[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
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
remove_circle_outline

Search Results (3,298)

Search Parameters:
Keywords = VEGF

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1909 KiB  
Review
Peptide Lv and Angiogenesis: A Newly Discovered Angiogenic Peptide
by Dylan L. Pham, Kelsey Cox, Michael L. Ko and Gladys Y.-P. Ko
Biomedicines 2024, 12(12), 2851; https://doi.org/10.3390/biomedicines12122851 (registering DOI) - 15 Dec 2024
Viewed by 338
Abstract
Peptide Lv is a small endogenous secretory peptide with ~40 amino acids and is highly conserved among certain several species. While it was first discovered that it augments L-type voltage-gated calcium channels (LTCCs) in neurons, thus it was named peptide “Lv”, it can [...] Read more.
Peptide Lv is a small endogenous secretory peptide with ~40 amino acids and is highly conserved among certain several species. While it was first discovered that it augments L-type voltage-gated calcium channels (LTCCs) in neurons, thus it was named peptide “Lv”, it can bind to vascular endothelial growth factor receptor 2 (VEGFR2) and has VEGF-like activities, including eliciting vasodilation and promoting angiogenesis. Not only does peptide Lv augment LTCCs in neurons and cardiomyocytes, but it also promotes the expression of intermediate-conductance KCa channels (KCa3.1) in vascular endothelial cells. Peptide Lv is upregulated in the retinas of patients with early proliferative diabetic retinopathy, a disease involving pathological angiogenesis. This review will provide an overview of peptide Lv, its known bioactivities in vitro and in vivo, and its clinical relevance, with a focus on its role in angiogenesis. As there is more about peptide Lv to be explored, this article serves as a foundation for possible future developments of peptide Lv-related therapeutics to treat or prevent diseases. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

Figure 1
<p>Peptide Lv is encoded as a propeptide in the <span class="html-italic">Vstm4</span> gene. Created with Biorender.com.</p>
Full article ">Figure 2
<p>Sequence homology of peptide Lv among four different species: mouse, rat, human, and chicken. Created with Biorender.com.</p>
Full article ">Figure 3
<p>Stages of vascular endothelial cell-mediated angiogenesis. The first stage of angiogenesis is initiated by the reaction to various factors, including VEGF. Local vasodilation and increased permeability occur in response to VEGF. An increase in MMPs, followed by basement membrane degradation ensues. A sprout then forms, followed by the proliferation and migration of endothelial stalk cells, led by a tip cell. Once the vessel tube is formed, the vessel will then mature through its association with pericytes. Created by Biorender.com.</p>
Full article ">Figure 4
<p>Various ion channels are involved in hyperpolarization. Endothelial K<sup>+</sup> channels, such as K<sub>Ca</sub>3.1, K<sub>Ca</sub>2.3, and Kir channels, release K<sup>+</sup>, leading to endothelial hyperpolarization. The electrical signal can be propagated to smooth muscle cells through myoendothelial junctions (MEJs). K<sup>+</sup> efflux into the myoendothelial space can activate smooth muscle Kir channels and Na<sup>+</sup>/K<sup>+</sup> pumps, leading to smooth muscle hyperpolarization. TRPV4 channel opening leads to increased calcium influx that can activate K<sub>Ca</sub> channels. Created with Biorender.com.</p>
Full article ">
14 pages, 5653 KiB  
Article
Uterine Repair Mechanisms Are Potentiated by Mesenchymal Stem Cells and Decellularized Tissue Grafts Through Elevated Vegf, Cd44, and Itgb1 Gene Expression
by Sara Bandstein, Lucia De Miguel-Gómez, Edina Sehic, Emy Thorén, Sara López-Martínez, Irene Cervelló, Randa Akouri, Mihai Oltean, Mats Brännström and Mats Hellström
Bioengineering 2024, 11(12), 1268; https://doi.org/10.3390/bioengineering11121268 - 14 Dec 2024
Viewed by 267
Abstract
Transplantation of decellularized uterus tissue showed promise in supporting regeneration following uterine injury in animal models, suggesting an alternative to complete uterus transplantation for uterine factor infertility treatment. However, most animal studies utilized small grafts, limiting their clinical relevance. Hence, we used larger [...] Read more.
Transplantation of decellularized uterus tissue showed promise in supporting regeneration following uterine injury in animal models, suggesting an alternative to complete uterus transplantation for uterine factor infertility treatment. However, most animal studies utilized small grafts, limiting their clinical relevance. Hence, we used larger grafts (20 × 10 mm), equivalent to nearly one uterine horn in rats, to better evaluate the bioengineering challenges associated with structural support, revascularization, and tissue regeneration. We analyzed histopathology, employed immunohistochemistry, and investigated gene expression discrepancies in growth-related proteins over four months post-transplantation in acellular grafts and those recellularized (RC) with bone marrow-derived mesenchymal stem cells (bmMSCs). RC grafts exhibited less inflammation and faster epithelialization and migration of endogenous cells into the graft compared with acellular grafts. Despite the lack of a significant difference in the density of CD31 positive blood vessels between groups, the RC group demonstrated a better organized myometrial layer and an overall faster regenerative progress. Elevated gene expression for Vegf, Cd44, and Itgb1 correlated with the enhanced tissue regeneration in this group. Elevated Tgfb expression was noted in both groups, potentially contributing to the rapid revascularization. Our findings suggest that large uterine injuries can be regenerated using decellularized tissue, with bmMSCs enhancing the endogenous repair mechanisms. Full article
(This article belongs to the Section Regenerative Engineering)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Experimental design. MMP, metalloproteinases; bmMSCs, bone marrow derived mesenchymal stem cells.</p>
Full article ">Figure 2
<p>Decision diagram for the evaluation of uterine tissue regeneration. Created with Lucidchart software.</p>
Full article ">Figure 3
<p>Decellularization of the rat uterus was confirmed by the white aspect of the organ (<b>A</b>) and by cell nuclei removal by DAPI staining before and after decellularization (<b>B</b>). Recellularization using GPF-labeled rat bone marrow derived mesenchymal stem cells and was assessed by immunofluorescence (anti-GFP ab290 from Abcam, 1:100 + DAPI stain) (<b>C</b>). The decellularized uterus was cut into 2 × 1 cm patches (<b>D</b>) for further transplantation into rats (<b>E</b>). Tissue regeneration was evaluated one month and four months post-transplantation (<b>F</b>,<b>G</b>). The graft is marked as a white dotted line, and the asterisks mark the native uterine horn. Scale bars: (<b>A</b>,<b>D</b>–<b>G</b>) = 500 µm; (<b>B</b>,<b>C</b>) = 75 µm.</p>
Full article ">Figure 4
<p>Histological assessment with hematoxylin and eosin staining (H&amp;E) and Masson’s trichrome staining (MT) on acellular grafts (DC) (<b>A</b>,<b>B</b>) and grafts recellularized with bone marrow-derived mesenchymal stem cells (RC) (<b>C</b>,<b>D</b>), one month (1 m) and four months (4 m).</p>
Full article ">Figure 5
<p>Morphological assessment of uterine tissue regeneration; evaluation of uterine thickness (<b>A</b>), number of glands (<b>B</b>,<b>C</b>), luminal epithelium morphology (<b>D</b>–<b>G</b>), myometrium regeneration (<b>G</b>–<b>I</b>). Immune labeling: (<b>C</b>–<b>F</b>,<b>H</b>,<b>I</b>), Ecad = red, aSMA = green, DAPI = blue; Scale bars; (<b>C</b>–<b>F</b>,<b>H</b>,<b>I</b>) = 50 µm. * <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 6
<p>Morphological assessment of uterine tissue regeneration by blood vessel quantification. (<b>A</b>,<b>B</b>) CD31 = red, blue-black = DNA; Scale bars; (<b>A</b>,<b>B</b>) = 100 µm. Black arrows specify identified blood vessels that were quantified (<b>C</b>).</p>
Full article ">Figure 7
<p>The gene expression for <span class="html-italic">Ki67</span> (<b>A</b>), <span class="html-italic">Vegf</span> (<b>B</b>), <span class="html-italic">Tgfb</span> (<b>C</b>), <span class="html-italic">Il1b</span> (<b>D</b>), <span class="html-italic">Cd44</span> (<b>E</b>), <span class="html-italic">Itgb1</span> (<b>F</b>), and <span class="html-italic">Cx</span>cl12 (<b>G</b>) was analyzed in cells within the scaffolds one and four months after transplantation and were compared to native uterus tissue gene expression. We confirmed an upregulation in <span class="html-italic">Vegf</span>, <span class="html-italic">Tgfb</span>, and <span class="html-italic">Cd44</span> in all grafts, and mesenchymal stem cells prolonged the elevated levels of <span class="html-italic">Ki67</span>, <span class="html-italic">Itgb1</span>, and <span class="html-italic">Vegf</span> genes. Expression plotted as Log<sub>2</sub>(fold change). * <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.</p>
Full article ">
21 pages, 35452 KiB  
Article
Assessment of Agrimonia eupatoria L. and Lipophosphonoxin (DR-6180) Combination for Wound Repair: Bridging the Gap Between Phytomedicine and Organic Chemistry
by Miriam Kaňuchová, Veronika Brindza Lachová, Kateřina Bogdanová, Jana Sabová, Petra Bonová, Tomáš Vasilenko, Ivan Kováč, Martin Novotný, Petra Mitrengová, Nitjawan Sahatsapan, Matúš Čoma, Emil Švajdlenka, Milan Kolář, Peter Bohuš, Pavel Mučaji, Robert Zajíček, Dominik Rejman and Peter Gál
Biomolecules 2024, 14(12), 1590; https://doi.org/10.3390/biom14121590 - 12 Dec 2024
Viewed by 341
Abstract
Agrimonia eupatoria L. (AE) has a rich tradition of use in wound healing improvement across various cultures worldwide. In previous studies, we revealed that Agrimonia eupatoria L. water extract (AE) possesses a rich polyphenolic composition, displaying remarkable antioxidant properties. Our investigations also demonstrated [...] Read more.
Agrimonia eupatoria L. (AE) has a rich tradition of use in wound healing improvement across various cultures worldwide. In previous studies, we revealed that Agrimonia eupatoria L. water extract (AE) possesses a rich polyphenolic composition, displaying remarkable antioxidant properties. Our investigations also demonstrated that lipophosphonoxin (LPPO) exhibited antibacterial efficacy in vitro while preserving the proliferation and differentiation of fibroblasts and keratinocytes. Building upon our prior findings, in this study, we intended to examine whether a combination of AE and LPPO could enhance skin wound healing while retaining antibacterial attributes. The antibacterial activity of AE/LPPO against Staphylococcus aureus was evaluated, alongside its effects on fibroblast-to-myofibroblast transition, the formation of extracellular matrix (ECM), and endothelial cells and keratinocyte proliferation/phenotype. We also investigated AE/LPPO’s impact on TGF-β1 and VEGF-A signaling in keratinocytes/fibroblasts and endothelial cells, respectively. Additionally, wound healing progression in rats was examined through macroscopic observation and histological analysis. Our results indicate that AE/LPPO promotes myofibroblast-like phenotypic changes and augments ECM deposition. Clinically relevant, the AE/LPPO did not disrupt TGF-β1 and VEGF-A signaling and accelerated wound closure in rats. Notably, while AE and LPPO individually exhibited antibacterial activity, their combination did not lead to synergism, rather decreasing antibacterial activity, warranting further examination. These findings underscore substantial wound healing improvement facilitated by AE/LPPO, requiring further exploration in animal models closer to human physiology. Full article
(This article belongs to the Section Natural and Bio-derived Molecules)
Show Figures

Figure 1

Figure 1
<p>High-performance liquid chromatography (HPLC) analysis of the <span class="html-italic">Agrimonia eupatoria</span> L. (AE) water extract and the second-generation lipophosphonoxin (LPPO) DR-6180. (<b>A</b>) Analysis of AE (blue) and LPPO (red) mixture. (<b>B</b>) Analysis of LPPO (blue), showing two isomers represented by two distinct peaks.</p>
Full article ">Figure 2
<p>MTS assay of HaCaT keratinocytes (<b>A</b>–<b>C</b>), human dermal fibroblasts (HDF) (<b>D</b>–<b>F</b>), and human dermal microvascular vein endothelial cells (HMVEC-d) (<b>G</b>–<b>I</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive control in HaCaT and HDF, while VEGF-A was used in HMVEC-d. The figures were generated using GraphPad Prism software (GraphPad Software, San Diego, CA, USA), which was also utilized to logarithmically transform the concentration values and generate fitting curves to visualize the data.</p>
Full article ">Figure 3
<p>Wound healing (2D migration) assay of HaCaT keratinocytes (<b>A</b>) and human dermal microvascular vein endothelial cells—HMVEC-d (<b>B</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive control in HaCaT, while VEGF-A was used in HMVEC-d (Magnification 100×).</p>
Full article ">Figure 4
<p>Western blot (WB) analysis of HaCaT keratinocytes (<b>A</b>), human dermal fibroblasts—HDF (<b>B</b>), and human dermal microvascular vein endothelial cells—HMVEC-d (<b>C</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive control in HaCaT and HDF, while VEGF-A was used in HMVEC-d.</p>
Full article ">Figure 5
<p>Immunofluorescence of HaCaT keratinocytes (<b>A</b>) and human dermal fibroblasts—HDF (<b>B</b>) in the presence of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract, lipophosphonoxin (LPPO) DR-6180, and combination of AE and LPPO. TGF-β1 was used as the positive. Magnification 200×.</p>
Full article ">Figure 6
<p>Antimicrobial effect of AE + LPPO (DR-6180) combination tested by checkerboard microdilution assay in 69-well microtiter plate. Colors indicate wells with positive bacterial growth (yellow color for growth control (GC), and AE and DR-6180 alone; green for the combinations). The numbers next to the green areas are ∑FICs (FIC<sub>LPPO</sub> + FIC<sub>AE</sub>) values. ∑FIC &gt;1 to &lt; 2 is evaluated as indifference, and ∑FIC ≥ 2 is evaluated as antagonism.</p>
Full article ">Figure 7
<p>Wounds on rats from control (untreated), LavaSurg-treated (positive control, moist healing), and combination of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract and lipophosphonoxin (LPPO) DR-6180 on day 14 post surgery. Scale bar = 1 cm.</p>
Full article ">Figure 8
<p>Histological assessment of wounds (hematoxylin and eosin) control (untreated), LavaSurg-treated (positive control, moist healing), and combination of <span class="html-italic">Agrimonia eupatoria</span> L. (AE) extract and lipophosphonoxin (LPPO) DR-6180 on day 14 post surgery. Scale bar = 100 µm.</p>
Full article ">
31 pages, 3194 KiB  
Review
The Role of MicroRNAs in the Pathogenesis of Doxorubicin-Induced Vascular Remodeling
by Ekaterina Podyacheva, Julia Snezhkova, Anatoliya Onopchenko, Vyacheslav Dyachuk and Yana Toropova
Int. J. Mol. Sci. 2024, 25(24), 13335; https://doi.org/10.3390/ijms252413335 - 12 Dec 2024
Viewed by 246
Abstract
Doxorubicin (DOX), a cornerstone chemotherapeutic agent, effectively combats various malignancies but is marred by significant cardiovascular toxicity, including endothelial damage, chronic heart failure, and vascular remodeling. These adverse effects, mediated by oxidative stress, mitochondrial dysfunction, inflammatory pathways, and dysregulated autophagy, underscore the need [...] Read more.
Doxorubicin (DOX), a cornerstone chemotherapeutic agent, effectively combats various malignancies but is marred by significant cardiovascular toxicity, including endothelial damage, chronic heart failure, and vascular remodeling. These adverse effects, mediated by oxidative stress, mitochondrial dysfunction, inflammatory pathways, and dysregulated autophagy, underscore the need for precise therapeutic strategies. Emerging research highlights the critical role of microRNAs (miRNAs) in DOX-induced vascular remodeling and cardiotoxicity. miRNAs, such as miR-21, miR-22, miR-25, miR-126, miR-140-5p, miR-330-5p, miR-146, miR-143, miR-375, miR-125b, miR-451, miR-34a-5p, and miR-9, influence signaling pathways like TGF-β/Smad, AMPKa/SIRT, NF-κB, mTOR, VEGF, and PI3K/AKT/Nrf2, impacting vascular homeostasis, angiogenesis, and endothelial-to-mesenchymal transition. Despite existing studies, gaps remain in understanding the full spectrum of miRNAs involved and their downstream effects on vascular remodeling. This review synthesizes the current knowledge on miRNA dysregulation during DOX exposure, focusing on their dual roles in cardiovascular pathology and tumor progression. Strategies to reduce DOX cardiotoxicity include modulating miRNA expression to restore signaling balance, targeting pro-inflammatory and pro-fibrotic pathways, and leveraging miRNA inhibitors or mimics. This review aims to organize and integrate the existing knowledge on the role of miRNAs in vascular remodeling, particularly in the contexts of DOX treatment and the progression of various cardiovascular diseases, including their potential involvement in tumor growth. Full article
Show Figures

Figure 1

Figure 1
<p>Alterations in the expressions of key known microRNAs during the development of specific cardiovascular pathologies (atherosclerosis, hypertension (arterial and pulmonary), and diabetic cardiovascular complications). ↑, overexpression; ↓, reduced expression.</p>
Full article ">Figure 2
<p>Effects of doxorubicin on microRNA expression in modulating apoptosis, mitochondrial biogenesis, ferroptosis, pyroptosis, endoplasmic reticulum (ER) stress, and endothelial dysfunction. ↑, overexpression; ↓, reduced expression.</p>
Full article ">Figure 3
<p>Key signaling pathways involved and their effects in vascular remodeling under doxorubicin exposure. ↑, activation; ↓, suppression; AKT, protein kinase B; AMPK, 5′ AMP-activated protein kinase; COX-2, cyclooxygenase-2; Cx43/Cx45, connexin 43/45; DOX, doxorubicin; ERK5, extracellular signal-regulated kinase 5; FOXO3a, forkhead box protein O3a; GSDMD, Gasdermin D; IL10, interleukin-10; JNK, c-Jun N-terminal kinases; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2, nuclear factor erythroid 2-related factor 2; PI3K, phosphoinositide 3-kinases; ROS, reactive oxygen species; SIRT, silent information regulator 2 protein; TGF-β, transforming growth factor beta; TNF-α, tumor necrosis factor alpha; VEGFR, vascular endothelial growth factor receptor; ZO-1, zonula occludens-1.</p>
Full article ">Figure 4
<p>The impact of doxorubicin on microRNA expression in vascular remodeling and tumor angiogenesis. ↑, activation; ↓, suppression.</p>
Full article ">
18 pages, 9960 KiB  
Article
Comparative Analysis of Extracorporeal Shockwave Therapy, Bisphosphonate, and Wharton Jelly-Derived Mesenchymal Stem Cells in Preserving Bone and Cartilage Integrity and Modulating IL31, IL33, and BMP2 in the Cartilage of Ovariectomized Rat Model
by Jai-Hong Cheng, Cheng-Wei Chen, Wen-Yi Chou, Po-Cheng Chen, Kuan-Ting Wu, Shun-Wun Jhan, Shan-Ling Hsu, Yi-No Wu and Hou-Tsung Chen
Biomedicines 2024, 12(12), 2823; https://doi.org/10.3390/biomedicines12122823 - 12 Dec 2024
Viewed by 276
Abstract
Background: Osteoporosis (OP) is a chronic inflammatory bone disease characterized by reduced bone structure and strength, leading to increased fracture risk. Effective therapies targeting both bone and cartilage are limited. This study compared the therapeutic effects of extracorporeal shockwave therapy (ESWT), bisphosphonate (Aclasta), [...] Read more.
Background: Osteoporosis (OP) is a chronic inflammatory bone disease characterized by reduced bone structure and strength, leading to increased fracture risk. Effective therapies targeting both bone and cartilage are limited. This study compared the therapeutic effects of extracorporeal shockwave therapy (ESWT), bisphosphonate (Aclasta), and human Wharton jelly-derived mesenchymal stem cells (WJMSCs) in a rat model of OP. Methods: Female rats were assigned to four groups: Sham (no surgery or treatment), OP (bilateral ovariectomy, OVX), ESWT (OVX + ESWT on both tibias at 0.25 mJ/mm2, 1500 impulses per tibia), Aclasta (OVX + zoledronic acid 0.1 mg/kg via tail vein injection), and WJMSC (OVX + 2 × 10⁶ WJMSCs). Pathological changes, bone microarchitecture (by micro-CT), serum cytokines (by ELISA), and tissue-specific molecular markers (by immunohistochemistry) were evaluated. Results: All treatments improved bone density, preserved cartilage, and modulated cytokines (IL31, IL33, VEGF, and BMP2), with Aclasta showing the greatest improvements in bone parameters and cartilage preservation. ESWT and WJMSC also demonstrated significant effects, with ESWT highlighting non-invasive chondroprotective potential. Conclusions: Aclasta provided the best overall therapeutic response, particularly in bone regeneration. However, ESWT and WJMSC also showed comparable chondroprotective effects. ESWT emerges as a promising non-invasive alternative for OP management when pharmacological or cell-based therapies are not feasible. Full article
(This article belongs to the Special Issue Diseases and Regeneration for Muscle, Joint and Bone)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The study design and application of ESWT. (<b>A</b>) Flowchart displaying the experiments and timing for knee surgery, shockwave application, Aclasta treatment, WJMSC treatments, and sacrifice of the animals. Eight rats were used for each group. (<b>B</b>) Focused ESWT application (0.25 mJ/mm<sup>2</sup>, 4 Hz, 1500 impulses for each location) on right and left tibia at 0.5 cm below the skin of rats at each location, indicated as black circles. OP group = osteoporosis, OVX = ovariectomy, ESWT group = extracorporeal shockwave therapy, WJMSC group = Wharton Jelly-derived mesenchymal stem cell treatment, micro-CT = micro computed tomography, IHC = immunohistochemistry, ELISA = enzyme-linked immunosorbent assay.</p>
Full article ">Figure 2
<p>Micro-CT scan of proximal femur, tibia, and spine in different groups. (<b>A</b>) The results displayed photomicrographs of the knee in transverse (tibia, up) and sagittal (femur and tibia, below) views from micro-CT. (<b>B</b>) The photomicrographs of the spine (T6 to T8) in transverse and sagittal views. The region of the red line was the region of interest in vertebral bone (T7). F indicated the femur and T indicated the tibia. R was the right lower limb and L was the left lower limb. Eight rats were used for each group. OP group was osteoporosis, OVX was ovariectomy, ESWT group was extracorporeal shockwave therapy, and WJMSC group was Wharton jelly-derived mesenchymal stem cell treatment.</p>
Full article ">Figure 3
<p>The microphotographs of the left knees and spine. (<b>A</b>) The hematoxylin and eosin staining showed the cartilage change in the articular cartilage and epiphyseal plate of the left tibia (×100 magnification) in different groups. (<b>B</b>) Safranin-O staining showing the articular cartilage of the left knee (1000 μm) and epiphyseal plate of the left tibia (×100 magnification). (<b>C</b>) Safranin-O staining showing the spine (×50 magnification), vertebral bone (×100 magnification), and vertebral cartilage (×100 magnification). Eight rats were used for each group. The red arrowhead indicates cartilage. The black arrowhead indicated epiphyseal plate. The black triangle indicated cancellous bone. The red triangle indicated primary spongiosa. The tibia indicated by T and the femur indicated by F. Osteoporosis indicated by OP group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group.</p>
Full article ">Figure 4
<p>Immunohistochemical analysis for (<b>A</b>) IL31, (<b>B</b>) IL33, and (<b>C</b>) ST2 in the articular cartilage (×100 magnification) of the left knee and epiphyseal plate of the left tibia in Sham, OP, ESWT, Aclasta, and WJMSC groups (right). The expression levels are assessed following the treatments (left). *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. Eight rats were used for each group. Osteoporosis indicated OP group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. T represented tibia. Red arrowhead indicated the expression of proteins.</p>
Full article ">Figure 4 Cont.
<p>Immunohistochemical analysis for (<b>A</b>) IL31, (<b>B</b>) IL33, and (<b>C</b>) ST2 in the articular cartilage (×100 magnification) of the left knee and epiphyseal plate of the left tibia in Sham, OP, ESWT, Aclasta, and WJMSC groups (right). The expression levels are assessed following the treatments (left). *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. Eight rats were used for each group. Osteoporosis indicated OP group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. T represented tibia. Red arrowhead indicated the expression of proteins.</p>
Full article ">Figure 5
<p>Immunohistochemical analysis for BMP2 in the articular cartilage of the (<b>A</b>) tibia, (<b>B</b>) femur, and (<b>C</b>) epiphyseal plate of the left tibia in Sham, OP, SW, Aclasta, and WJMSC groups (×100 magnification) (right). The expression level was assessed following the treatments (left). *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. T represented tibia and Eight rats were used for each group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. Red arrowhead indicated the expression of proteins.</p>
Full article ">Figure 6
<p>The immunohistochemical analysis of the vertebral cartilage in OVX rats. The immunohistochemical staining (×100 magnification, left panel) and the expression levels (right) of IL31, IL33, ST2, and BMP2 in the vertebral cartilage of the spines for each group. *** <span class="html-italic">p</span> &lt; 0.001 as compared with the OP group. Eight rats were used for each group. Extracorporeal shockwave therapy indicated ESWT group and Wharton jelly-derived mesenchymal stem cell treatment indicated WJMSC group. Red arrowhead indicated the expression of proteins.</p>
Full article ">
18 pages, 3242 KiB  
Systematic Review
Biological and Cellular Effects of Percutaneous Electrolysis: A Systematic Review
by Jacobo Rodríguez-Sanz, Sergi Rodríguez-Rodríguez, Carlos López-de-Celis, Miguel Malo-Urriés, Soledad Pérez-Amodio, Román Pérez-Antoñanzas, Sergio Borrella-Andrés, Isabel Albarova-Corral and Miguel Ángel Mateos-Timoneda
Biomedicines 2024, 12(12), 2818; https://doi.org/10.3390/biomedicines12122818 - 12 Dec 2024
Viewed by 412
Abstract
Background: Percutaneous electrolysis is an invasive physical therapy technique that is receiving attention. The objective of this article is to evaluate the biological and cellular effects of percutaneous electrolysis and its influence on tissue healing processes. Methods. The search strategy performed [...] Read more.
Background: Percutaneous electrolysis is an invasive physical therapy technique that is receiving attention. The objective of this article is to evaluate the biological and cellular effects of percutaneous electrolysis and its influence on tissue healing processes. Methods. The search strategy performed in PubMed, Cochrane Library, and Web of Sciences databases resulted in a total of 25 studies. Once inclusion and exclusion criteria were applied, seven studies were finally included in this systematic review. The biological effects of percutaneous electrolysis were evaluated and grouped into pro-inflammatory and anti-inflammatory effects, cell death, and extracellular matrix and tissue remodeling effects. Results. Percutaneous electrolysis generates a significant pro-inflammatory increase in the chronic tendon condition of IL1β-6-18-1α-1rn, NLRP3, and M1 polymorphonuclear cells and increased expression of COX2, TNFα, Cxcl10, and TGFβ1 during the first 7 days. This inflammation is regulated as of day 13. A significant increase in cell death markers, such as LDH, Yo-Pro, cytochrome C, and Smac/Diablo markers, was observed during the first 7 days. Finally, a significant increase in markers Mmp9, VEGF, VEGFR, PPAR-γ/tubulin, and COL-I was observed in the extracellular matrix and tissue remodeling, and a decrease in COL-III was observed during the first 7 days. In the acute inflammatory injury condition, an increase in anti-inflammatory markers, such as IL-10-13, CCL1, and IkB, and a significant decrease in pro-inflammatory cytokines, such as IL-6-1β, CCL3-4-5, CCR5-8, NFkB, and TNFα, were observed during the first 7 days. Finally, a significant increase in VEGF, VEGFR, and PPAR-γ/tubulin markers in the extracellular matrix and tissue remodeling was observed for this condition during the first 7 days. Conclusions. Percutaneous electrolysis generates a controlled local pro-inflammatory effect in chronic conditions and regulates inflammation in inflammatory injuries (during the first 7 days). Electrolysis has short-term effects (0–7 days post) of cell death and controlled extracellular matrix destruction. Additionally, it facilitates subsequent healing by improving extracellular matrix synthesis starting from 7 days after application. Full article
(This article belongs to the Special Issue Tendinopathy and Myopathy: From Molecular Basis to Therapy)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow chart.</p>
Full article ">Figure 2
<p>Risk of bias graph: review of authors’ judgements about each risk of bias item presented as percentages across all included studies.</p>
Full article ">Figure 3
<p>Risk of bias summary: review of authors’ judgements about each risk of bias item for each included study [<a href="#B5-biomedicines-12-02818" class="html-bibr">5</a>,<a href="#B6-biomedicines-12-02818" class="html-bibr">6</a>,<a href="#B7-biomedicines-12-02818" class="html-bibr">7</a>,<a href="#B8-biomedicines-12-02818" class="html-bibr">8</a>,<a href="#B9-biomedicines-12-02818" class="html-bibr">9</a>,<a href="#B10-biomedicines-12-02818" class="html-bibr">10</a>,<a href="#B11-biomedicines-12-02818" class="html-bibr">11</a>]. “+”: Low risk of bias. “?”: Unclear risk of bias.</p>
Full article ">Figure 4
<p>Diagram of the effects of percutaneous electrolysis depending on the environment of chronic tendon or acute muscle injury. Abbreviations: IL-6-10-13-18-1β-1α, interleukin-6-10-13-18-1β-1α; NLRP3, NOD-like receptor family pyrin domain containing 3; IL1rn, interleukin-1 receptor antagonist; TNFα, tumor necrosis factor alpha; Cxcl10, C–X–C motif chemokine ligand 10; TGFβ1, transforming growth factor beta 1; COX-2, cyclooxygenase-2; LDH, lactate dehydrogenase; Yo-Pro, Yo-Pro-1 (apoptosis marker); M1, macrophage phenotype 1; COL-I-III, collagen type I-III; Mmp9, matrix metalloproteinase 9; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; CCL1-3-4-5, C–C motif chemokine ligand 1-3-4-5; CCR5-8, C–C motif chemokine receptor 5-8; NFkB, nuclear factor kappa B; IkB, inhibitor of kappa B; Smac/Diablo, second mitochondria-derived activator of caspases/direct IAP-binding protein with low pI; PPAR-γ/tubulin, peroxisome proliferator-activated receptor gamma/tubulin; HK, hexokinase; PK, pyruvate kinase; FBPase, fructose-1,6-bisphosphatase; G6PDH, glucose-6-phosphate dehydrogenase; CS, citrate synthase; ME, malic enzyme; HOAD, 3-Hydroxyacyl-CoA dehydrogenase; FAS, fatty acid synthase; AST, aspartate aminotransferase; ALT, alanine aminotransferase.</p>
Full article ">
15 pages, 1759 KiB  
Article
Diagnostic and Prognostic Value of Angiogenic Status in Hereditary Hemorrhagic Telangiectasia
by Sherlyne Jaimes-Díaz, Gustavo Juan-Samper, Susana Torres-Martínez, Eva Escorihuela-Alares, Silvia Calabuig-Fariñas, Raquel Rodríguez-López, Nieves Prieto-Colodrero, Mercedes Ramon-Capilla and Estrella Fernández-Fabrellas
Diagnostics 2024, 14(24), 2783; https://doi.org/10.3390/diagnostics14242783 - 11 Dec 2024
Viewed by 276
Abstract
Background/Objectives: Angiogenesis is involved in the pathogenesis of hereditary hemorrhagic telangiectasia (HHT). VEGF, ANG2, TGFβ1, and ENG are the most studied angiogenic factors, but their clinical significance in blood samples is still not completely defined. The genetic study of HHT mutations is [...] Read more.
Background/Objectives: Angiogenesis is involved in the pathogenesis of hereditary hemorrhagic telangiectasia (HHT). VEGF, ANG2, TGFβ1, and ENG are the most studied angiogenic factors, but their clinical significance in blood samples is still not completely defined. The genetic study of HHT mutations is the test of choice for diagnosing the disease, but this route is expensive, and the causative mutation is not found in up to 10% of cases. Therefore, the use of angiogenic biomarkers could facilitate a cheaper and easier approach to the diagnosis of HHT. To determine the diagnostic and prognostic value of the VEGFA, TGFβ1, ANG2, and ENG plasmatic concentrations in patients with HHT. Methods: All the participants were clinically evaluated and the concentrations of these angiogenic factors were measured using MILLIPLEX®MAP immunoassays in plasma samples collected from 44 patients with HHT and 19 controls. To evaluate the diagnostic validity of these parameters, we estimated the maximum Youden index of the ROC curve and evaluated their diagnostic value using multiple logistic regression. Results: Patients with HHT had increased blood levels of TGFβ1 and decreased ENG compared to the control group. We could not identify any angiogenic markers related to the clinical severity or epistaxis. TGFβ1 and ENG exhibited a higher discriminant capacity for HHT, especially patients with HHT1, and it was possible to develop signatures of these factors with diagnostic value. Conclusions: We identified several angiogenic factors that may be important diagnostic biomarkers for HHT and propose that the combination of TGFβ1 and ENG could represent a signature with diagnostic value for this disease. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Box plots of the values of vascular endothelial growth factor (VEGFA), transforming growth factor beta 1 (TGFβ1), angiopoietin 2 (ANG2), and endoglin (ENG) in the studied population (patients with hereditary hemorrhagic telangiectasia type 1 or 2 and unaffected controls), with the boxes showing the median and interquartile ranges (25% and 75%) and the whiskers expressing the values in an acceptable range, with the outliers shown as circles and the extremes with asterisks.</p>
Full article ">Figure 2
<p>Graph relating the severity of epistaxis (epistaxis severity score (ESS): 0, not present; 1, mild, ≥1–≤4; 2, moderate, &gt;4–≤7; 3, severe, &gt;7–≤10) in patients with hereditary hemorrhagic telangiectasia against transforming growth factor beta 1 (TGFβ1) expression levels (<span class="html-italic">p</span> = 0.097).</p>
Full article ">Figure 3
<p>Receiver operating characteristic (ROC) curves for the transforming growth factor beta 1 (TGFβ1), vascular endothelial growth factor (VEGFA), angiopoietin 2 (ANG2), and endoglin (ENG) markers in patients with hereditary hemorrhagic telangiectasia.</p>
Full article ">
14 pages, 1279 KiB  
Article
Kallistatin as a Potential Marker of Therapeutic Response During Alpha-Lipoic Acid Treatment in Diabetic Patients with Sensorimotor Polyneuropathy
by Marcell Hernyák, László Imre Tóth, Sára Csiha, Ágnes Molnár, Hajnalka Lőrincz, György Paragh, Mariann Harangi and Ferenc Sztanek
Int. J. Mol. Sci. 2024, 25(24), 13276; https://doi.org/10.3390/ijms252413276 - 11 Dec 2024
Viewed by 310
Abstract
Diabetic sensorimotor neuropathy (DSPN) is strongly associated with the extent of cellular oxidative stress and endothelial dysfunction in type 2 diabetes (T2DM). Alpha-lipoic acid (ALA) attenuates the progression of DSPN through its antioxidant and vasculoprotective effects. Kallistatin has antioxidant and anti-inflammatory properties. We [...] Read more.
Diabetic sensorimotor neuropathy (DSPN) is strongly associated with the extent of cellular oxidative stress and endothelial dysfunction in type 2 diabetes (T2DM). Alpha-lipoic acid (ALA) attenuates the progression of DSPN through its antioxidant and vasculoprotective effects. Kallistatin has antioxidant and anti-inflammatory properties. We aimed to evaluate changes in kallistatin levels and markers of endothelial dysfunction in patients with T2DM and DSPN following six months of treatment with 600 mg/day of ALA. A total of 54 patients with T2DM and DSPN and 24 control patients with T2DM but without neuropathy participated in this study. The serum concentrations of kallistatin, ICAM-1, VCAM-1, oxLDL, VEGF, ADMA, and TNF-alpha were measured by an ELISA. Peripheral sensory neuropathy was assessed with neuropathy symptom questionnaires and determination of the current perception threshold. After ALA treatment, the level of kallistatin significantly decreased, as well as the levels of TNF-alpha and ADMA. Changes in kallistatin levels were positively correlated with changes in oxLDL. The improvement in DSPN symptoms following ALA treatment showed a positive correlation with changes in kallistatin, VEGF, oxLDL, and ADMA levels. Based on our results, kallistatin could represent a potential new biomarker for assessing therapeutic response during ALA treatment in patients with DSPN. Full article
(This article belongs to the Special Issue Molecular Diagnosis and Treatments of Diabetes Mellitus)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Concentrations of serum kallistatin in diabetic patients with neuropathy before and after 6 months of alpha-lipoic acid (ALA) treatment, as well as in diabetic controls without neuropathy. The boxes represent the interquartile range (25th–75th percentiles), with the 50th percentile (median) indicated by a solid line within the box. The whiskers denote the minimum and maximum values.</p>
Full article ">Figure 2
<p>Spearman’s correlation of the change in oxidized low-density lipoprotein (Δ oxidized LDL) levels with the change in kallistatin (Δ kallistatin) in type 2 diabetic patients with neuropathy during 6 months of alpha-lipoic acid (ALA) treatment.</p>
Full article ">Figure 3
<p>Spearman’s correlation of the change in the Neuropathy Total Symptom Score (NTSS) with (<b>a</b>) the change in kallistatin (Δ kallistatin); (<b>b</b>) the change in vascular endothelial growth factor (Δ VEGF); (<b>c</b>) the change in oxidized low-density lipoprotein (Δ oxidized LDL) levels; and (<b>d</b>) the change in asymmetric dimethylarginine (Δ ADMA) levels in type 2 diabetic patients with neuropathy during 6 months of alpha-lipoic acid (ALA) treatment.</p>
Full article ">
15 pages, 3251 KiB  
Article
Potential Use of Plasma Rich in Growth Factors in Age-Related Macular Degeneration: Evidence from a Mouse Model
by Eduardo Anitua, Francisco Muruzabal, Sergio Recalde, Patricia Fernandez-Robredo and Mohammad Hamdan Alkhraisat
Medicina 2024, 60(12), 2036; https://doi.org/10.3390/medicina60122036 - 10 Dec 2024
Viewed by 538
Abstract
Background and Objectives: Age-related macular degeneration (AMD) is the leading cause of low vision and legal blindness in adults in developed countries. Wet AMD can be successfully treated using vascular endothelial growth factor (VEGF) inhibitors; however, dry AMD currently has no effective [...] Read more.
Background and Objectives: Age-related macular degeneration (AMD) is the leading cause of low vision and legal blindness in adults in developed countries. Wet AMD can be successfully treated using vascular endothelial growth factor (VEGF) inhibitors; however, dry AMD currently has no effective treatment. The purpose of this study is to analyze the efficacy of intraocular injection of plasma rich in growth factors (PRGF) in an AMD mouse model induced by intraperitoneal administration of sodium iodate. Materials and Methods: Intravitreal application of PRGF (experimental group) and saline (control group) was performed immediately after intraperitoneal injection of sodium iodate. Retinographies were performed at 2 and 7 days after treatment administration. The eyes were retrieved for histological and immunohistological analysis. Statistical analysis was performed to compare the outcomes between the study groups. Results: In comparison to saline solution, PRGF significantly decreased the depigmentation of the RPE, showing a more reddened retina. PRGF intravitreal treatment significantly reduced the glial fibrillary acidic protein (GFAP) stained processes, suggesting a significant reduction in the risk of scar formation. Moreover, the myofibroblast invasion into the RPE cell layer was significantly reduced in the PRGF-treated group of mice. There was a tendency for better preservation of the photoreceptors in the PRGF group. Conclusions: Within the limitations of this study, intravitreal injection of PRGF provided significant protection against the degeneration of the photoreceptors and the RPE induced by the systemic administration of NaIO3. Full article
Show Figures

Figure 1

Figure 1
<p>Representative macroscopic images of retinas on the same day of treatment (t0) and after 2 and 7 days of treatment administration from female and male mice of the different groups (control, NaIO<sub>3</sub> -+ SS, and NaIO<sub>3</sub> + PRGF). The control group showed the typical retinal redness throughout the study period, while the disease group (NaIO<sub>3</sub> -+ SS) showed severe depigmentation of the retinal fundus at 2 days after intraperitoneal injection, which recovered at 7 days after treatment, but not to levels similar to the control group. The group of mice treated with PRGF (NaIO<sub>3</sub> + PRGF) showed significantly less retinal depigmentation than the disease group on day 2, with a more reddened retina. At day 7, retinal depigmentation was significantly reduced, showing a white patch throughout the retina, but this patch was smaller than in the disease group (NaIO<sub>3</sub> + SS). Macroscopic images show the different effects of NaIO<sub>3</sub> depending on the sex of the mice, showing a significant reduction effect on the retina of female mice compared to the retina of male mice.</p>
Full article ">Figure 2
<p>Representative histological images of female and male mice from the different treatment groups (control, NaIO<sub>3</sub> -+ SS, and NaIO<sub>3</sub> + PRGF) at two different magnifications (2× and 20×). Images at 20× magnification correspond to the black boxed area drawn in the lower magnification (2×) upper image. The histological images show remarkable retinal disorganization in the disease group of mice compared to the control group and the PRGF group, while the retinas of the PRGF-treated mice showed similar histological morphology to the control group.</p>
Full article ">Figure 3
<p>Immunohistochemical analysis of GFAP protein expression. The results showed that GFAP expression increased after intraperitoneal administration of NaIO<sub>3</sub> compared with the control group. On the other hand, intravitreal administration of PRGF reduced GFAP expression. However, no significant differences were observed between the different groups. Representative GFAP immunohistochemical images show an increased number of GFAP-positive processes throughout the retina of mice in the disease group (black arrowheads) compared with the control and PRGF groups. Noteworthy differences in GFAP protein expression were observed in relation to the mice’s genders.</p>
Full article ">Figure 4
<p>Smooth muscle alpha actin (SMA) expression in the mice retina of the different treatment groups. The squared area in the retina in each image has been enlarged in the upper left corner of the same image. SMA expression was significantly increased in the retinal choriocapillary area in mice in the disease group compared to the control group. PRGF treatment reduced SMA expression in the disease group. On the other hand, a clear difference in SMA expression was observed between female and male retinas.</p>
Full article ">Figure 5
<p>Immunohistochemical analysis of rhodopsin in the retina of treated mice. The results showed that rhodopsin was significantly reduced in the retina of mice in which NaIO<sub>3</sub> was administered intraperitoneally compared with the control group. On the other hand, PRGF increased rhodopsin expression with respect to the disease group, though without reaching statistical significance. Rhodopsin immunohistochemical imaging shows that, although the rhodopsin-positive area was markedly higher in the PRGF group than in the disease group, no significant differences were observed between them. The image in the upper left corner of each image corresponds to the magnification of the white-boxed retinal area of the same image. The enlarged image shows that the length of the photoreceptor outer segments, delimited between the black arrow and the white arrow, is significantly shorter in the retina of the disease group compared with the control group and the PRGF group. *: statistically significant (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
12 pages, 600 KiB  
Systematic Review
Bevacizumab for Vestibular Schwannomas in Neurofibromatosis Type 2: A Systematic Review of Tumor Control and Hearing Preservation
by Melina Screnci, Mathilde Puechmaille, Quentin Berton, Toufic Khalil, Thierry Mom and Guillaume Coll
J. Clin. Med. 2024, 13(23), 7488; https://doi.org/10.3390/jcm13237488 - 9 Dec 2024
Viewed by 464
Abstract
Background/Objectives: Vestibular schwannomas (VSs), also called acoustic neuromas, are benign tumors affecting the vestibulocochlear nerve, often leading to hearing loss and balance issues. This condition is particularly challenging in patients with neurofibromatosis type 2 (NF2), where VSs tend to develop bilaterally. Conventional [...] Read more.
Background/Objectives: Vestibular schwannomas (VSs), also called acoustic neuromas, are benign tumors affecting the vestibulocochlear nerve, often leading to hearing loss and balance issues. This condition is particularly challenging in patients with neurofibromatosis type 2 (NF2), where VSs tend to develop bilaterally. Conventional treatments, such as surgery and radiotherapy, although effective, carry risks like hearing loss and nerve damage. Bevacizumab, a VEGF-targeting monoclonal antibody, has emerged as a less invasive treatment option, showing potential for tumor volume reduction and hearing preservation. This systematic review aims to assess the efficacy of bevacizumab in controlling tumor volume, preserving hearing, and identifying associated adverse events. Methods: A comprehensive systematic review was performed using PRISMA guidelines. PubMed and Cochrane Library databases were searched for studies evaluating the effects of bevacizumab on VS, focusing on key outcomes like tumor volume reduction, hearing preservation, and adverse events. Data extraction and quality assessment were independently conducted by two reviewers using the Newcastle-Ottawa Scale. Results: Nine studies involving 176 patients were included. Bevacizumab showed a partial tumor volume reduction (≥20%) in 40% of cases and disease stabilization in 50%, while 10% experienced tumor progression. Hearing outcomes revealed improvement in 36% of patients, stabilization in 46%, and deterioration in 18%. Severe adverse effects, including hypertension and thromboembolic events, occurred in 13% of patients, while 18% reported no side effects. Tumor regrowth was observed in some patients after treatment discontinuation, emphasizing the need for long-term monitoring. Conclusions: Bevacizumab demonstrates effectiveness in managing VS, particularly in NF2 patients, by reducing tumor size and preserving hearing in a substantial proportion of cases. However, the variability in patient response and the risk of adverse events underscore the need for individualized treatment approaches and further research, including randomized controlled trials, to optimize its clinical application. Full article
(This article belongs to the Section Otolaryngology)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram illustrating the systematic review process. Out of 225 identified articles, 39 met the inclusion criteria. After applying exclusion criteria, 9 articles were included in the final analysis.</p>
Full article ">
31 pages, 929 KiB  
Review
Different Therapeutic Approaches for Dry and Wet AMD
by Nicoletta Marchesi, Martina Capierri, Alessia Pascale and Annalisa Barbieri
Int. J. Mol. Sci. 2024, 25(23), 13053; https://doi.org/10.3390/ijms252313053 - 4 Dec 2024
Viewed by 493
Abstract
Age-related macular degeneration (AMD) is the most common cause of irreversible loss of central vision in elderly subjects, affecting men and women equally. It is a degenerative pathology that causes progressive damage to the macula, the central and most vital part of the [...] Read more.
Age-related macular degeneration (AMD) is the most common cause of irreversible loss of central vision in elderly subjects, affecting men and women equally. It is a degenerative pathology that causes progressive damage to the macula, the central and most vital part of the retina. There are two forms of AMD depending on how the macula is damaged, dry AMD and wet or neovascular AMD. Dry AMD is the most common form; waste materials accumulate under the retina as old cells die, not being replaced. Wet AMD is less common, but can lead to vision loss much more quickly. Wet AMD is characterized by new abnormal blood vessels developing under the macula, where they do not normally grow. This frequently occurs in patients who already have dry AMD, as new blood vessels are developed to try to solve the problem. It is not known what causes AMD to develop; however, certain risk factors (i.e., age, smoking, genetic factors) can increase the risk of developing AMD. There are currently no treatments for dry AMD. There is evidence that not smoking, exercising regularly, eating nutritious food, and taking certain supplements can reduce the risk of acquiring AMD or slow its development. The main treatment for wet AMD is inhibitors of VEGF (vascular endothelial growth factor), a protein that stimulates the growth of new blood vessels. VEGF inhibitors can stop the growth of new blood vessels, preventing further damage to the macula and vision loss. In most patients, VEGF inhibitors can improve vision if macular degeneration is diagnosed early and treated accordingly. However, VEGF inhibitors cannot repair damage that has already occurred. Current AMD research is trying to find treatments for dry AMD and other options for wet AMD. This review provides a summary of the current evidence regarding the different treatments aimed at both forms of AMD with particular and greater attention to the dry form. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular and Cellular Biology 2024)
Show Figures

Figure 1

Figure 1
<p>Main AMD treatments. The purple section illustrates the treatments for dry AMD, whereas the red section depicts the treatments for wet AMD. The treatments that are not related to either form are summarized in the central area in blue. This figure has been created by the authors using Canva and Smart—Servier Medical Art.</p>
Full article ">
15 pages, 1362 KiB  
Article
MiRNA-34a, miRNA-145, and miRNA-222 Expression, Matrix Metalloproteinases, TNF-α and VEGF in Patients with Different Phenotypes of Coronary Artery Disease
by Alfiya Oskarovna Iusupova, Nikolay Nikolaevich Pakhtusov, Olga Alexandrovna Slepova, Natalia Vladimirovna Khabarova, Elena Vitalievna Privalova, Irina Vladimirovna Bure, Marina Vyacheslavovna Nemtsova and Yuri Nikitich Belenkov
Int. J. Mol. Sci. 2024, 25(23), 12978; https://doi.org/10.3390/ijms252312978 - 3 Dec 2024
Viewed by 326
Abstract
The development of different phenotypes of coronary artery (CA) lesions is regulated via many various factors, such as pro-inflammatory agents, zinc-dependent endopeptidases, growth factors and circulating microRNAs (miRs). To evaluate the expression levels of miR-34a, miR-145 and miR-222, tumor necrosis factor α (TNF-α), [...] Read more.
The development of different phenotypes of coronary artery (CA) lesions is regulated via many various factors, such as pro-inflammatory agents, zinc-dependent endopeptidases, growth factors and circulating microRNAs (miRs). To evaluate the expression levels of miR-34a, miR-145 and miR-222, tumor necrosis factor α (TNF-α), matrix metalloproteinases (MMP-1, -9, -13 and -14) and vascular endothelial growth factor (VEGF) in patients with different phenotypes of coronary artery disease (CAD): ischemia/angina with non-obstructive coronary arteries (INOCA/ANOCA) and obstructive CAD (oCAD) compared with a control group. This cross-sectional observational study included 157 subjects with a verified CAD diagnosis (51 patients with INOCA, 76 patients with oCAD and 30 healthy volunteers). The expression of miR-34a, miR-145 and miR-222 (RT-PCR) and the levels of VEGF, TNF-α, MMP-1, MMP-9, MMP-13 and MMP-14 (ELISA) were estimated in plasma samples. A higher concentration of MMP-9 was found in oCAD-group samples compared to the INOCA/ANOCA group. The INOCA/ANOCA group was characterized by higher levels of TNF-α. Based on multivariate regression analysis, a mathematical model predicting the type of CA lesion was constructed. MiR-145 was the independent predictor of INOCA/ANOCA (p = 0.006). Changes in concentrations of MMP-9 and MMP-14 were found in both investigated CAD groups, with MMP-9 levels being significantly higher in obstructive CAD samples than in INOCA/ANOCA, which confirms the role of inflammation in the development of atherosclerosis. A multivariate regression analysis allowed us to achieve a model that can predict the phenotype of stable CAD, and MiR-145 can be assumed as an independent predictor of INOCA/ANOCA. Full article
Show Figures

Figure 1

Figure 1
<p>The design of the study. CAD—coronary artery disease; INOCA/ANOCA—ischemia/angina with non-obstructive coronary arteries; qPCR—quantitative polymerase chain reaction; MRI—magnetic resonance imaging; MSCT—multi-detector computed tomography.</p>
Full article ">Figure 2
<p>MiRNAs expression in plasma of CAD patients and healthy volunteers (control). All values are presented as the median and CI. Statistically significant—<span class="html-italic">p</span> &lt; 0.05; CAD—coronary artery disease, INOCA/ANOCA—ischemia/angina with non-obstructive coronary arteries.</p>
Full article ">Figure 3
<p>Diagnostic value analysis of the model by ROC curve.</p>
Full article ">
9 pages, 229 KiB  
Article
Real-World Outcomes of Different Types of Treatment for Diabetic Macular Edema Before and After Approval of Anti-Vascular Endothelium Growth Factor Agents
by Masahiko Sugimoto, Shinichiro Chujo, Kumiko Kato, Masahiko Shimura, Shigehiko Kitano, Sentaro Kusuhara, Hiroto Terasaki, Mineo Kondo and Writing Committee of Japan-Clinical Retina Study Group (J-CREST)
J. Clin. Med. 2024, 13(23), 7336; https://doi.org/10.3390/jcm13237336 - 2 Dec 2024
Viewed by 392
Abstract
Background/Objectives: The object of this study was to determine the outcomes of treatments other than anti-vascular endothelial growth factor (VEGF) therapy for diabetic macular edema (DME) before and after the approval of anti-VEGF therapy in Japan. Methods: This was a retrospective [...] Read more.
Background/Objectives: The object of this study was to determine the outcomes of treatments other than anti-vascular endothelial growth factor (VEGF) therapy for diabetic macular edema (DME) before and after the approval of anti-VEGF therapy in Japan. Methods: This was a retrospective study registered in the database of the Survey of Treatment for DME (STREAT-DME). A total of 1683 patients treated from 2010 to 2017 were included. The patients were divided into two groups: (1) a pre-group, treated before the approval of anti-VEGF agents (2010–2013, n = 771), and (2) a post-group (2014–2017, n = 912). Each group was further categorized based on best-corrected visual acuity (BCVA): (i) improved from poor (>0.3 logMAR units) to good (≤0.3 logMAR units) or (ii) decreased from good to poor. Results: In the pre-group, 18.5% of patients improved from poor to good BCVA out of the total patient population (p < 0.0001), along with 17.3% out of those administered anti-VEGF therapy (p = 0.139), 20.5% of those administered a sub-tenon injection of triamcinolone acetonide (STTA, p = 0.02), 17.7% (p = 0.20) of those administered photocoagulation, and 14.2% of those who underwent pars plana vitrectomy (PPV, p = 0.0001). In the post-group, 21.8% had improved BCVA out of the total patient population (p < 0.0001), along with 27.2% of those undergoing anti-VEGF therapy (p < 0.0001), 16.7% of those administered STTA (p < 0.0001), and 27.2% of those who underwent PPV (p < 0.0001). Conclusions: STTA and PPV are effective to a certain extent, even after the approval of anti-VEGF agents. Full article
(This article belongs to the Special Issue An Update on Retinal Diseases: From Diagnosis to Treatment)
16 pages, 4204 KiB  
Article
Nebivolol Exerts Hepatoprotective Activity During CLP-Induced Sepsis by Modulating Oxidative Stress, Liver Regeneration, and AKT/MAPK Pathways in Rats
by Rahma Tharwat Sabra, Amany Abdlrehim Bekhit, Nourhan Tharwat Sabra, Nadia Ahmed Abd El-Moeze and Moustafa Fathy
Stresses 2024, 4(4), 800-815; https://doi.org/10.3390/stresses4040053 - 2 Dec 2024
Viewed by 375
Abstract
Sepsis is a potentially catastrophic organ dysfunction arising from an infection-induced immunologic reaction leading to severe inflammation, progression of septic shock, and damage to body organs. Sepsis is marked by noticeable hepatotoxicity caused by activating oxidative stress, inflammation, and apoptotic mechanisms. Through Cecal [...] Read more.
Sepsis is a potentially catastrophic organ dysfunction arising from an infection-induced immunologic reaction leading to severe inflammation, progression of septic shock, and damage to body organs. Sepsis is marked by noticeable hepatotoxicity caused by activating oxidative stress, inflammation, and apoptotic mechanisms. Through Cecal Ligation and Puncture (CLP) in rats, our study is the first to investigate the potential preventive effect of the antihypertensive medicine “Nebivolol” on sepsis-induced hepatotoxicity at a molecular level. Six groups of sixty albino Wistar rats (male) were randomly assigned. Biochemical and oxidative stress markers of liver function were measured. Additionally, apoptosis- and inflammatory-related gene and protein expressions were examined. Finally, the liver tissues were examined for histological assessments. The hepatic architecture was considerably altered by CLP, which also resulted in marked elevations of blood aspartate aminotransferase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP), total and direct bilirubin levels, and hepatic malondialdehyde (MDA). In contrast, it decreased serum albumin level, hepatic superoxide dismutase (SOD) activity, and glutathione (GSH) level. It also significantly elevated all hepatic inflammatory mediators (Interlukin-6 (IL-6), Tumor Necrosis Factor-alpha (TNF-α), and Interlukin-1 beta (IL-1β)) and alleviated Interlukin-10 (IL-10). It magnified the expression of p-AKT/t-AKT, p-JNK1/2/t-JNK1/2, and p-p38/t-p38 proteins, raised Matrix Metalloproteinase 2/9 (MMP 2/9) and nuclear factor-kappa B (NF-κB) gene transcriptions, and lessened Vascular Endothelial Growth Factor (VEGF) gene expression. In contrast, Nebivolol administration dramatically mitigated all biochemical and histological changes obtained by CLP. The present finding demonstrated that Nebivolol succeeded, for the first time, in improving the hepatic injury obtained from CLP-evoked sepsis through modulating oxidative stress, inflammatory mediators, and apoptotic pathways through targeting the crosstalk between protein kinase B (AKT), NF-κB, and mitogen-activated protein kinase (MAPK), making Nebivolol a hopeful treatment for hepatic injury. Full article
(This article belongs to the Collection Feature Papers in Human and Animal Stresses)
Show Figures

Figure 1

Figure 1
<p>Impact of Nebivolol on hepatic biochemical indicators in serum, (<b>a</b>) ALT, (<b>b</b>) AST, (<b>c</b>) ALP, (<b>d</b>) albumin, (<b>e</b>) direct bilirubin, and (<b>f</b>) total bilirubin after induction of CLP in rats. Data are exemplified as mean ± SEM. Following a one-way ANOVA test, the significant divisions among groups were analyzed using the Tukey–Kramar test, where ### <span class="html-italic">p</span>&lt; 0.001, contrasted with controlling sham control; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, contrasted with CLP group.</p>
Full article ">Figure 2
<p>Influence of Nebivolol on oxidative status of liver tissue. (<b>a</b>) MDA content, (<b>b</b>) SOD activity, and (<b>c</b>) GSH level. Data are exemplified as mean ± SEM. Following a one-way ANOVA test, the significant divisions among groups were analyzed using the Tukey–Kramar test, where ### <span class="html-italic">p</span> &lt; 0.001, contrasted with sham control; *** <span class="html-italic">p</span> &lt; 0.001, contrasted with CLP group; <span>$</span> <span class="html-italic">p</span> &lt; 0.05 and <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, contrasted with CLP + Neb4 group.</p>
Full article ">Figure 3
<p>Impact of Nebivolol on (<b>a</b>) <span class="html-italic">TNF-α</span>, (<b>b</b>) IL<span class="html-italic">-β</span>, (<b>c</b>) <span class="html-italic">IL-</span>6, and (<b>d</b>) <span class="html-italic">IL-</span>10 cytokines in hepatic tissues. Data are exemplified as mean ± SEM. Following a one-way ANOVA test, the significant divisions among groups were analyzed using the Tukey–Kramar test, where ### <span class="html-italic">p</span>&lt; 0.001, contrasted with sham control; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, contrasted with CLP group; <span>$</span> <span class="html-italic">p</span> &lt; 0.05, <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, and <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001, contrasted with CLP + Neb4 group.</p>
Full article ">Figure 4
<p>Effect of Nebivolol on (<b>a</b>) <span class="html-italic">MMP-2</span>, (<b>b</b>) <span class="html-italic">MMP-9</span>, (<b>c</b>) <span class="html-italic">VEGF</span>, and (<b>d</b>) <span class="html-italic">NF-κB</span> mRNA expression levels in liver samples. Data are exemplified as mean ± SEM. Following a one-way ANOVA test, the significant divisions among groups were analyzed using the Tukey–Kramar test, where ### <span class="html-italic">p</span> &lt; 0.001, contrasted with sham control; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001, contrasted with CLP group; <span>$</span> <span class="html-italic">p</span> &lt; 0.05 and <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, contrasted with CLP + Neb4 group.</p>
Full article ">Figure 5
<p>Effect of Nebivolol on p-AKT, p-JNK1/2, and p-p38 protein expressions. (<b>a</b>) Representative western blot membranes of t-AKT, p-AKT, t-JNK1/2, p-JNK1/2, t-p38, p-p38, and b-actin proteins for all studied groups; (<b>b</b>–<b>d</b>) expressions of p-AKT/t-AKT, p-JNK1/2/t-JNK1/2, and p-p38/t-p38 proteins were expressed densitometrically using bands in (<b>a</b>). Data are exemplified as mean ± SEM. Following a one-way ANOVA test, the significant divisions among groups were analyzed using the Tukey–Kramar test, where ### <span class="html-italic">p</span>&lt; 0.001, contrasted with sham control; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, contrasted with CLP group; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, contrasted with CLP + Neb4 group.</p>
Full article ">Figure 6
<p>Impact of Nebivolol on liver histological alterations. The figure demonstrates photographs of rat liver (H&amp;E staining, 400×). (<b>a</b>) Sham specimens depict a standard hepatic architecture, with hepatocytes organized in cords extending from the central vein (black arrow) and portal tract (blue arrow). (<b>b</b>) Nebivolol 10 group with the same structure as a sham group. (<b>c1</b>) CLP group showing hepatocyte necrosis, ballooning, and inflammatory cell infiltration (in the circle) with a higher magnification photomicrograph in (<b>c2</b>) (600×) showing binuclear cells (red arrow) with area of necrosis (brown arrow). (<b>d</b>) CLP/Nebivolol 4 showing decreased the inflammatory cell infiltration, ballooning deterioration, and hepatocyte necrosis (in the circle). (<b>e</b>) CLP/Nebivolol 10 improves the hepatic integrity with minimal inflammatory cells, slight centrilobular hepatocyte deterioration and ballooning with no necrosis. (<b>f</b>) CLP/vitamin C showing mild portal tract inflammation.</p>
Full article ">Figure 7
<p>Schematic illustration of the research design.</p>
Full article ">
21 pages, 2029 KiB  
Systematic Review
Addition of Bevacizumab to Chemotherapy and Its Impact on Clinical Efficacy in Cervical Cancer: A Systematic Review and Meta-Analysis
by Aleena Shahzad, Anees ur Rehman, Tehnia Naz, Muhammad Fawad Rasool, Alisha Saeed, Saba Rasheed, Sadia Shakeel, Saleh Karamah Al-Tamimi and Rabia Hussain
Pharmacy 2024, 12(6), 180; https://doi.org/10.3390/pharmacy12060180 - 1 Dec 2024
Viewed by 992
Abstract
Background and Objectives: Cervical cancer is the third leading cause of cancer-related mortality in females. One of the most successful therapeutic modalities to date is suppressing vascular endothelial growth factor (VEGF)-mediated angiogenesis. Bevacizumab is a monoclonal antibody that targets VEGF-A. The outcomes [...] Read more.
Background and Objectives: Cervical cancer is the third leading cause of cancer-related mortality in females. One of the most successful therapeutic modalities to date is suppressing vascular endothelial growth factor (VEGF)-mediated angiogenesis. Bevacizumab is a monoclonal antibody that targets VEGF-A. The outcomes for cervical cancer patients treated with bevacizumab in combination with platinum-based chemotherapy have been explored in several studies. This study aimed to assess the impact of bevacizumab on progression-free survival (PFS) and overall survival (OS) in patients with metastatic cervical cancer. Materials and Methods: This systematic review was registered in PROSPERO (CRD42023456755). Following PRISMA guidelines, a comprehensive literature search on PubMed and Google Scholar identified 28 studies meeting the inclusion criteria. The outcomes of interest were PFS and OS. The statistical analysis computed hazard ratios (HRs) with 95% confidence intervals (CIs). The study also included a subgroup analysis by cervical cancer stage. Results: The pooled analysis revealed that bevacizumab-based therapy significantly improved both PFS with HR 0.77 (95% CI: 0.58–0.96; p < 0.01; I2 = 58%) and OS with HR 0.63 (95% CI: 0.45–0.89; p < 0.01; I2 = 41%) in cervical cancer patients. Subgroup analysis by stage of cervical cancer demonstrated better efficacy of bevacizumab in metastatic stage IVB cervical cancer patients indicated by HR for PFS (0.69, 95% CI: 0.54–0.79; p < 0.01) and HR for OS (0.57, 95% CI: 0.46–0.73; p < 0.01). Conclusions: Bevacizumab exhibits a significant increase in PFS and OS, underscoring the efficacy of anti-angiogenesis therapy in cervical cancer, particularly in stage IVB metastatic cervical cancer patients. Full article
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram of the study selection process.</p>
Full article ">Figure 2
<p>Forest plot showing efficacy of bevacizumab on PFS in cervical cancer patients. The meta-analysis results are illustrated as pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for PFS across the included studies. Each square represents an individual study’s effect size [<a href="#B1-pharmacy-12-00180" class="html-bibr">1</a>,<a href="#B2-pharmacy-12-00180" class="html-bibr">2</a>,<a href="#B11-pharmacy-12-00180" class="html-bibr">11</a>,<a href="#B12-pharmacy-12-00180" class="html-bibr">12</a>,<a href="#B17-pharmacy-12-00180" class="html-bibr">17</a>,<a href="#B18-pharmacy-12-00180" class="html-bibr">18</a>,<a href="#B45-pharmacy-12-00180" class="html-bibr">45</a>,<a href="#B47-pharmacy-12-00180" class="html-bibr">47</a>,<a href="#B49-pharmacy-12-00180" class="html-bibr">49</a>,<a href="#B51-pharmacy-12-00180" class="html-bibr">51</a>,<a href="#B52-pharmacy-12-00180" class="html-bibr">52</a>,<a href="#B53-pharmacy-12-00180" class="html-bibr">53</a>,<a href="#B55-pharmacy-12-00180" class="html-bibr">55</a>,<a href="#B56-pharmacy-12-00180" class="html-bibr">56</a>]. The horizontal lines show 95% CIs, and the diamond represents the overall pooled estimate (0.77 [0.58–0.96]). The I<sup>2</sup> statistic (58%) indicates heterogeneity across the studies.</p>
Full article ">Figure 3
<p>Forest plot showing efficacy of bevacizumab on PFS in metastatic (stage IVB) cervical cancer patients. The meta-analysis results are illustrated as pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for PFS in stage IVB patients across the included studies. Each square represents an individual study’s effect size [<a href="#B2-pharmacy-12-00180" class="html-bibr">2</a>,<a href="#B5-pharmacy-12-00180" class="html-bibr">5</a>,<a href="#B11-pharmacy-12-00180" class="html-bibr">11</a>,<a href="#B18-pharmacy-12-00180" class="html-bibr">18</a>,<a href="#B43-pharmacy-12-00180" class="html-bibr">43</a>,<a href="#B47-pharmacy-12-00180" class="html-bibr">47</a>,<a href="#B49-pharmacy-12-00180" class="html-bibr">49</a>,<a href="#B53-pharmacy-12-00180" class="html-bibr">53</a>,<a href="#B55-pharmacy-12-00180" class="html-bibr">55</a>,<a href="#B56-pharmacy-12-00180" class="html-bibr">56</a>]. The horizontal lines show 95% CIs, and the diamond represents the overall pooled estimate (0.69 [0.54–0.79]). The I<sup>2</sup> statistic (48.51%) indicates heterogeneity across the studies.</p>
Full article ">Figure 4
<p>Forest plot showing efficacy of bevacizumab on OS in cervical cancer patients. The meta-analysis results are illustrated as pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for OS patients across the included studies. Each square represents an individual study’s effect size [<a href="#B1-pharmacy-12-00180" class="html-bibr">1</a>,<a href="#B2-pharmacy-12-00180" class="html-bibr">2</a>,<a href="#B5-pharmacy-12-00180" class="html-bibr">5</a>,<a href="#B12-pharmacy-12-00180" class="html-bibr">12</a>,<a href="#B14-pharmacy-12-00180" class="html-bibr">14</a>,<a href="#B44-pharmacy-12-00180" class="html-bibr">44</a>,<a href="#B45-pharmacy-12-00180" class="html-bibr">45</a>,<a href="#B48-pharmacy-12-00180" class="html-bibr">48</a>,<a href="#B49-pharmacy-12-00180" class="html-bibr">49</a>,<a href="#B52-pharmacy-12-00180" class="html-bibr">52</a>,<a href="#B53-pharmacy-12-00180" class="html-bibr">53</a>,<a href="#B57-pharmacy-12-00180" class="html-bibr">57</a>]. The horizontal lines show 95% CIs, and the diamond represents the overall pooled estimate (0.63 [0.45–0.89]). The I<sup>2</sup> statistic (41%) indicates heterogeneity across the studies.</p>
Full article ">Figure 5
<p>Forest plot showing efficacy of bevacizumab on OS in metastatic (stage IVB) cervical cancer patients. The meta-analysis results are illustrated as pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for OS in stage IVB patients across the included studies. Each square represents an individual study’s effect size [<a href="#B2-pharmacy-12-00180" class="html-bibr">2</a>,<a href="#B5-pharmacy-12-00180" class="html-bibr">5</a>,<a href="#B14-pharmacy-12-00180" class="html-bibr">14</a>,<a href="#B48-pharmacy-12-00180" class="html-bibr">48</a>,<a href="#B49-pharmacy-12-00180" class="html-bibr">49</a>,<a href="#B53-pharmacy-12-00180" class="html-bibr">53</a>,<a href="#B57-pharmacy-12-00180" class="html-bibr">57</a>]. The horizontal lines show 95% CIs, and the diamond represents the overall pooled estimate (0.57 [0.46–0.73]). The I<sup>2</sup> statistic (47.21%) indicates heterogeneity across the studies.</p>
Full article ">
Back to TopTop