) combined with the MSC spheroids significantly increased BV and BMC compared to the control (BV, p = 0.012; BMC, p = 0.015), bFGF alone (BV, p = 0.012; BMC, p = 0.008), and spheroid (BV, p < 0.001; BMC, p < 0.001) groups. A low dose of the bFGF alone did not significantly promote bone formation (p > 0.05). The non-viable (frozen) spheroids loaded with a low dose of the bFGF resulted in a higher BV and BMC compared to the spheroids alone (BV, p = 0.003; BMC, p = 0.017), though the effect was less pronounced than in the viable spheroids. These findings demonstrate the synergistic effect of the bFGF and MSC spheroids on bone regeneration. The increased expression of the BMP-2 and VEGF observed in the initial experiments, coupled with the enhanced bone formation in vivo, highlight the therapeutic potential of this combination. Future studies will aim to elucidate the underlying molecular mechanisms and assess the long-term outcomes for bone repair strategies. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flow cytometric analysis of KUM10 cells. Flow cytometry histograms showing the expression of surface markers in KUM10 cells. The (<b>A</b>) CD45, a pan-leukocyte marker, (<b>B</b>) CD11b, a macrophage marker, and (<b>C</b>) CD31, an endothelial cell marker, all show negative expression in KUM10 cells. In contrast, the mesenchymal stem cell markers (<b>D</b>) Sca1, (<b>E</b>) PDGFR-α, and (<b>F</b>) CD29 show positive expression in KUM10 cells. Blue histograms represent the isotype control, while red histograms represent specific antibody staining.</p>
Full article ">Figure 2
<p>Trophic factor-related gene expression in monolayer- and spheroid-cultured KUM10 cells. (<b>A</b>) Phase-contrast microscopy images of monolayer cells and spheroids. Scale bar indicates 200 μm. (<b>B</b>–<b>D</b>) qPCR analysis results for <span class="html-italic">bFGF</span> (<b>B</b>), <span class="html-italic">Bmp2</span> (<b>C</b>), and <span class="html-italic">Vegfa</span> (<b>D</b>). * indicates <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Binding assay showing (<b>A</b>) fluorescence microscopy images: scale bar indicates 1 mm. (<b>B</b>) Amount of bFGF bound to the spheroids.</p>
Full article ">Figure 4
<p>Spheroid injection into the fracture site. (<b>A</b>) Spheroid injection into the fracture site using a micropipette. The arrow indicates the fracture line, and the arrowheads point to the injected spheroids. (<b>B</b>) Enlarged view of the area highlighted by the white dashed box in (<b>A</b>). The arrowhead indicates the spheroids. (<b>C</b>) Post-injection image of the fracture site. The arrow indicates the fracture line, and the arrowheads point to the injected spheroids. Scale bar indicates 5 mm.</p>
Full article ">Figure 5
<p>Micro-CT analysis of femurs following transplantation of high-dose bFGF-loaded spheroids. Representative 3D micro-CT images of fractured femurs from the following groups: (<b>A</b>) control, (<b>B</b>) bFGF, (<b>C</b>) Spheroid, (<b>D</b>) bFGF-loaded spheroid (bFGF-spheroid), and (<b>E</b>) bFGF-loaded dead spheroid (bFGF-dSpheroid). (<b>F</b>,<b>G</b>) Quantification of callus area and bone mineral content at the fracture site 4 weeks post-fracture. (<b>F</b>) Analysis of bone volume (mm<sup>3</sup>) in calluses from the control, spheroid, bFGF, bFGF-loaded spheroid (bFGF-spheroid), and bFGF-loaded dead spheroid (bFGF-dSpheroid) groups. (<b>G</b>) Analysis of bone mineral content (mg) in the same groups. Data are presented as mean ± SD (<span class="html-italic">n</span> = 5). “a” indicates statistical significance (<span class="html-italic">p</span> &lt; 0.05) compared to the control group. “b” indicates statistical significance (<span class="html-italic">p</span> &lt; 0.05) compared to the spheroid group.</p>
Full article ">Figure 6
<p>Micro-CT analysis of femurs following transplantation of low-dose bFGF-Loaded spheroids. Representative 3D micro-CT images of fractured femurs from the following groups: (<b>A</b>) control, (<b>B</b>) bFGF, (<b>C</b>) Spheroid, (<b>D</b>) bFGF-loaded spheroid (bFGF-spheroid), and (<b>E</b>) bFGF-loaded dead spheroid (bFGF-dSpheroid). (<b>F</b>,<b>G</b>) Quantification of callus area and bone mineral content at the fracture site 4 weeks post-fracture. (<b>F</b>) Analysis of bone volume (mm<sup>3</sup>) in calluses from the control, spheroid, bFGF, bFGF-loaded spheroid (bFGF-spheroid), and bFGF-loaded dead spheroid (bFGF-dSpheroid) groups. (<b>G</b>) Analysis of bone mineral content (mg) in the same groups. Data are presented as mean ± SD (<span class="html-italic">n</span> = 10). “a” indicates statistical significance (<span class="html-italic">p</span> &lt; 0.05) compared to the control group. “b” indicates statistical significance (<span class="html-italic">p</span> &lt; 0.05) compared to the spheroid group.</p>
Full article ">
Back to TopTop