[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
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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (8,563)

Search Parameters:
Keywords = microRNAs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1645 KiB  
Article
Cross-Sectional Analysis of Hypoxia-Regulated miRNA-181a, miRNA-199a, HIF-1α, and SIRT1 in the Development of Type 2 Diabetes in Patients with Obstructive Sleep Apnea—Preliminary Study
by Filip Franciszek Karuga, Piotr Kaczmarski, Marcin Sochal, Bartosz Szmyd, Piotr Białasiewicz and Agata Gabryelska
J. Clin. Med. 2024, 13(24), 7644; https://doi.org/10.3390/jcm13247644 (registering DOI) - 15 Dec 2024
Abstract
Introduction: Obstructive sleep apnea (OSA) is recognized as an independent risk factor for diabetes mellitus type 2 (T2DM) development, which is twice as common in patients with OSA compared to non-OSA patients. Objectives: This study aimed to investigate changes in oxygen metabolism [...] Read more.
Introduction: Obstructive sleep apnea (OSA) is recognized as an independent risk factor for diabetes mellitus type 2 (T2DM) development, which is twice as common in patients with OSA compared to non-OSA patients. Objectives: This study aimed to investigate changes in oxygen metabolism and their role in T2DM development among OSA patients through epigenetic processes via miRNA-181a, miRNA-199a, and enzymatic processes via SIRT1 and HIF-1α. Methods: Based on polysomnography, apnea–hypopnea index and the presence of T2DM patients were divided into three groups: control group (n = 17), OSA group (n = 11), OSA&T2DM (n = 20) group. Total RNA was extracted from the buffy coat. Moreover, HOMA-IR (Homeostatic Model Assessment for Insulin Resistance) was counted. Results: Morning miRNA-181a expression was significantly higher in the OSA&T2DM group than in the control group: 67.618 vs. 32.685 (p = 0.036). Evening miRNA-199a expression was significantly higher in the OSA group than in the control group: 5.043 vs. 2.081 (p = 0.042), while its morning expression was significantly higher in the OSA&T2DM group when compared to the control: 4.065 vs. 1.605 (p = 0.036). MiRNA-181a evening expression revealed a negative correlation with the SIRT1 evening and morning expressions (R = −0.367, p = 0.010 and R = −0.405, p = 0.004, respectively). Moreover, morning miRNA-181a was positively correlated with HOMA-IR (R = 0.321, p = 0.034). MiRNA-199a evening expression presented a moderate positive correlation with the SIRT1 morning expressions (R = 0.48, p < 0.001) and HOMA-IR (R = 0.35, p = 0.02). Conclusions: Patients suffering from OSA and T2DM had an increased expression of miRNA-181a. Moreover, a negative correlation between miRNA-181a and SIRT1 expression was observed, while a correlation between miRNA-181a and insulin resistance was positive. This phenomenon might suggest a possible epigenetic pathway for an increased incidence of T2DM in OSA patients however further research is needed. Full article
21 pages, 1069 KiB  
Review
Extracellular Vesicles from Mesenchymal Stem Cells: Potential as Therapeutics in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD)
by Xue Zou and David Brigstock
Biomedicines 2024, 12(12), 2848; https://doi.org/10.3390/biomedicines12122848 (registering DOI) - 14 Dec 2024
Viewed by 297
Abstract
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by the accumulation of triglycerides within hepatocytes, which can progress to more severe conditions, such as metabolic dysfunction-associated steatohepatitis (MASH), which may include progressive fibrosis, leading to cirrhosis, cancer, and death. This goal of [...] Read more.
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by the accumulation of triglycerides within hepatocytes, which can progress to more severe conditions, such as metabolic dysfunction-associated steatohepatitis (MASH), which may include progressive fibrosis, leading to cirrhosis, cancer, and death. This goal of this review is to highlight recent research showing the potential of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in reducing the key pathogenic pathways of MASLD or MASH. Methods: Relevant published studies were identified using PubMed with one or more of the following search terms: MASLD, MASH, NAFLD, NASH, exosome, extracellular vesicle (EV), therapy, and/or mesenchymal stem cells (MSC). The primary literature were subsequently downloaded and summarized. Results: Using in vitro or in vivo models, MSC-EVs have been found to counteract oxidative stress, a significant contributor to liver injury in MASH, and to suppress disease progression, including steatosis, inflammation, and, in a few instances, fibrosis. Some of these outcomes have been attributed to specific EV cargo components including microRNAs and proteins. Thus, MSC-EVs enriched with these types of molecules may have improved the therapeutic efficacy for MASLD/MASH and represent a novel approach to potentially halt or reverse the disease process. Conclusions: MSC-EVs are attractive therapeutic agents for treating MASLD/MASH. Further studies are necessary to validate the clinical applicability and efficacy of MSC-EVs in human MASH patients, focusing on optimizing delivery strategies and identifying the pathogenic pathways that are targeted by specific EV components. Full article
30 pages, 883 KiB  
Review
Approaches and Challenges in Characterizing the Molecular Content of Extracellular Vesicles for Biomarker Discovery
by Suman Kumari, Christopher Lausted, Kelsey Scherler, Alphonsus H. C. Ng, Yue Lu, Inyoul Lee, Leroy Hood and Kai Wang
Biomolecules 2024, 14(12), 1599; https://doi.org/10.3390/biom14121599 (registering DOI) - 14 Dec 2024
Viewed by 512
Abstract
Extracellular vesicles (EVs) are lipid bilayer nanoparticles released from all known cells and are involved in cell-to-cell communication via their molecular content. EVs have been found in all tissues and body fluids, carrying a variety of biomolecules, including DNA, RNA, proteins, metabolites, and [...] Read more.
Extracellular vesicles (EVs) are lipid bilayer nanoparticles released from all known cells and are involved in cell-to-cell communication via their molecular content. EVs have been found in all tissues and body fluids, carrying a variety of biomolecules, including DNA, RNA, proteins, metabolites, and lipids, offering insights into cellular and pathophysiological conditions. Despite the emergence of EVs and their molecular contents as important biological indicators, it remains difficult to explore EV-mediated biological processes due to their small size and heterogeneity and the technical challenges in characterizing their molecular content. EV-associated small RNAs, especially microRNAs, have been extensively studied. However, other less characterized RNAs, including protein-coding mRNAs, long noncoding RNAs, circular RNAs, and tRNAs, have also been found in EVs. Furthermore, the EV-associated proteins can be used to distinguish different types of EVs. The spectrum of EV-associated RNAs, as well as proteins, may be associated with different pathophysiological conditions. Therefore, the ability to comprehensively characterize EVs’ molecular content is critical for understanding their biological function and potential applications in disease diagnosis. Here, we set out to provide an overview of EV-associated RNAs and proteins as well as approaches currently being used to characterize them. Full article
(This article belongs to the Special Issue Extracellular Vesicles as Biomarkers of Diseases)
Show Figures

Figure 1

Figure 1
<p>Depiction of EV subtypes and their biogenesis. Exosomes, a type of small EVs, are formed from the invagination of the endosomal membrane. Small ectosomes, another type of sEVs, are generated by the outward budding of the membrane. Microvesicles, indicated as large EVs, are produced through the direct budding of the plasma membrane. Apoptotic vesicles, a type of lEV, are produced by blebbing and fragmentation during apoptosis.</p>
Full article ">
16 pages, 1611 KiB  
Article
Multiple Myeloma: Genetic and Epigenetic Biomarkers with Clinical Potential
by Yuliya A. Veryaskina, Sergei E. Titov, Natalia V. Skvortsova, Igor B. Kovynev, Oksana V. Antonenko, Sergei A. Demakov, Pavel S. Demenkov, Tatiana I. Pospelova, Mikhail K. Ivanov and Igor F. Zhimulev
Int. J. Mol. Sci. 2024, 25(24), 13404; https://doi.org/10.3390/ijms252413404 (registering DOI) - 13 Dec 2024
Viewed by 243
Abstract
Multiple myeloma (MM) is characterized by the uncontrolled proliferation of monoclonal plasma cells and accounts for approximately 10% of all hematologic malignancies. The clinical outcomes of MM can exhibit considerable variability. Variability in both the genetic and epigenetic characteristics of MM undeniably contributes [...] Read more.
Multiple myeloma (MM) is characterized by the uncontrolled proliferation of monoclonal plasma cells and accounts for approximately 10% of all hematologic malignancies. The clinical outcomes of MM can exhibit considerable variability. Variability in both the genetic and epigenetic characteristics of MM undeniably contributes to tumor dynamics. The aim of the present study was to identify biomarkers with the potential to improve the accuracy of prognosis assessment in MM. Initially, miRNA sequencing was conducted on bone marrow (BM) samples from patients with MM. Subsequently, the expression levels of 27 microRNAs (miRNA) and the gene expression levels of ASF1B, CD82B, CRISP3, FN1, MEF2B, PD-L1, PPARγ, TERT, TIMP1, TOP2A, and TP53 were evaluated via real-time reverse transcription polymerase chain reaction in BM samples from patients with MM exhibiting favorable and unfavorable prognoses. Additionally, the analysis involved the bone marrow samples from patients undergoing examinations for non-cancerous blood diseases (NCBD). The findings indicate a statistically significant increase in the expression levels of miRNA-124, -138, -10a, -126, -143, -146b, -20a, -21, -29b, and let-7a and a decrease in the expression level of miRNA-96 in the MM group compared with NCBD (p < 0.05). No statistically significant differences were detected in the expression levels of the selected miRNAs between the unfavorable and favorable prognoses in MM groups. The expression levels of ASF1B, CD82B, and CRISP3 were significantly decreased, while those of FN1, MEF2B, PDL1, PPARγ, and TERT were significantly increased in the MM group compared to the NCBD group (p < 0.05). The MM group with a favorable prognosis demonstrated a statistically significant decline in TIMP1 expression and a significant increase in CD82B and CRISP3 expression compared to the MM group with an unfavorable prognosis (p < 0.05). From an empirical point of view, we have established that the complex biomarker encompassing the CRISP3/TIMP1 expression ratio holds promise as a prognostic marker in MM. From a fundamental point of view, we have demonstrated that the development of MM is rooted in a cascade of complex molecular pathways, demonstrating the interplay of genetic and epigenetic factors. Full article
(This article belongs to the Special Issue Molecular Mechanisms of mRNA Transcriptional Regulation: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Hierarchical cluster analysis between 16 multiple myeloma (MM) cases and six non-cancerous blood diseases (NCBD) cases for the microRNAs (miRNAs) that were chosen for validation by RT-PCR in the analyzed groups. Each column represents the expression of a miRNA, and each row denotes a nucleic acid sample. Yellow: upregulated miRNA; blue: downregulated miRNA; green: minor changes; red: a graphical representation of a group of samples.</p>
Full article ">Figure 2
<p>Comparative analysis of gene expression levels between multiple myeloma (MM) (n = 45) and non-cancerous samples (NCBD) (n = 43). The figure presents the median value, upper and lower quartiles, non-outlier range, and outliers appearing as circles.</p>
Full article ">Figure 3
<p>Comparative analysis of gene expression levels between multiple myeloma samples of patients with favorable (n = 28) and unfavorable (n = 17) prognosis. The figure presents the median value, upper and lower quartiles, non-outlier range, and outliers appearing as circles.</p>
Full article ">Figure 4
<p>ROC analysis for the (<b>A</b>) <span class="html-italic">CRISP3</span>, (<b>B</b>) <span class="html-italic">TIMP1</span>, and (<b>C</b>) <span class="html-italic">CRISP3</span>/<span class="html-italic">TIMP1</span> genes. AUC, sensitivity (Sn), and specificity (Sp) values are indicated. Red line is a diagonal support line, blue is a ROC curve.</p>
Full article ">Figure 4 Cont.
<p>ROC analysis for the (<b>A</b>) <span class="html-italic">CRISP3</span>, (<b>B</b>) <span class="html-italic">TIMP1</span>, and (<b>C</b>) <span class="html-italic">CRISP3</span>/<span class="html-italic">TIMP1</span> genes. AUC, sensitivity (Sn), and specificity (Sp) values are indicated. Red line is a diagonal support line, blue is a ROC curve.</p>
Full article ">Figure 5
<p>Interactions between microRNAs(miRNAs) and their target genes. Blue squares represent miRNAs, and purple circles indicate their target genes.</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 235
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 ">
15 pages, 2532 KiB  
Review
MicroRNA and Heart Failure: A Novel Promising Diagnostic and Therapeutic Tool
by Andrea D’Amato, Silvia Prosperi, Paolo Severino, Vincenzo Myftari, Michele Correale, Pasquale Perrone Filardi, Roberto Badagliacca, Francesco Fedele, Carmine Dario Vizza and Alberto Palazzuoli
J. Clin. Med. 2024, 13(24), 7560; https://doi.org/10.3390/jcm13247560 - 12 Dec 2024
Viewed by 271
Abstract
Heart failure (HF) has a multifaceted and complex pathophysiology. Beyond neurohormonal, renin–angiotensin–aldosterone system, and adrenergic hyperactivation, a role for other pathophysiological determinants is emerging. Genetic and epigenetic factors are involved in this syndrome. In many maladaptive processes, the role of microRNAs (miRNAs) has [...] Read more.
Heart failure (HF) has a multifaceted and complex pathophysiology. Beyond neurohormonal, renin–angiotensin–aldosterone system, and adrenergic hyperactivation, a role for other pathophysiological determinants is emerging. Genetic and epigenetic factors are involved in this syndrome. In many maladaptive processes, the role of microRNAs (miRNAs) has been recently demonstrated. MiRNAs are small endogenous non-coding molecules of RNA involved in gene expression regulation, and they play a pivotal role in intercellular communication, being involved in different biological and pathophysiological processes. MiRNAs can modulate infarct area size, cardiomyocytes restoration, collagen deposition, and macrophage polarization. MiRNAs may be considered as specific biomarkers of hypertrophy and fibrosis. MiRNAs have been proposed as a therapeutical tool because their administration can contrast with myocardial pathophysiological remodeling leading to HF. Antimir and miRNA mimics are small oligonucleotides which may be administered in several manners and may be able to regulate the expression of specific and circulating miRNAs. Studies on animal models and on healthy humans demonstrate that these molecules are well tolerated and effective, opening the possibility of a therapeutic use of miRNAs in cases of HF. The application of miRNAs for diagnosis, prognostic stratification, and therapy fits in with the new concept of a personalized and tailored approach to HF. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

Figure 1
<p><b>Molecular and pathophysiological mechanisms in which microRNAs (miRNAs) are involved in the myocardium.</b> MiRNAs are involved in several molecular processes in the myocardium, such as ion activity, and metabolic and inflammatory response regulation. MiRNAs contribute to heart failure through four main processes in the myocardium: inflammation, angiogenesis, myocardial hypertrophy, and fibrosis. MiRNAs: microRNAs.</p>
Full article ">Figure 2
<p><b>Main pathophysiological mechanisms on which microRNAs (miRNAs)-targeted therapy has been studied.</b> Myocardial fibrosis, hypertrophy, and ischemia are the main mechanisms leading to heart failure and, for this reason, they may represent crucial mechanisms to counteract. Many molecular pathways have been involved in each mechanism. MiRNAs: microRNAs; TGF-β: transforming growth factor- β; ERK-MAP: extracellular signal-regulated kinase/microtubule-associated protein kinase; IL-6/STAT3/PTEN: interleukin-6/signal transducer and activator of transcription 3/Phosphatase and tensin homolog; GPD2: Glycerol-3-Phosphate Dehydrogenase 2; Nrf2/ARE: nuclear erythroid 2-related factor 2.</p>
Full article ">
8 pages, 253 KiB  
Editorial
The 2024 Nobel Prize in Physiology or Medicine: microRNA Takes Center Stage
by George A. Calin, Florent Hubé, Michael R. Ladomery, Nicholas Delihas, Manuela Ferracin, Laura Poliseno, Luca Agnelli, Suresh K. Alahari, Ai-Ming Yu and Xiao-Bo Zhong
Non-Coding RNA 2024, 10(6), 62; https://doi.org/10.3390/ncrna10060062 - 12 Dec 2024
Viewed by 362
Abstract
The Non-coding Journal Editorial Board Members would like to congratulate Victor Ambros and Gary Ruvkun, who were jointly awarded the 2024 Nobel Prize in Physiology or Medicine for their groundbreaking discovery of microRNAs and the role of microRNAs in post-transcriptional gene regulation, uncovering [...] Read more.
The Non-coding Journal Editorial Board Members would like to congratulate Victor Ambros and Gary Ruvkun, who were jointly awarded the 2024 Nobel Prize in Physiology or Medicine for their groundbreaking discovery of microRNAs and the role of microRNAs in post-transcriptional gene regulation, uncovering a previously unknown layer of gene control in eukaryotes [...] Full article
16 pages, 7938 KiB  
Article
Identification and Potential Functions of Ebola Virus-Encoded MicroRNAs in EBOV-Infected Human ARPE Cells
by Melvin Mensah-Bonsu, Christopher Doss, Clay Gloster and Perpetua Muganda
Acta Microbiol. Hell. 2024, 69(4), 295-310; https://doi.org/10.3390/amh69040027 - 12 Dec 2024
Viewed by 400
Abstract
Ebola virus (EBOV) causes Ebola virus disease (EVD), a severe and often fatal hemorrhagic fever. Although much research has focused on host miRNA expression during EBOV infection, it has been discovered that EBOV itself also produces miRNAs. However, further studies are needed to [...] Read more.
Ebola virus (EBOV) causes Ebola virus disease (EVD), a severe and often fatal hemorrhagic fever. Although much research has focused on host miRNA expression during EBOV infection, it has been discovered that EBOV itself also produces miRNAs. However, further studies are needed to fully comprehend the role of these EBOV-encoded miRNAs in infection and disease development. This study aimed to identify known and novel EBOV-encoded miRNAs and their potential functions in the pathogenic mechanisms of EBOV. We reanalyzed previously available small RNASeq data to identify the miRNAs and predict their cellular targets and functions. We identified four EBOV-encoded miRNAs—EBOV-mir-M1 (4390–4414), EBOV-mir-M4, EBOV-mir-M2 (8288–8309), and EBOV-mir-M3 (9885–9906)—expressed specifically in Ebola-infected human adult retinal pigment epithelial (ARPE) cells. EBOV-mir-M1 (4390–4414) was expressed up to 19 times more than the other three miRNAs. The identified miRNAs were predicted to target genes associated with pathways such as calcium signaling, MAPK signaling, type I interferon signaling, and cytokine-mediated signaling, which play critical roles in Ebola infection and pathogenesis. This study contributes to our understanding of the role of EBOV-encoded miRNAs in infection and pathogenesis by demonstrating the expression of these miRNAs in human ARPE cells, providing insights into the mechanisms underlying EBOV pathogenesis. Full article
(This article belongs to the Special Issue Feature Papers in Medical Microbiology in 2024)
Show Figures

Figure 1

Figure 1
<p>Secondary (drawn using RNAFold webserver, FORNA view) and 3D structures (obtained using RNAComposer and PyMol) of EBOV-encoded precursor miRNAs with mature miRNAs (highlighted red) expressed in infected human RPE cells at 24 h post-infection. (<b>A</b>) EBOV-mir-M1 (4390–4414), (<b>B</b>) EBOV-mir-M2 (8288–8309), (<b>C</b>) EBOV-mir-M3 (9885–9906), (<b>D</b>) EBOV-mir-M4 (11,501–11,523). EBOV-encoded miRNAs found on the same precursor are shown with green markers.</p>
Full article ">Figure 2
<p>EBOV-encoded miRNAs expressed in infected human RPE cells at 24 h post-infection. Mock-infected and EBOV-infected datasets were analyzed for Ebola virus encoded miRNAs as described in the <a href="#sec2-amh-69-00027" class="html-sec">Section 2</a>. Normalized miRNA quantities in counts per million for fqgrep tool (<b>left two panels</b>) and IGV (<b>right two panels</b>) are plotted for each miRNA identified in infected cells (<b>two bottom panels</b> labelled infected) as compared to mock-infected cells (<b>two top panels</b> labelled control). * indicates significance at 5% level, ** indicates significance at 1% level, *** indicates significance at 1‰ level, **** indicates significance at 0.1‰ level.</p>
Full article ">Figure 3
<p>IGV visualization of EBOV-encoded miRNAs expressed in infected human RPE cells at 24 h post-infection. (<b>A</b>) EBOV-miR-M1 (4390–4414), EBOV-miR-M2 (8288–8309), EBOV-miR-M3 (9885–9906), and EBOV-miR-M4 (11,501–11,523) (<b>B</b>) Visualization of mock-infected (Blue control) vs. infected (Orange infected) samples showing the presence of an identified miRNA in the infected samples but not present in the mock-infected samples.</p>
Full article ">Figure 4
<p>(<b>A</b>) Heat map (datasets with same color bars represent replicates (blue, orange, green, red, black, grey) control/mock-infected and infected samples were made in triplicates and sequenced 4× to obtain 12 control/mock-infected and 12 infected samples). (<b>B</b>) Graph of all EBOV-encoded miRNAs expressed in infected human RPE cells at 24 h post-infection. (Left panel represents the study miRNAs and right panel represents previously reported miRNAs. * indicates significance at 5% level, ** indicates significance at 1% level, *** indicates significance at 1‰ level, **** indicates significance at 0.1‰ level). (<b>C</b>) EBOV-encoded miRNAs mapped to their coordinates on the EBOV genome. (Highlighted miRNAs represent miRNAs identified by this study) [<a href="#B29-amh-69-00027" class="html-bibr">29</a>,<a href="#B30-amh-69-00027" class="html-bibr">30</a>,<a href="#B31-amh-69-00027" class="html-bibr">31</a>,<a href="#B33-amh-69-00027" class="html-bibr">33</a>].</p>
Full article ">Figure 5
<p>(<b>A</b>) EBOV-encoded miRNAs identified in virus-infected human ARPE cells at 24 h post-infection. Sequences with similar regions in bold font and highlighted. Enriched KEGG pathways of EBOV-encoded miRNAs in RPE cells from DAVID analysis. (<b>B</b>) EBOV-mir-M1 (4390–4414), EBOV-mir-M2 (8288–8309), and EBOV-mir-M3 (9885–9906). (<b>C</b>) EBOV-mir-M4 (11,501–11,523).</p>
Full article ">
23 pages, 3021 KiB  
Article
Increased Levels of hsa-miR-199a-3p and hsa-miR-382-5p in Maternal and Neonatal Blood Plasma in the Case of Placenta Accreta Spectrum
by Angelika V. Timofeeva, Ivan S. Fedorov, Anastasia D. Nikonets, Alla M. Tarasova, Ekaterina N. Balashova, Dmitry N. Degtyarev and Gennady T. Sukhikh
Int. J. Mol. Sci. 2024, 25(24), 13309; https://doi.org/10.3390/ijms252413309 - 11 Dec 2024
Viewed by 358
Abstract
Despite the increasing number of placenta accreta spectrum (PAS) cases in recent years, its impact on neonatal outcomes and respiratory morbidity, as well as the underlying pathogenetic mechanism, has not yet been extensively studied. Moreover, no study has yet demonstrated the effectiveness of [...] Read more.
Despite the increasing number of placenta accreta spectrum (PAS) cases in recent years, its impact on neonatal outcomes and respiratory morbidity, as well as the underlying pathogenetic mechanism, has not yet been extensively studied. Moreover, no study has yet demonstrated the effectiveness of antenatal corticosteroid therapy (CT) for the prevention of respiratory distress syndrome (RDS) in newborns of mothers with PAS at the molecular level. In this regard, microRNA (miRNA) profiling by small RNA deep sequencing and quantitative real-time PCR was performed on 160 blood plasma samples from preterm infants (gestational age: 33–36 weeks) and their mothers who had been diagnosed with or without PAS depending on the timing of the antenatal RDS prophylaxis. A significant increase in hsa-miR-199a-3p and hsa-miR-382-5p levels was observed in the blood plasma of the newborns from mothers with PAS compared to the control group. A clear trend toward the normalization of hsa-miR-199a-3p and hsa-miR-382-5p levels in the neonatal blood plasma of the PAS groups was observed when CT was administered within 14 days before delivery, but not beyond 14 days. Direct correlations were found among the hsa-miR-382-5p level in neonatal blood plasma and the hsa-miR-199a-3p level in the same sample (r = 0.49; p < 0.001), the oxygen requirements in the NICU (r = 0.41; p = 0.001), the duration of the NICU stay (r = 0.31; p = 0.019), and the severity of the newborn’s condition based on the NEOMOD scale (r = 0.36; p = 0.005). Logistic regression models based on the maternal plasma levels of hsa-miR-199a-3p and hsa-miR-382-5p predicted the need for cardiotonic therapy, invasive mechanical ventilation, or high-frequency oscillatory ventilation in newborns during the early neonatal period, with a sensitivity of 95–100%. According to the literary data, these miRNAs regulate fetal organogenesis via IGF-1, the formation of proper lung tissue architecture, surfactant synthesis in alveolar cells, and vascular tone. Full article
(This article belongs to the Special Issue The Role of miRNA in Human Diseases)
Show Figures

Figure 1

Figure 1
<p>PLS-A analysis of deep sequencing data of miRNA in the peripheral blood plasma of day-old newborns from mothers with PAS and without PAS (control).</p>
Full article ">Figure 2
<p>The dependence of hsa-miR-382-5p and hsa-miR-199a-3p content in the blood plasma of newborns and their mothers on the severity of placenta accreta spectrum (PAS) and the timing of antenatal corticosteroid therapy (CT). Levels of miR-382-5p (−∆Ct, PCR data) in the blood plasma of newborns from mothers with placenta accreta or placenta increta or placenta percreta without CT or with CT 2–14 days before delivery in comparison with control group—without PAS and without CT (<b>A</b>). Levels of miR-382-5p (−∆Ct, PCR data) in the blood plasma of pregnant women with placenta accreta or placenta increta or placenta percreta without CT or with CT 2–14 days before delivery in comparison with control group—without PAS and without CT (<b>B</b>). Levels of miR-199a-3p (−∆Ct, PCR data) in the blood plasma of newborns from mothers with placenta accreta or placenta increta or placenta percreta without CT or with CT 2–14 days before delivery in comparison with control group—without PAS and without CT (<b>C</b>). Levels of miR-199a-3p (−∆Ct, PCR data) in the blood plasma of pregnant women with placenta accreta or placenta increta or placenta percreta without CT or with CT 2–14 days before delivery in comparison with control group—without PAS and without CT (<b>D</b>). “Wo” means “without”.</p>
Full article ">Figure 3
<p>Dynamics of changes in hsa-miR-199a-3p levels in the blood plasma of newborns relative to their mothers’ blood plasma, with and without PAS, depending on the antenatal corticosteroid therapy (CT). “Wo” means “without”.</p>
Full article ">Figure 4
<p>Levels of hsa-miR-199a-3p and hsa-miR-382-5p in the blood plasma of newborns with PAS, categorized by their severity score according to the Neomod scale.</p>
Full article ">Figure 5
<p>Levels of miR-181a-5p, miR-199a-3p and miR-382-5p in blood plasma of pregnant women with/without PAS and with/without antenatal corticosteroid therapy. “Wo” means “without”.</p>
Full article ">Figure 6
<p>Logistic regression models for predicting neonatal complications by plasma miR-199a-3p and/or miR-382-5p levels in pregnant women with PAS using miR-181a-5p as a reference endogenous RNA. (<b>A</b>) Respiratory complications probability models. (<b>B</b>) Cardiovascular complications probability models. Se—sensitivity, Sp—specificity.</p>
Full article ">Figure 7
<p>Enrichment analysis of gene targets of hsa-miR-382-5p and hsa-miR-199a-3p using FunRich software tool.</p>
Full article ">
14 pages, 10055 KiB  
Article
MicroRNA Profiling of PRELI-Modulated Exosomes and Effects on Hepatic Cancer Stem Cells
by Boyong Kim
Int. J. Mol. Sci. 2024, 25(24), 13299; https://doi.org/10.3390/ijms252413299 - 11 Dec 2024
Viewed by 255
Abstract
The increasing incidence and mortality rates of liver cancer have heightened the demand for the development of effective anticancer drugs with minimal side effects. In this study, the roles of exosomes derived from liver cancer stem cells (LCSCs) with PRELI (Protein of Relevant [...] Read more.
The increasing incidence and mortality rates of liver cancer have heightened the demand for the development of effective anticancer drugs with minimal side effects. In this study, the roles of exosomes derived from liver cancer stem cells (LCSCs) with PRELI (Protein of Relevant Evolutionary and Lymphoid Interest) modulation and their miRNAs were investigated to explore their therapeutic properties for liver cancer. Various techniques, such as miRNA profiling, microRNA transfection, overexpression, flow cytometry, Western blotting, and immunocytochemistry, were used to evaluate the effects of exosomes under PRELI up- and downregulation. Downregulated PRELI cellular exosomes (DPEs) reduced the levels of five markers—CD133, CD90, CD24, CD13, and EpCAM—in LCSCs, with the exception of OV-6. Conversely, upregulated PRELI cellular exosomes (UPEs) significantly increased the expression of CD90, CD24, and CD133 in NHs, with the maximum increase in CD24. PRELI upregulation altered expression levels of miRNAs, including hsa-miR-378a-3p (involved in stem-like properties), hsa-miR-25-3p (contributing to cell proliferation), and hsa-miR-423-3p (driving invasiveness). Exosomes with downregulated PRELI inhibited the AKT/mTORC1 signaling pathway, whereas LCSCs transfected with the candidate miRNAs activated it. Additionally, under PRELI upregulation, exosomes showed increased surface marker expression, promoting cancer progression. The modulation of PRELI in LCSCs affected miRNA expression significantly, revealing candidate miRNA targets for liver cancer treatment. Exosomes with PRELI downregulation show potential as a novel therapeutic strategy. Consequently, this study proposes the potential of PRELI-induced exosomes and the three miRNAs as a liver anticancer therapeutic candidate. Full article
(This article belongs to the Section Molecular Nanoscience)
Show Figures

Figure 1

Figure 1
<p>Purification of exosomes isolated from PRELI-modulated LCSCs. (<b>a</b>) Marker detection (CD63) and expression levels of PRELI in purified exosomes, as evaluated using Western blotting. Evaluation of purified exosomes with FITC-CD63 antibodies using flow cytometry (<b>b</b>) and fluorescence microscopy (<b>c</b>). CE: control cellular exosome, UPE: upregulated PRELI cellular exosome, DPE: downregulated PRELI cellular exosome, NC: unstained exosome (** <span class="html-italic">p</span> &lt; 0.01), scale bar = 20 µm.</p>
Full article ">Figure 2
<p>Profiling of exosomes from PRELI-modulated LCSCs. (<b>a</b>) Analyzed miRNAs (heat map) and identification of significant miRNAs in the exosomes. (<b>b</b>) Comparison of miRNA levels among three types of exosomes. Biochemical categories of the pie chart (1. aging, 2. angiogenesis, 3. apoptosis, 4. autophagy, 5. cell cycle, 6. cell differentiation, 7. cell migration, 8. cell proliferation, 9. DNA repair, 10. immune response, 11. inflammatory response, 12. neurogeneration, 13. secretion) associated with significant miRNAs in the induced exosomes and alterations of miRNA levels in each of the functional categories. The numbers above bar graphs indicate the number of differentially expressed miRNAs. CE: control cellular exosome, UPE: upregulated PRELI cellular exosome, DPE: downregulated PRELI cellular exosome.</p>
Full article ">Figure 3
<p>Alterations of miRNAs associated with six biochemical categories in three types of exosomes. (CE: control cellular exosome, UPE: upregulated PRELI cellular exosome, DPE: downregulated PRELI cellular exosome) (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Expression of markers in LCSCs exposed to various exosomes. (CE: control cellular exosomes, UPE: upregulated PRELI cellular exosome, DPE: downregulated PRELI cellular exosome). Fill-patterned bar graphs indicate markers with significant changes. Ns: not significant (* <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
<p>Expression of typical LCSC markers in normal hepatocytes exposed to various exosomes. (CE: control cellular exosome, UPE: upregulated PRELI cellular exosome, DPE: downregulated PRELI cellular exosome). Fill-patterned bar graphs indicate markers with significant changes. Ns: not significant (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Levels of AKT signaling molecules (phosphorylated AKT and phosphorylated mTORC1) in normal hepatocytes and LCSCs under various exosomes. (CE: control cellular exosome, UPE: upregulated PRELI cellular exosome, DPEs: downregulated PRELI cellular exosome) (* <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 7
<p>Expression of AKT signaling molecules in LCSCs under miRNA treatment. The merged images show the internalized fluorescence-labeled GAPDH-siRNAs (GFP: green fluorescent proteins) and miRNA (Cy5: cyanine 5) in LCSCs. The expression levels of AKT signaling molecules (phosphorylated AKT and phosphorylated mTORC1) in LCSCs transfected with the fluorescent (Cy5)-labeled miRNAs including hsa-miR378a-3p, hsa-miR25-3p, and hsa-miR423-3p and positive control siRNAs (GFP-GAPDH -siRNA) (* <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) (scale bars = 20 μm).</p>
Full article ">Figure 8
<p>Attenuation of drug resistance and cellular viability in SRHs upon inhibition of three miRNAs. (<b>a</b>) Results from modeling drug resistance. (<b>b</b>) Expression levels of PRELI in SSHs (sorafenib-sensitive hepatocytes) and SRH (sorafenib-resistant hepatocytes). (<b>c</b>) Drug resistance in cells transfected with anti-miRNA siRNAs. Fill-patterned bar graphs indicate sensitivity for Sorafenib.CC<sub>50</sub>: cytotoxic concentration 50; NT: non-transfected; T: transfected, Ns: not significant (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
31 pages, 2779 KiB  
Review
Emerging Sensing Technologies for Liquid Biopsy Applications: Steps Closer to Personalized Medicine
by Panagiota M. Kalligosfyri, Eleni Lamprou and Despina P. Kalogianni
Sensors 2024, 24(24), 7902; https://doi.org/10.3390/s24247902 - 11 Dec 2024
Viewed by 300
Abstract
Liquid biopsy is an efficient diagnostic/prognostic tool for tumor-derived component detection in peripheral circulation and other body fluids. The rapid assessment of liquid biopsy techniques facilitates early cancer diagnosis and prognosis. Early and precise detection of tumor biomarkers provides crucial information about the [...] Read more.
Liquid biopsy is an efficient diagnostic/prognostic tool for tumor-derived component detection in peripheral circulation and other body fluids. The rapid assessment of liquid biopsy techniques facilitates early cancer diagnosis and prognosis. Early and precise detection of tumor biomarkers provides crucial information about the tumor that guides clinicians towards effective personalized medicine. Point-of-care-testing remains still a great challenge in cancer diagnostics. Liquid biopsy is a promising alternative to tissue biopsy with the great advantages of less invasion and real-time monitoring of the disease, also providing information about tumor heterogeneity. The field is continuously and rapidly expanding. Numerous sophisticated biosensors have been developed targeting several biomarkers to achieve low detection limits, increased specificity and robustness. Current biosensors include mainly optical sensors, such as colorimetric, fluorescent, SPR, SERS and lateral flow assays. Electrochemical sensors have also been developed, providing very low detection limits. Colorimetric sensors exhibited simplicity in signal interpretation, while fluorescent sensors contributed to low analysis times, and SPR/SERS enabled label-free and rapid analysis. Novel target amplification and signal enhancement techniques have been exploited to increase the detectability of the sensors. In this context, this review is focused on the recent advances in biosensing technology for cutting-edge liquid biopsy applications towards point-of-care testing. Full article
(This article belongs to the Special Issue Feature Review Papers in Biosensors Section 2024)
Show Figures

Figure 1

Figure 1
<p>Overview of sensors for cutting-edge liquid biopsy applications.</p>
Full article ">Figure 2
<p>A colorimetric sensor for the detection of exosomes based on AuNP aggregation after aptamer-based capturing of exosomes, TdT elongation of the aptamers and NaCl addition [<a href="#B16-sensors-24-07902" class="html-bibr">16</a>].</p>
Full article ">Figure 3
<p>An SPR sensor based on polystyrene beads with a Ag/Au layer for signal enhancement coupled to antibodies for specific detection of neuron-specific enolase [<a href="#B38-sensors-24-07902" class="html-bibr">38</a>].</p>
Full article ">Figure 4
<p>A CRISPR/Cas12a system based on AuNPs and dark-field microscopy (DFM) for detecting BRCA-1 mutations related to breast cancer. Meanshift and partial least-square algorithms were used for signal interpretation [<a href="#B45-sensors-24-07902" class="html-bibr">45</a>].</p>
Full article ">Figure 5
<p>Detection of exosomes through RCA using an aptamer specific for exosomes and dimer-G4/thioflavin-based fluorescence output [<a href="#B49-sensors-24-07902" class="html-bibr">49</a>].</p>
Full article ">Figure 6
<p>An overview of the sensors and biomarkers of interest that have emerged for cutting-edge liquid biopsy towards point-of-care testing.</p>
Full article ">
17 pages, 1120 KiB  
Review
The Role of Bovine Milk-Derived Exosomes in Human Health and Disease
by Monika Jabłońska, Tomasz Sawicki, Justyna Żulewska, Katarzyna Staniewska, Adriana Łobacz and Katarzyna E. Przybyłowicz
Molecules 2024, 29(24), 5835; https://doi.org/10.3390/molecules29245835 - 11 Dec 2024
Viewed by 470
Abstract
Bovine milk is widely recognized as one of the most valuable sources of nutrients such as proteins, fats, vitamins, and minerals that support the development and health of the body. In recent years, there has been increasing scientific interest in exosomes, the small [...] Read more.
Bovine milk is widely recognized as one of the most valuable sources of nutrients such as proteins, fats, vitamins, and minerals that support the development and health of the body. In recent years, there has been increasing scientific interest in exosomes, the small membrane-bound vesicles found in milk. Through their content (e.g., microRNA), exosomes can influence gene expression and modulate key signaling pathways within target cells. Results from in vitro and in vivo studies have shown that bovine milk-derived exosomes can alleviate intestinal inflammation by regulating signaling pathways and positively influencing the composition of the gut microbiota. They also improve cognitive function and support nervous system regeneration. In addition, exosomes promote bone health by stimulating osteoblast formation and inhibiting bone resorption, helping to prevent osteoporosis. Studies have shown that exosomes have beneficial effects on skin health by promoting collagen production, protecting cells from oxidative stress, and delaying the ageing process. Bovine milk-derived exosomes are a promising tool for the treatment and prevention of a variety of diseases, particularly those related to inflammation and tissue regeneration. Although these results are promising, further studies are needed to fully understand the mechanisms of action and the potential clinical application of milk exosomes in the prevention and treatment of different diseases. Full article
Show Figures

Figure 1

Figure 1
<p>The effects of bovine milk-derived exosomes on human health and diseases. The graphics presented in this figure were created with the assistance of ChatGPT4.</p>
Full article ">
16 pages, 2759 KiB  
Article
Dysregulation of MiR-21, MiR-221 and MiR-451 During Neoadjuvant Treatment of Breast Cancer: A Prospective Study
by Carine Bispo Patury, Brenda Luanny Maia Santos, Anna Lucia Carvalho Matos, José Slabi, Luciene Cristina Campos Gastalho and Carla Martins Kaneto
Biomolecules 2024, 14(12), 1580; https://doi.org/10.3390/biom14121580 - 11 Dec 2024
Viewed by 308
Abstract
Background: Breast cancer is highly heterogeneous disease in which different responses are observed to the same treatment for different subtypes and extents of similar diseases. Considering this scenario, the search for tumor biomarkers is indispensable, with current evidence suggesting that circulating microRNAs are [...] Read more.
Background: Breast cancer is highly heterogeneous disease in which different responses are observed to the same treatment for different subtypes and extents of similar diseases. Considering this scenario, the search for tumor biomarkers is indispensable, with current evidence suggesting that circulating microRNAs are viable biomarkers. This study evaluated the expression of miR-21, miR-221, miR-195, and miR-451 in patients with breast cancer undergoing neoadjuvant treatment at oncology outpatient facilities in Brazil. Methods: We conducted a prospective and observational study in which blood samples were collected for microRNA expression analysis, comparing control and breast cancer patients who were candidates for neoadjuvant treatment groups. The expression of microRNAs was investigated by qRT-PCR method. For parametric data analysis, one-way ANOVA with Tukey’s post hoc test was used. Results: Thirty-three participants (all female) were included in the control group and twenty-seven participants were included in the study group. The non-special subtype of breast cancer was found in 96% of the study group participants; 88.9% were locally advanced tumors (T3, T4), 40.7% were luminal tumors, 33.3% were HER-2-positive, and 26% were triple negative tumors. Expression analysis of microRNAs during neoadjuvant treatment, using miR-16 as a normalizer, showed higher expression levels of miR-21 and miR-221 at the end of treatment, and high expression levels for miR-451 were also observed at the beginning of treatment. Conclusion: This is the first study that evaluates the expression of microRNAs in the context of neoadjuvant treatment of breast cancer in the Brazilian population. Our results suggest that there is a deregulation of miR-21, miR-221, and miR-451 during neoadjuvant treatment in these patients. Full article
(This article belongs to the Special Issue Emerging Biomarkers Discovery for Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Analysis of the cycling threshold by qRT-PCR of miR-320a, miR-16, and U6 and their respective significance levels (<span class="html-italic">p</span>) in samples from the control group (n: 33) and the study group (n: 58). The evaluation was performed in 33 samples of the control group and in 58 samples of the group of patients with breast cancer, with collection 1 being performed before the neoadjuvant treatment (22 samples), collection 2 when 50% of the neoadjuvant treatment had been performed (21 samples), and collection 3 after the end of the neoadjuvant treatment (15 samples).</p>
Full article ">Figure 2
<p>Stability levels of MiR-320a, MiR-16, and U6 evaluated in samples from the control group (n: 33) and the study group with breast cancer (n: 58) by several platforms and by Delta CT: (<b>a</b>) BestKeeper; (<b>b</b>) Delta CT (cycling threshold); (<b>c</b>) Genorm; (<b>d</b>) NormFinder; (<b>e</b>) Global analysis by RefFinder.</p>
Full article ">Figure 3
<p>Stability levels of MiR-320a, MiR-16, and U6 evaluated in samples from the control group (n: 33) and the study group with breast cancer (n: 58) by the NormFinder platform.</p>
Full article ">Figure 4
<p>Relative expression of miR-21. Total N: 85 samples; control, n: 29; collection 1 (before neoadjuvant treatment), n: 22; collection 2 (after 50% of the neoadjuvant treatment), n: 20; collection 3 (after the end of neoadjuvant treatment and before surgery), n: 14.</p>
Full article ">Figure 5
<p>Relative expression of miR-195. Total N: 85 samples; control, n: 29; collection 1 (before neoadjuvant treatment), n: 22; collection 2 (after 50% of the neoadjuvant treatment), n: 20; collection 3 (after the end of neoadjuvant treatment and before surgery), n: 14.</p>
Full article ">Figure 6
<p>Relative expression of miR-451. Total N: 85 samples; control, n: 29; collection 1 (before neoadjuvant treatment), n: 22; collection 2 (after 50% of the neoadjuvant treatment), n: 20; collection 3 (after the end of neoadjuvant treatment and before surgery), n: 14.</p>
Full article ">Figure 7
<p>Relative expression of miR-221. Total N: 85 samples; control, n: 29; collection 1 (before neoadjuvant treatment), n: 22; collection 2 (after 50% of the neoadjuvant treatment), n: 20; collection 3 (after the end of neoadjuvant treatment and before surgery), n: 14.</p>
Full article ">
12 pages, 782 KiB  
Article
Prognostic Significance of miRNA Subtypes in Melanoma: A Survival Analysis and Correlation with Treatment Response Across Patient Stages
by Mihaela Prodan, Alis Dema, Bianca Roxana Nataras, Edward Seclaman, Vlad Bloanca, Zorin Crainiceanu, Ilona Emoke Deak, Claudia Raluca Balasa Virzob, Ana-Olivia Toma and Roxana Manuela Fericean
Biomedicines 2024, 12(12), 2809; https://doi.org/10.3390/biomedicines12122809 - 11 Dec 2024
Viewed by 323
Abstract
Background and Objectives: Melanoma remains a leading cause of skin cancer mortality despite advancements in targeted therapies and immunotherapies. MicroRNAs (miRNAs) have emerged as potential biomarkers for cancer prognosis and treatment response. This study aims to analyze survival outcomes according to various miRNA [...] Read more.
Background and Objectives: Melanoma remains a leading cause of skin cancer mortality despite advancements in targeted therapies and immunotherapies. MicroRNAs (miRNAs) have emerged as potential biomarkers for cancer prognosis and treatment response. This study aims to analyze survival outcomes according to various miRNA subtypes, assess the association between specific miRNAs and treatment response, and include patient staging to evaluate their prognostic significance. Methods: A retrospective cohort study was conducted on 90 patients from the Pius Brinzeu County Emergency Clinical Hospital, Timisoara, between 2019 and 2022. The cohort included 45 patients with advanced-stage melanoma and 45 with benign nevi. miRNA expression levels were quantified using the miRNeasy Kit and the Human Cancer PathwayFinder miScript miRNA PCR Array. Survival analysis was performed using the Kaplan–Meier method, and Cox proportional hazards models were used to assess the impact of miRNA expression on survival. Logistic regression analyzed the association between miRNA markers and treatment response, adjusting for patient staging. Results: Elevated levels of hsa-miR-200a-3p and hsa-miR-335-5p were significantly associated with poorer overall survival (p < 0.01), particularly in stage III and IV patients. Conversely, higher expression of hsa-miR-451a correlated with improved survival rates (p = 0.02). Patients with increased hsa-miR-29b-3p expression showed a better response to immunotherapy (OR = 2.35, 95% CI: 1.15–4.79). Multivariate analysis confirmed that miRNA expression levels and patient staging were independent predictors of survival and treatment response. Conclusions: Specific miRNA subtypes are significant prognostic markers in melanoma, influencing survival outcomes and treatment responses across different patient stages. Incorporating miRNA profiling into clinical practice could enhance personalized treatment strategies and improve patient prognoses. Full article
(This article belongs to the Section Immunology and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier survival analysis based on miRNA expression.</p>
Full article ">Figure 2
<p>Forest plot analysis.</p>
Full article ">
21 pages, 4080 KiB  
Review
MicroRNA-Based Liquid Biopsy for Cervical Cancer Diagnostics and Treatment Monitoring
by Maria A. Kepsha, Angelika V. Timofeeva, Vasiliy S. Chernyshev, Denis N. Silachev, Elena A. Mezhevitinova and Gennadiy T. Sukhikh
Int. J. Mol. Sci. 2024, 25(24), 13271; https://doi.org/10.3390/ijms252413271 - 10 Dec 2024
Viewed by 699
Abstract
Despite prevention strategies, cervical cancer remains a significant public health issue. Human papillomavirus plays a critical role in its development, and early detection is vital to improve patient outcomes. The incidence of cervical cancer is projected to rise, necessitating better diagnostic tools. Traditional [...] Read more.
Despite prevention strategies, cervical cancer remains a significant public health issue. Human papillomavirus plays a critical role in its development, and early detection is vital to improve patient outcomes. The incidence of cervical cancer is projected to rise, necessitating better diagnostic tools. Traditional screening methods like the cytological examination and human papillomavirus testing have limitations in sensitivity and reproducibility. Liquid-based cytology offers some improvements, but the need for more reliable and sensitive techniques persists, particularly for detecting precancerous lesions. Liquid biopsy is a non-invasive method that analyzes cancer-derived products in biofluids like blood, offering potential for real-time monitoring of tumor progression, metastasis, and treatment response. It can be based on detection of circulating tumor cells (CTCs), circulating free DNA (cfDNA), and microRNAs (miRNAs). This review particularly underlines the potential of microRNAs, which are transported by extracellular vesicles. Overall, this article underscores the importance of continued research into non-invasive diagnostic methods like liquid biopsy to enhance cervical cancer screening and treatment monitoring. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Illustration summarizing the stages of CC progression with comparison to a normal cervix and highlighting biofluids that can potentially be used for liquid biopsy. HPV—human papillomavirus; LSIL—low-grade squamous intraepithelial lesions; HSIL—high-grade squamous intraepithelial lesions; CTCs—circulating tumor cells; EVs—extracellular vesicles; ccfRNA—cell-free circulating RNA; ccfDNA—cell-free circulating DNA.</p>
Full article ">Figure 2
<p>Illustration depicting liquid biopsy where blood or cervicovaginal lavage is obtained from a patient to isolate and characterize EVs, miRNA, and/or IncRNA for eventual diagnostics. EVs—extracellular vesicles.</p>
Full article ">Figure 3
<p>Cellular localization of experimentally validated gene-targets protein products of CC microRNA markers presented in <a href="#ijms-25-13271-t001" class="html-table">Table 1</a> and <a href="#ijms-25-13271-t002" class="html-table">Table 2</a>, generated by the FunRich (Version 3.1, last accessed on 1 September 2024) program.</p>
Full article ">Figure 4
<p>Organ expression of target genes for CC miRNA markers. Data are generated using the FunRich (Version 3.1, last accessed on 1 September 2024) tool. The red diamonds indicate the main sites of metastasis for the CC.</p>
Full article ">Figure 5
<p>Molecular function of gene-targets protein products for CC miRNA markers. Data are generated using the FunRich (Version 3.1, last accessed on 1 September 2024) tool.</p>
Full article ">Figure 6
<p>Signaling pathways formed by gene-targets protein products for CC miRNA markers. Data are generated by using the FunRich (Version 3.1, last accessed on 1 September 2024) tool.</p>
Full article ">
Back to TopTop