[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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (284)

Search Parameters:
Keywords = p21/CDKN1A

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 991 KiB  
Review
A Critical Review on microRNAs as Prognostic Biomarkers in Laryngeal Carcinoma
by Kristina S. Komitova, Lyuben D. Dimitrov, Gergana S. Stancheva, Silva G. Kyurkchiyan, Veronika Petkova, Stoyan I. Dimitrov, Silviya P. Skelina, Radka P. Kaneva and Todor M. Popov
Int. J. Mol. Sci. 2024, 25(24), 13468; https://doi.org/10.3390/ijms252413468 - 16 Dec 2024
Viewed by 371
Abstract
During the past decade, a vast number of studies were dedicated to unravelling the obscurities of non-coding RNAs in all fields of the medical sciences. A great amount of data has been accumulated, and consequently a natural need for organization and classification in [...] Read more.
During the past decade, a vast number of studies were dedicated to unravelling the obscurities of non-coding RNAs in all fields of the medical sciences. A great amount of data has been accumulated, and consequently a natural need for organization and classification in all subfields arises. The aim of this review is to summarize all reports on microRNAs that were delineated as prognostic biomarkers in laryngeal carcinoma. Additionally, we attempt to allocate and organize these molecules according to their association with key pathways and oncogenes affected in laryngeal carcinoma. Finally, we critically analyze the common shortcomings and biases of the methodologies in some of the published papers in this area of research. A literature search was performed using the PubMed and MEDLINE databases with the keywords “laryngeal carcinoma” OR “laryngeal cancer” AND “microRNA” OR “miRNA” AND “prognostic marker” OR “prognosis”. Only research articles written in English were included, without any specific restrictions on study type. We have found 43 articles that report 39 microRNAs with prognostic value associated with laryngeal carcinoma, and all of them are summarized along with the major characteristics and methodology of the respective studies. A second layer of the review is structural analysis of the outlined microRNAs and their association with oncogenes and pathways connected with the cell cycle (p53, CCND1, CDKN2A/p16, E2F1), RTK/RAS/PI3K cascades (EGFR, PI3K, PTEN), cell differentiation (NOTCH, p63, FAT1), and cell death (FADD, TRAF3). Finally, we critically review common shortcomings in the methodology of the papers and their possible effect on their results. Full article
(This article belongs to the Special Issue The Role of RNAs in Cancers: Recent Advances)
Show Figures

Figure 1

Figure 1
<p>Representation of miRs with prognostic value in laryngeal carcinoma and their association with major oncogenes/pathways (Euler diagram). Asterix displays available data for association with <span class="html-italic">FAT1</span> gene.</p>
Full article ">
11 pages, 1582 KiB  
Article
PIK3CA Mutations and Co-Mutations in Operated Non-Small Cell Lung Carcinoma
by Salih Cokpinar, Ibrahim Halil Erdogdu, Seda Orenay-Boyacioglu, Olcay Boyacioglu, Nesibe Kahraman-Cetin and Ibrahim Meteoglu
J. Clin. Med. 2024, 13(23), 7472; https://doi.org/10.3390/jcm13237472 - 8 Dec 2024
Viewed by 612
Abstract
Background: Understanding PIK3CA mutations and co-mutations in non-small cell lung carcinoma (NSCLC) is critical to developing personalized treatment strategies. Therefore, this study aims to investigate PIK3CA mutations and the accompanying somatic variations in NSCLC. Methods: This retrospective study included 98 patients over 18 [...] Read more.
Background: Understanding PIK3CA mutations and co-mutations in non-small cell lung carcinoma (NSCLC) is critical to developing personalized treatment strategies. Therefore, this study aims to investigate PIK3CA mutations and the accompanying somatic variations in NSCLC. Methods: This retrospective study included 98 patients over 18 years of age who were diagnosed with NSCLC, operated on, and referred to the Molecular Pathology Laboratory between January 2019 and June 2024 for next-generation sequencing panel tests and ALK-ROS1 FISH analysis. Results: All patients were found to carry PIK3CA mutations. Among the 98 NSCLC patients analyzed, 16 (16.33%) were female and 82 (83.67%) were male. The average age of the patients was 64.53 ± 9.63 years, with an age range of 38–84 years, and the majority were 50 years or older. Of the cases, 51 presented the adenocarcinoma subtype, while the remaining 47 showed the squamous cell carcinoma subtype. A smoking history was present in 77 (78.57%) patients, while 21 (21.43%) had no smoking history. The most frequently detected pathogenic or likely pathogenic PIK3CA variations were c.1633G>A p.E545K (32.65%), c.1624G>A p.E542K (11.22%), c.3140A>G p.H1047R (11.22%), c.3140A>T p.H1047L (5.10%), c.1357G>C p.E453Q (4.08%), and c.3143A>G p.H1048R (2.04%). The top 10 mutations that most commonly accompanied PIK3CA variations were KRAS, NF1, TP53, EGFR, PTEN, BRAF, KIT, CDKN2A, SMARCA4, and ATM mutations, respectively. Conclusions: PIK3CA variations, along with other gene variations, may influence cancer progression and thus may play a crucial role in the determination of targeted treatment strategies. Full article
(This article belongs to the Section Pulmonology)
Show Figures

Figure 1

Figure 1
<p>Oncoprint diagram of all mutations co-occuring with <span class="html-italic">PIK3CA</span> variants.</p>
Full article ">Figure 2
<p>Distribution of <span class="html-italic">PIK3CA</span> co-mutations based on smoking status in NSCLC patients.</p>
Full article ">
26 pages, 3816 KiB  
Article
Genomic Profiling in Glioma Patients to Explore Clinically Relevant Markers
by Viacheslav Varachev, Olga Susova, Alexei Mitrofanov, David Naskhletashvili, George Krasnov, Anna Ikonnikova, Svetlana Bezhanova, Vera Semenova, Nadezhda Sevyan, Evgenii Prozorenko, Yulia Ammour, Ali Bekyashev and Tatiana Nasedkina
Int. J. Mol. Sci. 2024, 25(23), 13004; https://doi.org/10.3390/ijms252313004 - 3 Dec 2024
Viewed by 667
Abstract
Gliomas are a heterogeneous group of brain tumors, among which the most aggressive subtype is glioblastoma, accounting for 60% of cases in adults. Available systemic treatment options are few and ineffective, so new approaches to therapies for glioblastoma are in high demand. In [...] Read more.
Gliomas are a heterogeneous group of brain tumors, among which the most aggressive subtype is glioblastoma, accounting for 60% of cases in adults. Available systemic treatment options are few and ineffective, so new approaches to therapies for glioblastoma are in high demand. In total, 131 patients with diffuse glioma were studied. Paired tumor–normal samples were sequenced on the Illumina platform; the panel included 812 genes associated with cancer development. Molecular profiles in clinically distinct groups were investigated. In low-grade glioma (LGG) patients (n = 18), the most common mutations were IDH1/2 (78%), ATRX (33%), TP53 (33%), PIK3CA (17%), and co-deletion 1p/19q (22%). In high-grade glioma (HGG) patients (n = 113), more frequently affected genes were CDKN2A/B (33%), TERTp (71%), PTEN (60%), TP53 (27%), and EGFR (40%). The independent predictors of better prognosis were tumor grade and IDH1/2 mutations. In IDH—wildtype glioblastoma patients, a history of other precedent cancer was associated with worse overall survival (OS), while re-operation and bevacizumab therapy increased OS. Also, among genetic alterations, TERTp mutation and PTEN deletion were markers of poor prognosis. Nine patients received molecular targeted therapy, and the results were evaluated. The search for molecular changes associated with tumor growth and progression is important for diagnosis and choice of therapy. Full article
(This article belongs to the Special Issue Current Developments in Glioblastoma Research and Therapy)
Show Figures

Figure 1

Figure 1
<p>Genomic landscape of low-grade glioma (LGG) and high-grade glioma (HGG) patients. (amp—amplification, mut—mutation, met—methylated, unmet—unmethylated, na—not applicable, BEV(+)—patients received bevacizumab, BEV(-)—patients not receiving bevacizumab, SPGB—second primary glioblastoma, GB—glioblastoma, ODG—oligodendroglioma, DA2-3—diffuse astrocytoma Grade 2-3, DA4—diffuse astrocytoma Grade 4, co-del—co-deletion, chr—chromosome) (<a href="#app1-ijms-25-13004" class="html-app">Table S1</a>).</p>
Full article ">Figure 2
<p>Frequency of genetic alterations in patients with LGG and HGG in comparison with data from public databases (cBioPortal, Glioblastoma multiforme TCGA PanCancer Atlas 2018). Significant difference between LGG and HGG is marked by (*).</p>
Full article ">Figure 3
<p>Overall survival rate depending on tumor grade (<b>a</b>) and <span class="html-italic">IDH1/2</span> mutational status (<b>b</b>). HGG: high-grade glioma, LGG: low:grade glioma.</p>
Full article ">Figure 4
<p>Impact of clinical features on overall survival (months) in patients with IDH—wildtype glioblastoma: age (<b>a</b>), cancer history (<b>b</b>), tumor location (<b>c</b>), number of surgical resections (<b>d</b>), treatment with bevacizumab (<b>e</b>), MGMT promoter methylation (<b>f</b>).</p>
Full article ">Figure 5
<p>Impact of gene alteration on overall survival (months) in patients with <span class="html-italic">IDH</span>—wildtype glioblastoma: <span class="html-italic">CDKN2A/B</span> deletion (<b>a</b>), <span class="html-italic">TERT</span> promoter mutation (<b>b</b>), <span class="html-italic">PTEN</span> mutation (<b>c</b>), <span class="html-italic">PTEN</span> deletion (<b>d</b>), <span class="html-italic">TP53</span> mutation (<b>e</b>), <span class="html-italic">EGFR</span> alteration (mutation or amplification) (<b>f</b>), chromosome 7 gain (<b>g</b>), and chromosome 10 loss (<b>h</b>).</p>
Full article ">Figure 6
<p>Follow-up of patients during treatment with targeted therapies.</p>
Full article ">
17 pages, 2902 KiB  
Article
Variable Expression of Oncogene-Induced Senescence/SASP Surrogates in HPV-Associated Precancerous Cervical Tissue
by Tareq Saleh, Nisreen Himsawi, Amani Al Rousan, Ahmad Alhesa, Mohammed El-Sadoni, Suzan Khawaldeh, Nisreen Abu Shahin, Ala’ Abu Ghalioun, Bayan Shawish, Kholoud Friehat, Moureq R. Alotaibi, Ola Abu Al Karsaneh, Anas Abu-Humaidan, Rame Khasawneh, Ashraf I. Khasawneh and Sofian Al Shboul
Curr. Issues Mol. Biol. 2024, 46(12), 13696-13712; https://doi.org/10.3390/cimb46120818 - 2 Dec 2024
Viewed by 562
Abstract
Oncogene-induced senescence (OIS) is a form of cellular senescence triggered by oncogenic signaling and, potentially, by infection with oncogenic viruses. The role of senescence, along with its associated secretory phenotype, in the development of cervical cancer remains unclear. Additionally, the expression of the [...] Read more.
Oncogene-induced senescence (OIS) is a form of cellular senescence triggered by oncogenic signaling and, potentially, by infection with oncogenic viruses. The role of senescence, along with its associated secretory phenotype, in the development of cervical cancer remains unclear. Additionally, the expression of the senescence-associated secretory phenotype (SASP) has not yet been explored in cervical premalignant lesions infected by the Human Papilloma Virus (HPV). This study aimed to investigate the expression of OIS and SASP markers in HPV-infected cervical precancerous lesions. We used a set of patient-derived precancerous (n = 32) and noncancerous (chronic cervicitis; n = 10) tissue samples to investigate the gene expression of several OIS (LMNB1, CDKN2A, CDKN2B, and CDKN1A), and SASP (IL1A, CCL2, TGFB1, CXCL8, and MMP9) biomarkers using qRT-PCR. OIS status was confirmed in precancerous lesions based on Lamin B1 downregulation by immunohistochemical staining. HPV status for all precancerous lesions was tested. Most of the noncancerous samples showed high Lamin B1 expression, however, precancerous lesions exhibited significant Lamin B1 downregulation (p < 0.001). Fifty-five percent of the precancerous samples were positive for HPV infection, with HPV-16 as the dominant genotype. Lamin B1 downregulation coincided with HPV E6 positive expression. CDKN2A and CDKN2B expression was higher in precancerous lesions compared to noncancerous tissue, while LMNB1 was downregulated. The SASP profile of premalignant lesions included elevated CXCL8 and TGFB1 and reduced IL1A, CCL2, and MMP9. this work shall provide an opportunity to further examine the role of OIS and the SASP in the process of malignant cervical transformation. Full article
(This article belongs to the Special Issue Molecular Mechanism of HPV’s Involvement in Cancers)
Show Figures

Figure 1

Figure 1
<p>Distribution of HPV genotypes in cervical precancerous lesions. The bar chart shows HPV genotype distribution among cervical precancerous samples. Of these, 55% tested HPV-positive, with 78% associated with high-risk HPV subtypes. HPV-16 was the most common (72%), followed by HPV-18 (61%). Negative cases accounted for 45% of the sample. No variability metrics are shown as the data were derived from tabular representation.</p>
Full article ">Figure 2
<p>Immunohistochemical analysis of Lamin B1 expression in precancerous and noncancerous cervical tissue. A representative image of Lamin B1 expression in noncancerous cervical tissue (<b>A</b>) and (<b>B</b>) precancerous cervical tissues. (<b>C</b>) Quantification of Lamin B1-positive cells showing a significant reduction in precancerous cervical samples compared to noncancerous tissue (** <span class="html-italic">p</span> &lt; 0.001). Data are presented as the median with 95% confidence intervals.</p>
Full article ">Figure 3
<p>Fold change in mRNA expression levels of OIS markers in precancerous relative to noncancerous cervical tissue. <span class="html-italic">LMNB1</span> expression was significantly downregulated in precancerous lesions, confirming its immunohistochemically detected, reduced protein expression. In contrast, <span class="html-italic">CDKN2B</span> and <span class="html-italic">CDKN2A</span> (p15<sup>INK4b</sup> and p16<sup>INK4a</sup>, respectively) were both significantly upregulated, indicating senescence activation. <span class="html-italic">CDKN1A</span> (p21<sup>Cip1</sup>) exhibited a variable expression pattern without consistent upregulation, suggesting its differential regulation in the precancerous state. Data are presented as log<sub>2</sub> fold change, with each dot representing an individual (precancerous) sample. Created in BioRender.com.</p>
Full article ">Figure 4
<p>Co-immunofluorescent staining of HPV oncoprotein E6 and Lamin B1 in cervical precancerous lesions. Representative images show DAPI-stained nuclei (blue), Lamin B1 staining (red), E6 staining (green), and merged channels. The graph below quantifies the percentage of positive cells for E6, Lamin B1, and the co-localization of E6+/Lamin B1 cells. A total of 41% of the E6-positive cells exhibit downregulation of Lamin B1, suggesting that HPV E6 may be involved in driving the OIS phenotype in these lesions. Scale bar = 20 µm.</p>
Full article ">Figure 5
<p>Gene expression analysis of SASP-related markers in cervical precancerous lesions compared to noncancerous inflammatory lesions. The box plot illustrates the fold change (log<sub>2</sub>) in expression of <span class="html-italic">MMP9</span>, <span class="html-italic">IL1A</span>, <span class="html-italic">CCL2</span>, <span class="html-italic">CXCL8</span>, and <span class="html-italic">TGFB1</span>. <span class="html-italic">IL1A</span> and <span class="html-italic">CCL2</span> expression was reduced in precancerous lesions, while <span class="html-italic">CXCL8</span> and <span class="html-italic">TGFB1</span> exhibited mild upregulation. <span class="html-italic">MMP9</span> expression was heterogeneous, with some samples showing upregulation and others showing downregulation. These data reflect the variability in SASP marker expression in precancerous lesions. Created in BioRender.com.</p>
Full article ">
19 pages, 3392 KiB  
Article
Impact of Short-Term Exposure to Non-Functionalized Polystyrene Nanoparticles on DNA Methylation and Gene Expression in Human Peripheral Blood Mononuclear Cells
by Kinga Malinowska, Kateryna Tarhonska, Marek Foksiński, Paulina Sicińska, Ewa Jabłońska, Edyta Reszka, Ewelina Zarakowska, Daniel Gackowski, Karolina Górecka, Aneta Balcerczyk and Bożena Bukowska
Int. J. Mol. Sci. 2024, 25(23), 12786; https://doi.org/10.3390/ijms252312786 - 28 Nov 2024
Viewed by 416
Abstract
The aim of the present study was to investigate the concentration- and size-dependent effects of non-functionalized polystyrene nanoparticles (PS-NPs) of varying diameters (29 nm, 44 nm, and 72 nm) on specific epigenetic modifications and gene expression profiles related to carcinogenesis in human peripheral [...] Read more.
The aim of the present study was to investigate the concentration- and size-dependent effects of non-functionalized polystyrene nanoparticles (PS-NPs) of varying diameters (29 nm, 44 nm, and 72 nm) on specific epigenetic modifications and gene expression profiles related to carcinogenesis in human peripheral blood mononuclear cells (PBMCs) in vitro. This in vitro human-cell-based model is used to investigate the epigenetic effect of various environmental xenobiotics. PBMCs were exposed to PS-NPs at concentrations ranging from 0.001 to 100 µg/mL for 24 h period. The analysis encompassed epigenetic DNA modifications, including levels of 5-methyl-2′-deoxycytidine (5-mdC) and 5-(hydroxymethyl)-2′-deoxycytidine (5-hmdC), as well as the levels of 2′-deoxyuridine (dU) and 5-(hydroxymethyl)-2′-deoxyuridine (5-hmdU) by mass spectrometry methods, methylation in the promoter regions of selected tumor suppressor genes TP53 (P53), CDKN2A (P16), and CDKN1A (P21) and proto-oncogenes (CCND1, BCL2, BCL6), along with the expression profile of the indicated genes by real-time PCR assays. The results obtained revealed no significant changes in global DNA methylation/demethylation levels in PBMCs after short-term exposure to non-functionalized PS-NPs. Furthermore, there were no changes observed in the level of dU, a product of cytosine deamination. However, the level of 5-hmdU, a product of both 5-hmdC deamination and thymine oxidation, was increased at the highest concentrations of larger PS-NPs (72 nm). None of the PS-NPs caused a change in the methylation pattern of the promoter regions of the TP53, CDKN2A, CDKN1A, CCND1, BCL2 and BCL6 genes. However, gene profiling indicated that PS-NPs with a diameter of 29 nm and 44 nm altered the expression of the TP53 gene. The smallest PS-NPs with a diameter of 29 nm increased the expression of the TP53 gene at a concentration of 10 µg/mL, while PS-NPs with a diameter of 44 nm did so at a concentration of 100 µg/mL. An increase in the expression of the CDKN2A gene was also observed when PBMCs were exposed to PS-NPs with 29 nm in diameter at the highest concentration. The observed effect depended on both the concentration and the size of the PS-NPs. Full article
(This article belongs to the Topic Environmental Toxicology and Human Health—2nd Edition)
Show Figures

Figure 1

Figure 1
<p>The level of (<b>A</b>) 5-mdC and (<b>B</b>) 5-hmdC per 10<sup>3</sup> dN (10<sup>3</sup> deoxyribonucleotides) in human PBMCs incubated with PS-NPs of 29, 44, and 72 nm in diameter (0.001–100 μg/mL) for 24 h period. The data are presented as mean ± SD, n = 4 independent experiments. Statistical analysis was conducted using one-way ANOVA or Kruskal–Wallis test.</p>
Full article ">Figure 2
<p>The level of (<b>A</b>) dU and (<b>B</b>) 5-hmdU per 10<sup>6</sup> dN (10<sup>6</sup> deoxyribonucleotides) in human PBMCs incubated with PS-NPs of 29, 44, and 72 nm in diameter (0.001–100 μg/mL) for 24 h period. The data are presented as mean ± SD, n = 4 independent experiments. Statistical analysis was conducted using one-way ANOVA or Kruskal–Wallis test, followed by relevant post hoc; * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Methylation of suppressor genes (<span class="html-italic">TP53</span>, <span class="html-italic">CDKN2A</span>, <span class="html-italic">CDKN1A</span>) in human PBMCs incubated with PS-NPs of 29, 44, and 72 nm in diameter (0.001–100 μg/mL) for 24 h period. The data are presented as mean ± SD, n = 3 to 4 independent experiments. The promoter methylation experiments for 29, 44, and 72 nm were performed on separate plates, each including a respective control group (cells incubated in medium only). Consequently, baseline MI levels varied for some genes across different PS-NP sizes. Statistical analysis was conducted using one-way ANOVA or Kruskal–Wallis test.</p>
Full article ">Figure 4
<p>Methylation of proto-oncogenes (<span class="html-italic">CCND1</span>, <span class="html-italic">BCL2</span>, <span class="html-italic">BCL6</span>) in human PBMCs incubated with PS-NPs of 29, 44, and 72 nm in diameter (0.001–100 μg/mL) for 24 h period. The data are presented as mean ± SD, n = 3 to 4 independent experiments. The promoter methylation experiments for 29, 44, and 72 nm were performed on separate plates, each including a respective control group (cells incubated in medium only). Consequently, baseline MI levels varied for some genes across different PS-NP sizes. Statistical analysis was conducted using one-way ANOVA or Kruskal–Wallis test.</p>
Full article ">Figure 5
<p>Expression of suppressor genes (<span class="html-italic">TP53</span>, <span class="html-italic">CDKN2A</span> and <span class="html-italic">CDKN1A</span>) in human PBMCs incubated with PS-NPs of 29, 44, and 72 nm in diameter (0.001–100 μg/mL) for 24 h period. The data are presented as mean ± SD, n = 3 to 4 independent experiments. Statistical analysis was conducted using one-way ANOVA or Kruskal–Wallis test, followed by relevant post hoc test, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Expression of proto-oncogenes (<span class="html-italic">CCND1</span>, <span class="html-italic">BCL2</span>, <span class="html-italic">BCL6</span>) in human PBMCs incubated with PS-NPs of 29, 44, and 72 nm in diameter (0.001–100 μg/mL) for 24 h period. The data are presented as mean ± SD, n = 3 to 4 independent experiments. Statistical analysis was conducted using one-way ANOVA or Kruskal–Wallis test.</p>
Full article ">Figure 7
<p>Flow chart of experimental design.</p>
Full article ">
11 pages, 551 KiB  
Article
Genomic Analysis of Advanced Phyllodes Tumors Using Next-Generation Sequencing and Their Chemotherapy Response: A Retrospective Study Using the C-CAT Database
by Shuhei Suzuki and Yosuke Saito
Medicina 2024, 60(11), 1898; https://doi.org/10.3390/medicina60111898 - 19 Nov 2024
Viewed by 861
Abstract
Background and Objectives: Phyllodes tumors are rare breast neoplasms with limited therapeutic options and poorly understood molecular characteristics. This study aimed to analyze genomic alterations and treatment outcomes in advanced phyllodes tumors using Japan’s national clinical genomic testing registry (C-CAT database) to [...] Read more.
Background and Objectives: Phyllodes tumors are rare breast neoplasms with limited therapeutic options and poorly understood molecular characteristics. This study aimed to analyze genomic alterations and treatment outcomes in advanced phyllodes tumors using Japan’s national clinical genomic testing registry (C-CAT database) to identify potential therapeutic targets and predictive markers. Materials and Methods: We conducted a retrospective analysis of 60 phyllodes tumor cases from 80,329 patients registered in the C-CAT database between June 2019 and August 2024. Comprehensive genomic profiling was performed using multiple platforms including FoundationOne CDx, NCC OncoPanel, and other approved tests. Treatment responses were evaluated according to RECIST criteria, and pathogenic variants were assessed using established databases including ClinVar and OncoKB. Results: The cohort’s median age was 54 years (range: 13–79), with TERT promoter variants (70%), MED12 (52%), and TP53 (50%) mutations being the most frequent alterations. Forty patients received first-line chemotherapy, predominantly anthracycline-based regimens (n = 29). Although not reaching statistical significance, cases with CDKN2A and TERT alterations showed trends toward treatment resistance (OR > 3.0). One patient with a high tumor mutational burden (37/Mb) responded to pembrolizumab. Potential germline variants were identified in two cases (3.3%), involving MSH6 and TP53 alterations. Notably, no cases with CDKN2B alterations demonstrated treatment response (p = 0.09). Conclusions: Our findings suggest distinct molecular patterns in phyllodes tumors compared to other soft tissue sarcomas, with potential implications for treatment selection. The identification of specific genetic alterations associated with treatment resistance may guide therapeutic decision-making, while the presence of actionable mutations in select cases indicates potential opportunities for targeted therapy approaches. Full article
(This article belongs to the Collection Frontiers in Breast Cancer Diagnosis and Treatment)
Show Figures

Figure 1

Figure 1
<p>Genomic alterations and treatment responses of phyllodes tumor cases registered in the Center for Cancer Genomics and Advanced Therapeutics. Notable germline findings include MSH6 (T719fs*17, AF = 0.28) and TP53 (G245S, AF = 0.77) alterations, both classified as presumed germline pathogenic variants (PGPVs) according to Japanese PGPV criteria (Kosugi Group Criteria Ver.4; AF cutoff &gt;0.2 for truncating variants in MSS tumors and &gt;0.3 for TP53 variants in patients ≤30 years). TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated; F: FoundationOne<sup>®</sup> CDx; FL: FoundationOne<sup>®</sup> Liquid CDx; N: NCC Oncopanel System; T: GenMine™ TOP Cancer Panel. For treatment regimens, unmarked cases indicate adriamycin-based therapy while “N” indicates non-adriamycin regimens (including eribulin, pazopanib, and docetaxel). Y marks indicate patients aged 40 years or younger at diagnosis.</p>
Full article ">Figure 1 Cont.
<p>Genomic alterations and treatment responses of phyllodes tumor cases registered in the Center for Cancer Genomics and Advanced Therapeutics. Notable germline findings include MSH6 (T719fs*17, AF = 0.28) and TP53 (G245S, AF = 0.77) alterations, both classified as presumed germline pathogenic variants (PGPVs) according to Japanese PGPV criteria (Kosugi Group Criteria Ver.4; AF cutoff &gt;0.2 for truncating variants in MSS tumors and &gt;0.3 for TP53 variants in patients ≤30 years). TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated; F: FoundationOne<sup>®</sup> CDx; FL: FoundationOne<sup>®</sup> Liquid CDx; N: NCC Oncopanel System; T: GenMine™ TOP Cancer Panel. For treatment regimens, unmarked cases indicate adriamycin-based therapy while “N” indicates non-adriamycin regimens (including eribulin, pazopanib, and docetaxel). Y marks indicate patients aged 40 years or younger at diagnosis.</p>
Full article ">
15 pages, 4067 KiB  
Article
p21Waf1/Cip1 Is a Novel Downstream Target of 40S Ribosomal S6 Kinase 2
by Alakananda Basu and Zhenyu Xuan
Cancers 2024, 16(22), 3783; https://doi.org/10.3390/cancers16223783 - 10 Nov 2024
Viewed by 621
Abstract
Background/Objectives: The ribosomal S6 kinase 2 (S6K2) acts downstream of the mechanistic target of rapamycin complex 1 and is a homolog of S6K1 but little is known about its downstream effectors. The objective of this study was to use an unbiased transcriptome [...] Read more.
Background/Objectives: The ribosomal S6 kinase 2 (S6K2) acts downstream of the mechanistic target of rapamycin complex 1 and is a homolog of S6K1 but little is known about its downstream effectors. The objective of this study was to use an unbiased transcriptome profiling to uncover how S6K2 promotes breast cancer cell survival. Methods: RNA-Seq analysis was performed to identify novel S6K2 targets. Cells were transfected with siRNAs or plasmids containing genes of interest. Western blot analyses were performed to quantify total and phosphorylated proteins. Apoptosis was monitored by treating cells with different concentrations of doxorubicin. Results: Silencing of S6K2, but not S6K1, decreased p21 in MCF-7 and T47D breast cancer cells. Knockdown of Akt1 but not Akt2 decreased p21 in MCF-7 cells whereas both Akt1 and Akt2 knockdown attenuated p21 in T47D cells. While Akt1 overexpression enhanced p21 and partially reversed the effect of S6K2 deficiency on p21 downregulation in MCF-7 cells, it had little effect in T47D cells. S6K2 knockdown increased JUN mRNA and knockdown of cJun enhanced p21. Low concentrations of doxorubicin increased, and high concentrations decreased p21 levels in T47D cells. Silencing of S6K2 or p21 sensitized T47D cells to doxorubicin via c-Jun N-terminal kinase (JNK)-mediated downregulation of Mcl-1. Conclusions: S6K2 knockdown enhanced doxorubicin-induced apoptosis by downregulating the cell cycle inhibitor p21 and the anti-apoptotic protein Mcl-1 via Akt and/or JNK. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>RNA-Seq analysis. (<b>A</b>). Venn graph of DEGs detected using both Cuffdiff and DESeq following S6K2 KD. FDR &lt; 0.05 was applied for each method. (<b>B</b>). The representative gene ontology terms of functional annotation clusters, which are significantly enriched in 118 shared DEGs (FDR &lt; 0.05). (<b>C</b>). Densitometric quantification of <span class="html-italic">CDKN1A</span> mRNA normalized with GAPDH control. The asterisk (*) indicates a significant difference from control siRNA-transfected cells (<span class="html-italic">p</span> &lt; 0.05) using paired Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 2
<p>T47D (<b>A</b>,<b>B</b>) or MCF-7 (<b>C</b>,<b>D</b>) cells were transfected with or without control non-targeting siRNA or SMARTpool (SP) S6K1 or S6K2 siRNA. Western blot analyses were performed with indicated antibodies. The intensity of p21 was determined using ImageJ and normalized with respect to loading control. Each bar represents mean ± S.E. <span class="html-italic">p</span> values were calculated using a paired Student’s <span class="html-italic">t</span> test. (<b>E</b>). Different concentrations of cell lysates from MCF-7 cells transfected with an empty vector pcDNA3 (PC) or a vector containing S6K2 construct were subjected to Western blot analyses with indicated antibodies.</p>
Full article ">Figure 3
<p>T47D (<b>A</b>,<b>B</b>) or MCF-7 (<b>C</b>,<b>D</b>) cells were transfected with indicated siRNAs and Western blot analyses were performed with indicated antibodies. Each bar represents the mean ± S.E of four independent experiments. <span class="html-italic">p</span> values were calculated using paired Student’s <span class="html-italic">t</span> test of control versus individual siRNA as described under <a href="#cancers-16-03783-f002" class="html-fig">Figure 2</a>. ***, <span class="html-italic">p</span> ≤ 0.0005; **, <span class="html-italic">p</span> ≤ 0.005; *, <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 4
<p>T47D (<b>A</b>,<b>B</b>) or MCF-7 (<b>C</b>,<b>D</b>) cells were transfected with indicated siRNAs and Western blot analyses were performed with indicated antibodies. Each bar represents mean ± S.E of at least six independent experiments. <span class="html-italic">p</span> values were calculated using paired Student’s <span class="html-italic">t</span> test.</p>
Full article ">Figure 5
<p>T47D (<b>A</b>,<b>B</b>) or MCF-7 (<b>C</b>,<b>D</b>) cells were transfected with control or S6K2 siRNA and then infected with or without adenoviral vectors containing Akt1. Western blot analyses were performed with indicated antibodies. Each bar represents mean ± S.E of six independent experiments. <span class="html-italic">p</span> values calculated using paired <span class="html-italic">t</span> test of control versus Akt1 overexpressing cells: T47D, <span class="html-italic">p</span> = 0.0007; MCF-7, <span class="html-italic">p</span> = 0.0005; Light gray bar, control siRNA; black bar, S6K2 siRNA.</p>
Full article ">Figure 6
<p>T47D cells were transfected with indicated siRNAs. Western blot analyses were performed with indicated antibodies (<b>A</b>,<b>C</b>,<b>E</b>). The intensities of cJun (<b>B</b>) and p21 (<b>D</b>) were determined using ImageJ and normalized with respect to loading controls. Each bar represents mean ± S.E. <span class="html-italic">p</span> values were calculated using paired Student’s <span class="html-italic">t</span> test.</p>
Full article ">Figure 7
<p>T47D cells were transfected with control non-targeting siRNA or S6K2 siRNA and then treated with indicated concentrations of doxorubicin (Dox). Western blot analyses were performed with indicated antibodies. The band corresponding to cleaved caspase-7 was quantified using ImageJ and the intensities of bands normalized with loading controls are shown.</p>
Full article ">Figure 8
<p>T47D cells were transfected with control non-targeting siRNA or p21 siRNA and then treated with indicated concentrations of doxorubicin. Western blot analyses were performed with indicated antibodies. The bands corresponding to p21, cleaved caspase-3, caspase-7, and PARP were quantified using ImageJ, and the intensities of bands normalized with loading controls are shown.</p>
Full article ">Figure 9
<p>T47D cells were transfected with control non-targeting siRNA, S6K2 and/or c-Jun siRNA and then treated with or without 0.3 and 1.0 µM (<b>A</b>) or 10 µM (<b>B</b>) doxorubicin. Western blot analysis was performed with indicated antibodies. The band corresponding to cleaved caspase-7 or PARP was quantified using ImageJ and the intensities of bands were normalized with tubulin.</p>
Full article ">Figure 10
<p>T47D cells were transfected with control non-targeting siRNA or JNK1 siRNA and then treated with indicated concentrations of doxorubicin. Western blot analyses were performed with indicated antibodies.</p>
Full article ">
22 pages, 1438 KiB  
Article
Association of Genetic Variants at the CDKN1B and CCND2 Loci Encoding p27Kip1 and Cyclin D2 Cell Cycle Regulators with Susceptibility and Clinical Course of Chronic Lymphocytic Leukemia
by Lidia Ciszak, Agata Kosmaczewska, Edyta Pawlak, Irena Frydecka, Aleksandra Szteblich and Dariusz Wołowiec
Int. J. Mol. Sci. 2024, 25(21), 11705; https://doi.org/10.3390/ijms252111705 - 31 Oct 2024
Viewed by 681
Abstract
Beyond the essential role of p27Kip1 and cyclin D2 in cell cycle progression, they are also shown to confer an anti-apoptotic function in peripheral blood (PB) lymphocytes. Although the aberrant longevity and expression of p27Kip1 and cyclin D2 in leukemic cells [...] Read more.
Beyond the essential role of p27Kip1 and cyclin D2 in cell cycle progression, they are also shown to confer an anti-apoptotic function in peripheral blood (PB) lymphocytes. Although the aberrant longevity and expression of p27Kip1 and cyclin D2 in leukemic cells is well documented, the exact mechanisms responsible for this phenomenon have yet to be elucidated. This study was undertaken to determine the associations between polymorphisms in the CDKN1B and CCND2 genes (encoding p27Kip1 and cyclin D2, respectively) and susceptibility to chronic lymphocytic leukemia (CLL), as well as their influence on the expression of both cell cycle regulators in PB leukemic B cells and non-malignant T cells from untreated CLL patients divided according to the genetic determinants studied. Three CDKN1B single-nucleotide polymorphisms (SNPs), rs36228499, rs34330, and rs2066827, and three CCND2 SNPs, rs3217933, rs3217901, and rs3217810, were genotyped using a real-time PCR system. The expression of p27Kip1 and cyclin D2 proteins in both leukemic B cells and non-malignant T cells was determined using flow cytometry. We found that the rs36228499A and rs34330T alleles in CDKN1B and the rs3217810T allele in the CCND2 gene were more frequent in patients and were associated with increased CLL risk. Moreover, we observed that patients possessing the CCND2rs3217901G allele had lower susceptibility to CLL (most pronounced in the AG genotype). We also noticed that the presence of the CDKN1Brs36228499CC, CDKN1Brs34330CC, CDKN1Brs2066827TT, and CCND2rs3217901AG genotypes shortened the time to CLL progression. Statistically significant functional relationships were limited to T cells and assigned to CDKN1B polymorphic variants; carriers of the polymorphisms rs34330CC and rs36228499CC (determining the aggressive course of CLL) expressed a decrease in p27Kip1 and cyclin D2 levels, respectively. We indicate for the first time that genetic variants at the CDKN1B and CCND2 loci may be considered as a potentially low-penetrating risk factor for CLL and determining the clinical outcome. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>Cumulative probability of progression to a higher Rai stage, lymph node and organ progression, and treatment-free survival in CLL patients stratified according to the <span class="html-italic">CDKN1B</span> and <span class="html-italic">CCND2</span> gene polymorphisms. (<b>a</b>–<b>c</b>): Cumulative probability of progression to a higher Rai stage-free survival in CLL patients divided according to the genetic variants of the <span class="html-italic">CDKN1B</span>rs36228499 (<b>a</b>) and <span class="html-italic">CDKN1B</span>rs2066827 (<b>b</b>) polymorphic sites as well as the <span class="html-italic">CCND2</span>rs3217901 polymorphic locus (<b>c</b>). (<b>d</b>,<b>e</b>): Cumulative probability of lymph node-free survival in CLL patients stratified according to the genetic variants of the <span class="html-italic">CDKN1B</span>rs34330 (<b>d</b>) and <span class="html-italic">CCND2</span>rs3217810 (<b>e</b>) polymorphic sites. (<b>f</b>): Cumulative probability of treatment-free survival in CLL patients divided according to the genetic variants of the <span class="html-italic">CCND2</span>rs3217810 polymorphic locus. The <span class="html-italic">p</span>-value was obtained using the log rank test.</p>
Full article ">Figure 2
<p>Association between genetic variants of the <span class="html-italic">CDKN1B</span>rs36228499 polymorphic locus and cyclin D2 expression in CLL patients. (<b>a</b>,<b>d</b>) The graphs show the mean fluorescence intensity (MFI) of cyclin D2 protein in PB CD19+CD5+ (<b>a</b>) and CD3+ (<b>d</b>) cells in A− and A+ carriers of the <span class="html-italic">CDKN1B</span>rs36228499 polymorphic site. The horizontal lines represent the median values. Differences between studied groups were evaluated using the Mann–Whitney U test. (**) signifies a statistically significant difference <span class="html-italic">p</span> &lt; 0.01. (<b>b</b>,<b>c</b>,<b>e</b>,<b>f</b>): Cytometric analysis of cyclin D2 protein expression in CLL patients divided according to the genetic variants of the <span class="html-italic">CDKN1B</span>rs36228499 polymorphic locus. Histograms show cytometric analysis of cyclin D2 expression in PB CD19+CD5+ (<b>b</b>,<b>c</b>) and CD3+ (<b>e</b>,<b>f</b>) cells co-expressing cyclin D2 protein in A− and A+ carriers. PBMCs were gated using FSC/SSC profiles followed by gating on CD19+CD5+ (<b>b</b>,<b>c</b>) or CD3+ (<b>e</b>,<b>f</b>) to identify CD19+CD5+ and CD3+ cells for further analysis of cyclin D2 protein expression in PB CD19+CD5+ and CD3+ cells. Black line curves show cyclin D2-fluorescence of cells within PB CD19+CD5+ and CD3+ cells. Gray areas represent the isotype controls. The numbers located on the histograms represent the cyclin D2-dependent signal intensity (MFI).</p>
Full article ">
14 pages, 2545 KiB  
Article
Investigating the p21 Ubiquitin-Independent Degron Reveals a Dual Degron Module Regulating p21 Degradation and Function
by Marianna Riutin, Pnina Erez, Julia Adler, Assaf Biran, Nadav Myers and Yosef Shaul
Cells 2024, 13(19), 1670; https://doi.org/10.3390/cells13191670 - 9 Oct 2024
Viewed by 882
Abstract
A group of intrinsically disordered proteins (IDPs) are subject to 20S proteasomal degradation in a ubiquitin-independent manner. Recently, we have reported that many IDPs/IDRs are targeted to the 20S proteasome via interaction with the C-terminus of the PSMA3 subunit, termed the PSMA3 Trapper. [...] Read more.
A group of intrinsically disordered proteins (IDPs) are subject to 20S proteasomal degradation in a ubiquitin-independent manner. Recently, we have reported that many IDPs/IDRs are targeted to the 20S proteasome via interaction with the C-terminus of the PSMA3 subunit, termed the PSMA3 Trapper. In this study, we investigated the biological significance of the IDP–Trapper interaction using the IDP p21. Using a split luciferase reporter assay and conducting detailed p21 mutagenesis, we first identified the p21 RRLIF box, localized at the C-terminus, as mediating the Trapper interaction in cells. To demonstrate the role of this box in p21 degradation, we edited the genome of HEK293 and HeLa cell lines using a CRISPR strategy. We found that the p21 half-life increased in cells with either a deleted or mutated p21 RRLIF box. The edited cell lines displayed an aberrant cell cycle pattern under normal conditions and in response to DNA damage. Remarkably, these cells highly expressed senescence hallmark genes in response to DNA damage, highlighting that the increased p21 half-life, not its actual level, regulates senescence. Our findings suggest that the p21 RRLIF box, which mediates interactions with the PSMA3 Trapper, acts as a ubiquitin-independent degron. This degron is positioned adjacent to the previously identified ubiquitin-dependent degron, forming a dual degron module that functionally regulates p21 degradation and its physiological outcomes. Full article
Show Figures

Figure 1

Figure 1
<p><b>p21 interacts with the PSMA3 Trapper.</b> (<b>A</b>) Schematic presentation of the proposed model whereby IDP/IDR interacts with the PSMA3 (A3) trapper as a prerequisite step for degradation by the 20S proteasome particle. (<b>B</b>) Illustration of the luciferase complementation assay system demonstrating the direct interaction between the PSMA3 Trapper and the p21 protein, both fused to luciferase split fragments. Upon interaction, a bioluminescent signal is detected. (<b>C</b>) The boxplot represents the obtained bioluminescent signals, calculated as the fold increase over the signal of the control plasmids (number of repeats N = 10). (<b>D</b>) SDS–PAGE and immunoblot analysis of the overexpressed proteins tested in panel (<b>C</b>). Ponceau staining was used as a loading control.</p>
Full article ">Figure 2
<p><b>The C-terminal segment of p21 is the most potent Trapper binding region.</b> (<b>A</b>) Peptide array analysis of p21 to assess which regions preferably bind the Trapper. p21 was divided into 15 amino acid–long peptides with an overlap of 5 amino acids. From left to right, each square indicates a different peptide from the N-terminus to the C-terminus of the protein. Hits that imply an interaction with the recombinant Trapper appear as darker spots. Colored frames refer to the regions that are further tested. A simplified illustration of p21 protein positive regions labeled as N, M and C. (<b>B</b>) The boxplot represents the obtained bioluminescent signals measured per pair of overexpressed positive p21 regions in binding either the Trapper or the full-length PSMA3 (N = 5). (<b>C</b>) As in panel (<b>B</b>), but truncated p21 C region deleted mutant (2–131) and full-length p21 (2–164) were tested (N = 5). (<b>D</b>) Bioluminescent signals correlating to an interaction of full-length p21, separated p21 peptides, or deletion versions of p21 with either PSMA3 (A3) or the Trapper (Tr). Error bars refer to 3 technical repetitions. Illustrated on the left are the corresponding p21 mutants (N = 3). (<b>E</b>) SDS-PAGE and immunoblot analysis of the overexpressed proteins that were tested for binding in panel (<b>D</b>). Ponceau staining was used as a loading control. (<b>F</b>) The AlphaFold predicted structure of p21, and the regions of interest are shown.</p>
Full article ">Figure 3
<p><b>Identification of the p21 C region sequence in mediating Trapper binding.</b> (<b>A</b>) Sub-fragments of the p21 C region, numbered from 1 to 7, were used for further analysis. (<b>B</b>) The fold increase in luciferase activity over the control, resulting from the transfection of the C fragment presented in panel (<b>A</b>), along with the Trapper construct, is shown (N = 3). (<b>C</b>) Similar to panel (<b>B</b>), but using the full-length PSMA3 instead of the Trapper construct (N = 3). (<b>D</b>) The RRLIF sequence within the C fragment is compared to that corresponding sequence in the N fragment. (<b>E</b>) In the context of the p21 131–164 aa C fragment, the RRLIF motif was mutated to RRLAF and subjected to the split luciferase assay with both Trapper and the PSMA3 (N = 3). (<b>F</b>) The expression levels of the plasmids used in panel (<b>E</b>) are shown, with Ponceau staining serving as a loading control. (<b>G</b>) The RRLIF box in the full-length p21 was mutated to RRLAF and analyzed with both Trapper and the PSMA3 (N = 3). (<b>H</b>) The expression level of the plasmids used in panel (<b>G</b>), with Ponceau staining as a loading control.</p>
Full article ">Figure 4
<p><b>Analysis of p21 edited HEK293 cells</b>. (<b>A</b>) Protein sequence of wild type p21 and the edited p21 in HEK293 cells using CRISPR technology. Two alleles were edited differently: p21<sup>156t</sup> is a truncated mutant missing the last 9 amino acids of the C-terminus, including the RLIF box, while p21<sup>158+43</sup> is a frameshift mutant where the C-terminus 9 amino acids are replaced by a new reading frame of 43 amino acids. The red letters in the sequence represent the wild-type p21 sequence. (<b>B</b>) p21 expression in the edited cells was analyzed in the absence or presence of MG132 for 4 h, a proteasome inhibitor. The asterisk (*) indicates the position of wild-type p21. The band labeled with a question mark (?) represents an unidentified one. (<b>C</b>) p21 protein decay was assessed using cycloheximide (CHX), a translation inhibitor, for the indicated time points. (<b>D</b>) Decay of p21 in the edited HEK293 clone, as described in panel (<b>A</b>), was examined using CHX treatment as outlined in panel. The band labeled with a question mark (?) represents an unidentified one. (<b>C</b>). (<b>E</b>) Comparison of relative growth between control HEK293 cells and the HEK293 p21-edited clone. (<b>F</b>) Cell cycle distribution of the cells was analyzed using FACS.</p>
Full article ">Figure 5
<p><b>Analysis of p21-edited HeLa cells.</b> (<b>A</b>) The panel displays the protein sequence of the wild-type p21 and the edited p21 in HeLa p21 cells (p21 5A), where the RRLIF box was mutated to AAAAA using CRISPR technology. (<b>B</b>) The HeLa cells and p21-5A mutant were treated with bortezomib (250 nM) for 18 h. After removing bortezomib, cells were allowed to recover for 1 h, then treated with 0.5 mM cycloheximide (CHX). Cells were harvested at specified times and analyzed by Western blotting to assess the levels of p21 and p21 5A over the CHX treatment time course. The band labeled with a question mark (p21?) represents an unidentified one. (<b>C</b>) The graph shows the degradation rate of different forms of p21 over time. The <span class="html-italic">y</span>-axis represents the log percentage of the remaining p21, and the <span class="html-italic">x</span>-axis indicates time in hours (h). Error bars reflect variability of data points at each time interval, and the R<sup>2</sup> values were calculated. (<b>D</b>) Cells were irradiated (10 Gy) and the fractions of the G0/G1 and G2/M phases were quantified after 24 h. Error bars represent standard deviation or standard error of the mean. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p><b>The stable p21 mutants induce the expression of senescence-associated genes.</b> (<b>A</b>–<b>F</b>) the mRNA levels of several genes in wild-type and p21edited HEK293. (<b>G</b>–<b>L</b>) mRNA levels of the same set of genes in wild-type HeLa and p21–5A HeLa cells. Cells were either untreated or exposed to 10 Gy (X-ray) radiation. Error bars indicate the standard deviation or standard error of the mean. * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>The list of proteins containing the RRLIF box and the p21 double degron composition. (<b>A</b>) Sequence alignment of proteins containing the ubiquitin-independent RRLIF degron. For each protein, the p21 sequence similarity is shown in red. (<b>B</b>) The sequence of the p21 C-terminal region, showing the functional motifs and highlighting the structure of the double degron. The potential modified residues described in PhosphoSitePlus<sup>®</sup> are shown in red and marked as follows: <b>p</b> for phosphorylation sites, <b>u</b> for ubiquitination sites, <b>a</b> for acetylation sites, and <b>m2</b> for methylation sites.</p>
Full article ">
12 pages, 449 KiB  
Article
Heterogeneous Transcriptional Landscapes in Human Sporadic Parathyroid Gland Tumors
by Chiara Verdelli, Silvia Carrara, Riccardo Maggiore, Paolo Dalino Ciaramella and Sabrina Corbetta
Int. J. Mol. Sci. 2024, 25(19), 10782; https://doi.org/10.3390/ijms251910782 - 7 Oct 2024
Viewed by 866
Abstract
The expression of several key molecules is altered in parathyroid tumors due to gene mutations, the loss of heterozygosity, and aberrant gene promoter methylation. A set of genes involved in parathyroid tumorigenesis has been investigated in sporadic parathyroid adenomas (PAds). Thirty-two fresh PAd [...] Read more.
The expression of several key molecules is altered in parathyroid tumors due to gene mutations, the loss of heterozygosity, and aberrant gene promoter methylation. A set of genes involved in parathyroid tumorigenesis has been investigated in sporadic parathyroid adenomas (PAds). Thirty-two fresh PAd tissue samples surgically removed from patients with primary hyperparathyroidism (PHPT) were collected and profiled for gene, microRNA, and lncRNA expression (n = 27). Based on a gene set including MEN1, CDC73, GCM2, CASR, VDR, CCND1, and CDKN1B, the transcriptomic profiles were analyzed using a cluster analysis. The expression levels of CDC73 and CDKN1B were the main drivers for clusterization. The samples were separated into two main clusters, C1 and C2, with the latter including two subgroups of five PAds (C2A) and nineteen PAds (C2B), both differing from C1 in terms of their lower expression of CDC73 and CDKN1B. The C2A PAd profile was also associated with the loss of TP73, an increased expression of HAR1B, HOXA-AS2, and HOXA-AS3 lncRNAs, and a trend towards more severe PHPT compared to C1 and C2B PAds. C2B PAds were characterized by a general downregulated gene expression. Moreover, CCND1 levels were also reduced as well as the expression of the lncRNAs NEAT1 and VLDLR-AS1. Of note, the deregulated lncRNAs are predicted to interact with the histones H3K4 and H3K27. Patients harboring C2B PAds had lower ionized and total serum calcium levels, lower PTH levels, and smaller tumor sizes than patients harboring C2A PAds. In conclusion, PAds display heterogeneous transcriptomic profiles which may contribute to the modulation of clinical and biochemical features. The general downregulated gene expression, characterizing a subgroup of PAds, suggests the tumor cells behave as quiescent resting cells, while the severity of PHPT may be associated with the loss of p73 and the lncRNA-mediated deregulation of histones. Full article
(This article belongs to the Special Issue State-of-the-Art Molecular Endocrinology and Metabolism in Italy)
Show Figures

Figure 1

Figure 1
<p>Hierarchical cluster analysis of analyzed PAd series based on the expression of the genes known to be pathogenic in parathyroid tumorigenesis. (<b>a</b>) Dendrogram identifying 3 clusters; distance, also known as dissimilarity, is represented on the vertical scale. (<b>b</b>) Heatmap representing the expression levels of each analyzed gene in each PAd sample.</p>
Full article ">
13 pages, 3017 KiB  
Article
Platycladus orientalis Leaf Extract Promotes Hair Growth via Non-Receptor Tyrosine Kinase ACK1 Activation
by Jaeyoon Kim, Jang Ho Joo, Juhyun Kim, Heena Rim, Jae young Shin, Yun-Ho Choi, Kyoungin Min, So Young Lee, Seung-Hyun Jun and Nae-Gyu Kang
Curr. Issues Mol. Biol. 2024, 46(10), 11207-11219; https://doi.org/10.3390/cimb46100665 - 5 Oct 2024
Viewed by 1096
Abstract
Platycladus orientalis is a traditional oriental herbal medicinal plant that is widely used as a component of complex prescriptions for alopecia treatment in Eastern Asia. The effect of PO on hair growth and its underlying mechanism, however, have not been demonstrated or clarified. [...] Read more.
Platycladus orientalis is a traditional oriental herbal medicinal plant that is widely used as a component of complex prescriptions for alopecia treatment in Eastern Asia. The effect of PO on hair growth and its underlying mechanism, however, have not been demonstrated or clarified. In this study, we investigated the hair-growth-promoting effect of PO in cultured human dermal papilla cells (hDPCs). Platycladus orientalis leaf extract (POLE) was found to stimulate the proliferation of hDPCs. POLE with higher quercitrin concentration, especially, showed a high level of cellular viability. In the context of cellular senescence, POLE decreased the expression of p16 (CDKN2A) and p21(CDKN1A), which resulted in enhanced proliferation. In addition, growth factor receptors, FGFR1 and VEGFR2/3, and non-receptor tyrosine kinases, ACK1 and HCK, were significantly activated. In addition, LEF1, a transcription factor of Wnt/β-catenin signaling, was enhanced, but DKK1, an inhibitor of Wnt/β-catenin signaling, was downregulated by POLE treatment in cultured hDPCs. As a consequence, the expression of growth factors such as bFGF, KGF, and VEGF were also increased by POLE. We further investigated the hair-growth-promoting effect of topically administered POLE over a 12-week period. Our data suggest that POLE could support terminal hair growth by stimulating proliferation of DPCs and that enhanced production of growth factors, especially KGF, occurred as a result of tyrosine kinase ACK1 activation. Full article
Show Figures

Figure 1

Figure 1
<p>POLE enhanced cell viability in cultured hDPCs. (<b>a</b>) chemical structure of Quercitrin (Quercitrin-3-Rhamnoside (<b>b</b>) Cell viability was assessed using CCK-8 assay kit after POLE treatment (0.25, 0.5, 1, and 2%) for 24 h. (<b>c</b>) Quercitrin of POLE was examined following the lots number. (<b>d</b>) Comparison of cellular viability between POLE extracts following quercitrin concentration. N.T, non-treated control; MNX, minoxidil. Significantly different compared with N.T (* <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 2
<p>POLE modulated the mRNA and protein expression of cell proliferation and senescence in hDPCs. (<b>a</b>) The mRNA expression levels of cell cycle arresters (CDKN1A and CDKN2A) were evaluated by RT-PCR. (<b>b</b>) The protein levels of p21, p27 and Survivin were significantly changed by POLE treatment. Minoxidil was used as a positive control. MNX—minoxidil; N.T—non-treated control; significantly different compared with N.T (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>Activation of receptor Tyr kinase and Tyr kinase by POLE treatment. The DPCs were treated with or without POLE (0.25, 0.5 and 1%) for 24 h, and then collected. (<b>a</b>) The phosphorylation of three growth factor receptors (FGFR1, VEGFR2, and VEFR3) were increased. (<b>b</b>) In addition, activation of five Tyr kinases (ACK1, HCK, SRMS, FYN and MATK) were quantitated. Significantly different compared with N.T (* <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). N.T, non-treated control. (<b>c</b>) Schematic of signaling cascade for interaction between Tyr kinases and receptor Tyr kinases. (<b>d</b>) Using anti-phosphor-Tyr antibody, the relative phosphorylation of five Tyr kinases was evaluated. Significantly different compared with p-ACK-1 (**** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>POLE regulated Wnt/β-catenin Pathway Target Gene in cultured hDPCs. The target genes of β-catenin were evaluated by RT-PCR after treatment with adenosine for 24 h. The mRNA expression level of (<b>a</b>) LEF1 was increased and (<b>b</b>) DKK-1 was decreased by POLE treatment. The data represent the means of five independent samples. NT, non-treated control; Significantly different compared with N.T (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>Effect of POLE on mRNA expression levels of growth factor genes in cultured hDPCs. The cells were harvested after POLE treatment (0.25, 0.5, and 1%) for 24h. The mRNA expression levels of (<b>a</b>) FGF7 and (<b>b</b>) IGF-1 gene in cultured hDPCs were measured by real-time PCR. N.T, non-treated control; MNX, minoxidil. Significantly different compared with N.T (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Increase of growth factor levels by POLE treatment in cultured hDPCs. The DPCs were treated with POLE (0.25, 0.5 and 1%) for 24 h, and then collected. Cells cultured with vehicle medium were used as non-treated control. The 9 types of growth factors and receptors were (<b>a</b>) displayed and (<b>b</b>) quantitated. N.T, non-treated control. Positive, biotin-conjugated IgG. Significantly different compared with N.T (* <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>Terminal hair density changes by POLE treatment. The change in terminal hair density with the administration of (<b>a</b>) POLE and (<b>b</b>) Placebo were evaluated. Significantly different compared with the baseline (** <span class="html-italic">p</span> &lt; 0.01, ns: not significant).</p>
Full article ">
16 pages, 1322 KiB  
Article
Expression of HMGB1, TGF-β1, BIRC3, ADAM17, CDKN1A, and FTO in Relation to Left Ventricular Remodeling in Patients Six Months after the First Myocardial Infarction: A Prospective Study
by Jovana Kuveljic, Ana Djordjevic, Ivan Zivotic, Milica Dekleva, Ana Kolakovic, Maja Zivkovic, Aleksandra Stankovic and Tamara Djuric
Genes 2024, 15(10), 1296; https://doi.org/10.3390/genes15101296 - 2 Oct 2024
Cited by 1 | Viewed by 948
Abstract
Background: After myocardial infarction (MI), adverse left ventricular (LV) remodeling may occur. This is followed by LV hypertrophy and eventually heart failure. The remodeling process is complex and goes through multiple phases. The aim of this study was to investigate the expression of [...] Read more.
Background: After myocardial infarction (MI), adverse left ventricular (LV) remodeling may occur. This is followed by LV hypertrophy and eventually heart failure. The remodeling process is complex and goes through multiple phases. The aim of this study was to investigate the expression of HMGB1, TGF-β1, BIRC3, ADAM17, CDKN1A, and FTO, each involved in a specific step of LV remodeling, in association with the change in the echocardiographic parameters of LV structure and function used to assess the LV remodeling process in the peripheral blood mononuclear cells (PBMCs) of patients six months after the first MI. The expression of selected genes was also determined in PBMCs of controls. Methods: The study group consisted of 99 MI patients, who were prospectively followed-up for 6 months, and 25 controls. Cardiac parameters, measured via conventional 2D echocardiography, were evaluated at two time points: 3–5 days and 6 months after MI. The mRNA expression six-months-post-MI was detected using TaqMan® technology (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA). Results:HMGB1 mRNA was significantly higher in patients with adverse LV remodeling six-months-post-MI than in patients without adverse LV remodeling (p = 0.04). HMGB1 mRNA was significantly upregulated in patients with dilated LV end-diastolic diameter (LVEDD) (p = 0.03); dilated LV end-diastolic volume index (LVEDVi) (p = 0.03); severely dilated LV end-systolic volume index (LVESVi) (p = 0.006); impaired LV ejection fraction (LVEF) (p = 0.01); and LV enlargement (p = 0.03). It was also significantly upregulated in PBMCs from patients compared to controls (p = 0.005). TGF-β1 and BIRC3 mRNA were significantly lower in patients compared to controls (p = 0.02 and p = 0.05, respectively). Conclusions: Our results suggest that HMGB1 is involved in adverse LV remodeling six-months-post-MI, even on the mRNA level. Further research and validation are needed. Full article
(This article belongs to the Special Issue Genetic and Genomic Research of Cardiovascular Diseases)
Show Figures

Figure 1

Figure 1
<p><b>Relative expression of <span class="html-italic">HMGB1</span>, <span class="html-italic">TGF-β1</span>, <span class="html-italic">BIRC3</span>, <span class="html-italic">ADAM17</span>, <span class="html-italic">CDKN1A</span>, and <span class="html-italic">FTO</span> in controls and MI patients’ PBMCs.</b> Relative mRNA expression is presented as 2<sup>−∆Ct</sup> value for each sample. The cDNAs from the PBMCs if controls (N = 24) and patients with MI (N = 95) were used to quantify gene expression. The difference between the Ct values of the reference gene, <span class="html-italic">Cyclophilin A</span>, and the gene of interest was used to compute the delta Ct value. Data are presented as 2<sup>−∆Ct</sup> as mean for both groups (controls—circles; patients—squares) ± SD. The difference of mRNAs relative expression between groups was calculated using the Mann–Whitney <span class="html-italic">U</span> test. (<b>A</b>) Significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in PBMCs from patients compared to controls (0.038 ± 0.015 vs. 0.028 ± 0.009, <span class="html-italic">p</span> = 0.005); (<b>B</b>) <span class="html-italic">TGF-β1</span> mRNA was significantly downregulated in PBMCs from patients compared to controls (0.510 ± 0.155 vs. 0.600 ± 0.168, <span class="html-italic">p</span> = 0.02); (<b>C</b>) <span class="html-italic">BIRC3</span> mRNA was significantly downregulated in PBMCs from patients compared to controls (0.014 ± 0.007 vs. 0.016 ± 0.007, <span class="html-italic">p</span> = 0.05); (<b>D</b>) <span class="html-italic">ADAM17</span> mRNA expression was not significantly different between patients and controls (0.009 ± 0.004 vs. 0.009 ± 0.005, <span class="html-italic">p</span> = 0.30); (<b>E</b>) <span class="html-italic">CDKN1A</span> mRNA expression did not differ significantly between patients and controls (0.028 ± 0.017 vs. 0.031 ± 0.017, <span class="html-italic">p</span> = 0.40); (<b>F</b>) there was no significant difference in <span class="html-italic">FTO</span> mRNA expression between patients and controls (0.005 ± 0.002 vs. 0.006 ± 0.003, <span class="html-italic">p</span> = 0.39). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p><b>Relative expression of <span class="html-italic">HMGB1</span> and <span class="html-italic">TGF-β1</span> in MI patients’ PBMCs, according to the occurrence of adverse LV remodeling six-months-post-MI, based on the &gt;20% increase in LVEDV:</b> (<b>A</b>) significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in patients’ PBMCs with adverse remodeling (N = 18) compared to patients without adverse remodeling (N = 77) (0.046 ± 0.018 vs. 0.036 ± 0.013, <span class="html-italic">p</span> = 0.04); (<b>B</b>) <span class="html-italic">TGF-β1</span> mRNA expression was not significantly different between patients with adverse remodeling and patients without adverse remodeling (0.565 ± 0.173 vs. 0.497 ± 0.148, <span class="html-italic">p</span> = 0.15); (<b>C</b>) <span class="html-italic">BIRC3</span> mRNA expression did not differ significantly between patients with adverse remodeling and patients without adverse remodeling (0.013 ± 0.005 vs. 0.014 ± 0.007, <span class="html-italic">p</span> = 0.48); (<b>D</b>) <span class="html-italic">ADAM17</span> mRNA expression did not differ significantly between the two patient groups (adverse LVR: 0.009 ± 0.002 vs. without adverse LVR: 0.009 ± 0.005, <span class="html-italic">p</span> = 0.15); (<b>E</b>) <span class="html-italic">CDKN1A</span> mRNA expression was not significantly different between patients with adverse remodeling and patients without adverse remodeling (0.031 ± 0.014 vs. 0.027 ± 0.017, <span class="html-italic">p</span> = 0.22); (<b>F</b>) <span class="html-italic">FTO</span> mRNA expression did not differ significantly between patients with adverse remodeling and patients without adverse remodeling (0.006 ± 0.003 vs. 0.005 ± 0.002, <span class="html-italic">p</span> = 0.14). * <span class="html-italic">p</span> &lt; 0.05. ns: no significance.</p>
Full article ">Figure 3
<p><b>Relative expression of <span class="html-italic">HMGB1</span> in the PBMCs of patients with MI with regard to the reference values of the following echocardiographic parameters: LVEDD, LVESD, LVEDVi, LVESVi, LVEF, and LVE measured six-months-post-MI.</b> Relative mRNA expression is presented as the 2<sup>−∆Ct</sup> value for each sample. The delta Ct value was calculated from the difference between the Ct value of the gene of interest and that of <span class="html-italic">Cyclophilin A</span>. Data are presented as mean 2<sup>−∆Ct</sup> ± SD for both groups (normal, reference—circles; dilated/impaired—squares). The Mann–Whitney <span class="html-italic">U</span> test was used to calculate the difference in mRNAs’ relative expression between groups. (<b>A</b>) Significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in PBMCs from patients with dilated LVEDD (N = 40) compared to the patients with normal reference LVEDD (N = 55) (0.042 ± 0.017 vs. 0.035 ± 0.012, <span class="html-italic">p</span> = 0.03); (<b>B</b>) <span class="html-italic">HMGB1</span> mRNA expression did not differ significantly between patients with dilated LVESD (N = 19) and patients with normal reference LVESD (N = 76) (0.044 ± 0.018 vs. 0.037 ± 0.013, <span class="html-italic">p</span> = 0.11); (<b>C</b>) significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in PBMCs from patients with (mildly, moderately, and severely) dilated LVEDVi (N = 12) compared to the patients with normal reference LVEDVi (N = 83) (0.047 ± 0.018 vs. 0.037 ± 0.014, <span class="html-italic">p</span> = 0.03); (<b>D</b>) significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in PBMCs from patients with severely dilated LVESVi (N = 19) compared to the patients with reference and mildly and moderately dilated LVESVi (N = 76) (0.045 ± 0.015 vs. 0.036 ± 0.014, <span class="html-italic">p</span> = 0.006); (<b>E</b>) significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in PBMCs from patients with borderline low, impaired, and severely impaired LVEF (N = 76) compared to the patients with normal reference LVEF (N = 19) (0.040 ± 0.014 vs. 0.032 ± 0.013, <span class="html-italic">p</span> = 0.01); (<b>F</b>) significant upregulation of <span class="html-italic">HMGB1</span> mRNA was detected in PBMCs from patients with LV enlargement (N = 37) compared to the patients without LV enlargement (N = 58) (0.043 ± 0.016 vs. 0.035 ± 0.012, <span class="html-italic">p</span> = 0.03). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. ns: no significance.</p>
Full article ">
11 pages, 2273 KiB  
Article
Biallelic Loss of 7q34 (TRB) and 9p21.3 (CDKN2A/2B) in Adult Ph-Negative Acute T-Lymphoblastic Leukemia
by Natalya Risinskaya, Abdulpatakh Abdulpatakhov, Yulia Chabaeva, Olga Aleshina, Maria Gladysheva, Elena Nikulina, Ivan Bolshakov, Anna Yushkova, Olga Dubova, Anastasia Vasileva, Tatiana Obukhova, Hunan Julhakyan, Nikolay Kapranov, Irina Galtseva, Sergey Kulikov, Andrey Sudarikov and Elena Parovichnikova
Int. J. Mol. Sci. 2024, 25(19), 10482; https://doi.org/10.3390/ijms251910482 - 29 Sep 2024
Viewed by 1118
Abstract
Tumor cells of acute lymphoblastic leukemia (ALL) may have various genetic abnormalities. Some of them lead to a complete loss of certain genes. Our aim was to reveal biallelic deletions of genes in Ph–negative T-ALL. Chromosomal microarray analysis (CMA) was performed for 47 [...] Read more.
Tumor cells of acute lymphoblastic leukemia (ALL) may have various genetic abnormalities. Some of them lead to a complete loss of certain genes. Our aim was to reveal biallelic deletions of genes in Ph–negative T-ALL. Chromosomal microarray analysis (CMA) was performed for 47 patients with de novo Ph–negative T-ALL, who received treatment according to RALL-2016m clinical protocol at the National Medical Research Center for Hematology (Moscow, Russia) from 2017 to 2023. Out of forty-seven patients, only three had normal molecular karyotype. The other 44 patients had multiple gains, losses, and copy neutral losses of heterozygosity. Biallelic losses were found in 14 patients (30%). In ten patients (21%), a biallelic deletion of 9p21.3 involved a different number of genes, however CDKN2A gene loss was noted in all ten cases. For seven patients (15%), a biallelic deletion of 7q34 was found, including two genes—PRSS1, PRSS2 located within the T-cell receptor beta (TRB) locus. A clonal rearrangement of the TRB gene was revealed in 6 out of 7 cases with 7q34 biallelic loss. Both biallelic deletions can be considered favorable prognostic factors, with an association with 9p21 being statistically significant (p = 0.01) and a trend for 7q34 (p = 0.12) being observed. Full article
(This article belongs to the Special Issue Hematological Malignancies: Molecular Mechanisms and Therapy)
Show Figures

Figure 1

Figure 1
<p>Molecular karyotype of patient # 27 with biallelic deletions of 9p21.3 (<b>a</b>) and 7q34 (<b>b</b>). Chromosomes 7 and 9 are highlighted in blue frames, and the deepred indicate the loci of the biallelic deletions. A standard cytogenetic study also revealed a translocation t(6;7)(q23;q34) in this patient, as shown in the <a href="#app1-ijms-25-10482" class="html-app">Supplementary Table S1</a>.</p>
Full article ">Figure 2
<p>Detail views of biallelic loss 7q34 and TRB clonality assay with monoclonal peaks for patients 15 (<b>a</b>) and 27 (<b>b</b>) and polyclonal Gaussian curves for patient 44 (<b>c</b>).</p>
Full article ">Figure 3
<p>Histogram of the distribution of <span class="html-italic">TRB</span> clonality in groups of patients with biallelic loss of 7q34, monoallelic loss of 7q34, and intact 7q34 locus. (<b>a</b>)—CMA results with a cutoff of 50 kb for losses, (<b>b</b>)—a cutoff of 1 kb for losses. Dark red color indicates biallelic <span class="html-italic">TRB</span> clonality, red indicates <span class="html-italic">TRB</span> clonality with one monoclonal PCR peak, green indicates polyclonal samples. On the <span class="html-italic">X</span>-axis, the groups are arranged in the following order: with biallelic 7q34 loss, monoallelic 7q34 loss, no 7q34 lesions.</p>
Full article ">Figure 4
<p>Diagrams of the distribution of the MRD status depending on the presence of loss 7q34 (<b>a</b>) or 9p23.1 (<b>b</b>). The number of patients and percentage are indicated on the bars. Green indicates MRD-negative patients, red indicates MRD-positive patients. Along the X axis the bars are arranged in the following order—without loss, with monoallelic loss, with biallelic loss.</p>
Full article ">Figure 5
<p>Overall survival curves of patients depending on the presence of 7q34 (<b>a</b>) and 9p21.3 (<b>b</b>) deletions. Patients with biallelic loss (red curve), monoallelic loss (blue curve) and patients without deletions (yellow curve) were analyzed. The <span class="html-italic">X</span>-axis indicates the time after the start of therapy (months).</p>
Full article ">Figure 6
<p>The location of biallelic deletions of 7q34 in the studied cohort of patients, relative to the <span class="html-italic">TRB</span> locus, is shown in red lines. This figure was adapted from <a href="https://www.ncbi.nlm.nih.gov/gene/6957" target="_blank">https://www.ncbi.nlm.nih.gov/gene/6957</a> (accessed on 19 August 2024).</p>
Full article ">
13 pages, 12191 KiB  
Article
MTAP and p16 IHC as Markers for CDKN2A/B Loss in Meningiomas
by Hanim I. Ozkizilkaya, Anjali Vinocha, Antonio Dono, Oluwaseun Basit Ogunbona, Gokce A. Toruner, Phyu P. Aung, Carlos Kamiya Matsuoka, Yoshua Esquenazi, Franco DeMonte and Leomar Y. Ballester
Cancers 2024, 16(19), 3299; https://doi.org/10.3390/cancers16193299 - 27 Sep 2024
Viewed by 893
Abstract
Background: Homozygous cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) loss is one of the parameters that support the designation of meningiomas as Central Nervous System (CNS) WHO grade 3 tumors. Evaluation of CDKN2A/B by sequencing or Fluorescence in situ hybridization (FISH) is costly and not [...] Read more.
Background: Homozygous cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) loss is one of the parameters that support the designation of meningiomas as Central Nervous System (CNS) WHO grade 3 tumors. Evaluation of CDKN2A/B by sequencing or Fluorescence in situ hybridization (FISH) is costly and not always readily accessible. An immunohistochemistry (IHC)-based marker for the evaluation of CDKN2A/B loss would provide faster results at a lower cost. Methods: This retrospective study included patients diagnosed with meningioma at our institution between 2016 and 2019. Archival tumor tissue was used for analysis. MTAP immunohistochemistry (IHC) was performed at various dilutions (1:1200, 1:400, 1:200, 1:100) using two different antibodies, and p16 IHC was conducted simultaneously. These analyses were carried out at two different institutions. To determine the sensitivity and specificity of MTAP and p16 as surrogate markers for CDKN2A/B loss, CDKN2A FISH was utilized as the gold standard. Results: Overall, 46/49 tumors showed strong MTAP staining (94%) at institution 1, and 44/49 (90%) showed either faint positive or positive results at institution 2. One grade 3 meningioma that demonstrated homozygous CDKN2A loss by FISH also showed loss of MTAP expression by IHC. One grade 2 meningioma showed regional CDKN2A loss by FISH and variable MTAP expression under different IHC conditions. MTAP expression evaluation was superior at a dilution of 1:100 with the Abnova Anti-MTAP Monoclonal antibody. Conclusions: P16 expression was variable and did not correlate with either MTAP expression or CDKN2A FISH results. MTAP IHC is a promising surrogate marker for the evaluation of CDKN2A status in meningiomas. Full article
Show Figures

Figure 1

Figure 1
<p>MTAP, p16, and FISH results for case #36. (<b>A</b>) H&amp;E-stained section. (<b>B</b>) MTAP expression with 1:200 dilution at institution 1. (<b>C</b>,<b>D</b>) Faint positive p16 expression, institution 1 and institution 2, respectively. (<b>E</b>) Retained <span class="html-italic">CDKN2A</span> with two green and two red signals. (magnification = 600X).</p>
Full article ">Figure 2
<p>MTAP, p16, and FISH results for a grade 3 meningioma with <span class="html-italic">CDKN2A</span> deletion (case #47). (<b>A</b>) H&amp;E-stained section. (<b>B</b>) Absence of MTAP expression (1:100 dilution). Tumor cells do not stain with MTAP, whereas macrophages show positive staining. (<b>C</b>) Absence of MTAP expression in tumor cells (1:100 dilution). (<b>D</b>) Absence of p16 expression in tumor cells (institution 2). (<b>E</b>) Faint staining with p16 (institution 1). (<b>F</b>) <span class="html-italic">CDKN2A</span> loss with the absence of red signals. (<b>A</b>–<b>E</b>) scale bars = 200 um, (<b>F</b>) magnification = 600X.</p>
Full article ">Figure 3
<p>MTAP, p16, and FISH results of #30. (<b>A</b>,<b>B</b>) Grade 2 meningioma showing focal MTAP expression at 1:200 and 1:100 dilutions (institution 1), respectively. (<b>C</b>) Positive p16 expression in tumor cells. (<b>D</b>) Absence of p16 expression in tumor cells. (<b>E</b>) Areas with retained <span class="html-italic">CDKN2A</span> with two red and two green signals. (<b>F</b>) Area with <span class="html-italic">CDKN2A</span> loss with the absence of red signals in most nuclei. (<b>A</b>–<b>D</b>) scale bars = 200 um. (<b>E</b>,<b>F</b>): magnification = 600X.</p>
Full article ">Figure 3 Cont.
<p>MTAP, p16, and FISH results of #30. (<b>A</b>,<b>B</b>) Grade 2 meningioma showing focal MTAP expression at 1:200 and 1:100 dilutions (institution 1), respectively. (<b>C</b>) Positive p16 expression in tumor cells. (<b>D</b>) Absence of p16 expression in tumor cells. (<b>E</b>) Areas with retained <span class="html-italic">CDKN2A</span> with two red and two green signals. (<b>F</b>) Area with <span class="html-italic">CDKN2A</span> loss with the absence of red signals in most nuclei. (<b>A</b>–<b>D</b>) scale bars = 200 um. (<b>E</b>,<b>F</b>): magnification = 600X.</p>
Full article ">Figure 4
<p>Analysis of MTAP protein expression for case #21 (grade 1 meningioma) at different dilutions. (<b>A</b>) MTAP expression at 1:100 dilution at institution 1. (<b>B</b>) MTAP expression at 1:200 dilution at institution 1. (<b>C</b>) MTAP expression at 1:1200 dilution at institution 2. Note strong expression in panels (<b>A</b>,<b>B</b>) and very faint staining in panel (<b>C</b>) Scale bars = 200 um.</p>
Full article ">Figure 5
<p>Analysis of MTAP protein expression for case #12 (grade 1 meningioma) at different dilutions. (<b>A</b>) MTAP expression with 1:100 dilution at institution 1. (<b>B</b>) MTAP expression with 1:200 dilution at institution 1. (<b>C</b>) MTAP expression with 1:1200 dilution at institution 2. Scale bars = 200 um.</p>
Full article ">Figure 6
<p>Vysis LSI <span class="html-italic">CDKN2A</span> (P16) (9p21)—Spectrum Orange)/D9Z1 (9p11q11)—CEP 9 Spectrum Green probes (Abbott Molecular, Abbott Park, IL, USA). (<b>A</b>) <span class="html-italic">CDKN2A</span> intact tumor with two red and two green signals. (<b>B</b>) Tumor with loss of red signals but presence of two green signals, indicating homozygous loss of <span class="html-italic">CDKN2A</span>. Magnification = 600X.</p>
Full article ">
21 pages, 58464 KiB  
Article
Injectable Hydrogel-Encapsulating Pickering Emulsion for Overcoming Lenvatinib-Resistant Hepatocellular Carcinoma via Cuproptosis Induction and Stemness Inhibition
by Xin Li, Chuanyu Tang, Hanjie Ye and Chihua Fang
Polymers 2024, 16(17), 2418; https://doi.org/10.3390/polym16172418 - 26 Aug 2024
Viewed by 1402
Abstract
Lenvatinib resistance (LenR) presents a significant challenge in hepatocellular carcinoma (HCC) treatment, leading to high cancer-related mortality rates globally. Unlike traditional chemotherapy resistance mechanisms, LenR in HCC is primarily driven by increased cancer cell stemness. Disulfiram, (DSF), functioning as a Cu ionophore, can [...] Read more.
Lenvatinib resistance (LenR) presents a significant challenge in hepatocellular carcinoma (HCC) treatment, leading to high cancer-related mortality rates globally. Unlike traditional chemotherapy resistance mechanisms, LenR in HCC is primarily driven by increased cancer cell stemness. Disulfiram, (DSF), functioning as a Cu ionophore, can coordinate with Cu2+ to overcome LenR in HCC by inhibiting cancer cell stemness and cuproptosis. However, DSF faces challenges due to its poor water solubility, while copper ions present issues related to systemic toxicity during widespread use. To address this, DSF and CuO nanoparticles (NPs) were co-encapsulated to form an oil-in-water Pickering emulsion (DSF@CuO), effectively elevating DSF and copper ion concentrations within the tumor microenvironment (TME). DSF@CuO was then combined with sodium alginate (SA) to form a DSF@CuO-SA solution, which gelatinizes in situ with Ca2+ in the TME to form a DSF@CuO Gel, enhancing Pickering emulsion stability and sustaining DSF and copper ion release. A DSF@CuO Gel exhibits enhanced stability and therapeutic efficacy compared to conventional administration methods. It effectively induces mitochondrial dysfunction and cuproptosis in LenR HCC cells by downregulating DLAT, LIAS, and CDKN2A, while upregulating FDX1. Furthermore, it suppresses cancer stemness pathways through activation of the JNK/p38 MAPK pathway and inhibition of the NF-κB and NOTCH signaling pathways. These findings suggest that DSF@CuO Gels are a promising therapeutic strategy for treating LenR HCC. In vivo and in vitro LenR HCC models demonstrated significant therapeutic efficacy. In conclusion, this novel approach underscores DSF@CuO Gel’s potential to overcome LenR in HCC, offering a novel approach to address this clinical challenge. Full article
(This article belongs to the Special Issue Advances in Natural Biodegradable Polymers)
Show Figures

Figure 1

Figure 1
<p>Characterization of injectable hydrogel DSF@CuO Gel. (<b>A</b>) Schematic diagram of DSF@CuO Gel synthesis. (<b>B</b>) TEM images of CuO NPs (Scale bar = 100 nm). (<b>C</b>) DLS was used to measure diameter of CuO NPs. (<b>D</b>) Images of DSF@CuO under confocal microscope (scale bar = 20 μm). (<b>E</b>,<b>F</b>) Photos of DSF@CuO Gel before and after addition of Ca<sup>2+</sup>. (<b>G</b>) SEM images of DSF@CuO Gel hydrogel (scale bar = 200 μm). (<b>H</b>) Zeta potential of CuO NPs, DSF@CuO, and DSF@CuO Gel. (<b>I</b>,<b>J</b>) Rheological analyses were conducted to investigate mechanical properties of DSF@CuO Gel. (<b>K</b>) Drug release profile of hydrogel at different pH levels. (<b>L</b>,<b>M</b>) Hemolysis assessment of DSF@CuO Gel with different concentrations of treatment group. Data are presented as mean ± SD and are representative of three independent experiments.</p>
Full article ">Figure 2
<p>Cytotoxicity of DSF@CuO in LenR HCC. (<b>A</b>,<b>B</b>) The IC50 for LenR Huh7 and LenR Hep 3B. (<b>C</b>–<b>E</b>) The CCK-8 assay used to assess the cytotoxic effects of different treatment groups on LenR Huh7, LenR Hep3B, and LO-2. (<b>F</b>,<b>G</b>) The impact of different treatment groups on cell cycle in LenR Huh7. (<b>H</b>,<b>I</b>) The impact of different treatment groups on cell clone formation experiment in LenR Huh7. (<b>J</b>,<b>K</b>) The impact of different treatment groups on apoptosis in LenR-Huh7. (<b>L</b>,<b>M</b>) The cytotoxic effects of different treatment groups on tumor spheroids in LenR-Huh7 (scale bar = 100 μm). (G1: corn oil, G2: DSF, G3: CuO NPs, G4: Len@CuO, and G5: DSF@CuO). Data are presented as mean ± SD and are representative of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>DSF@CuO induced the upregulation of ROS, LPO, and severe mitochondria damage, resulting in cuproptosis. (<b>A</b>) The impact of different treatment groups on copper ion transport. (<b>B</b>,<b>C</b>) FCM was used to analyze the expression of JC-1 in different treatment groups. (<b>D</b>,<b>E</b>) Flow cytometry was used to analyze the expression of LPO in different treatment groups. (<b>F</b>) Fluorescence microscopy is used to detect changes in ROS and JC-1 probe levels in different treatment groups. (<b>G</b>,<b>H</b>) WB was used to verify the protein expression of cuproptosis across different treatment groups. (<b>I</b>) Representative Bio-TEM images of WT-Huh7 cells and LenR HCC cells before and after treatment with DSF@CuO. (G1: corn oil, G2: DSF, G3: CuO NPs, G4: Len@CuO, and G5: DSF@CuO). Data are presented as mean ± SD and are representative of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>DSF@CuO could suppress cell stemness-related characteristics in LenR HCC. (<b>A</b>) The expression of CD44 and CD133 in WT-Huh7 and LenR-Huh7. (<b>B</b>) Tumor sphere-forming capacity in both LenR cell lines and the WT (Scale bar = 50 μm). (<b>C</b>,<b>D</b>) A wound-healing assay showing the migration of corn oil, DSF, CuO NPs, lenvatinib, and DSF@CuO-treated LenR Huh7 cells in 0, 24, and 48 h. (<b>E</b>,<b>F</b>) A transwell assay was used to evaluate the impact of different treatment groups on the invasion ability of LenR Huh7. (<b>G</b>,<b>H</b>) The impact of different treatment groups on tumor spheroid formation in LenR-Huh7 (scale bar = 50 μm). (<b>I</b>,<b>K</b>) Western blot analysis of EpCAM, SOX9, and CD24 protein levels in LenR Huh7 cells after incubation with different groups. (<b>J</b>,<b>L</b>) Western blot analysis of E-cadherin, N-cadherin, and Vimentin protein levels in LenR Huh7 cells after incubation with different groups. (G1: corn oil, G2: DSF, G3: CuO NPs, G4: Len@CuO, and G5: DSF@CuO). Data are presented as mean ± SD and are representative of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Mechanisms of DSF@CuO in inhibiting cell stemness of LenR HCC. (<b>A</b>,<b>B</b>) GO and KEGG enrichment analysis of DEGs in DSF+Cu<sup>2+</sup> group. (<b>C</b>) GSEA plots showing pathways upregulated and downregulated after DSF+Cu<sup>2+</sup> treatment. (<b>D</b>) GSVA plots showing pathways upregulated and downregulated after DSF treatment. (<b>E</b>,<b>F</b>) Western blot analysis of p-p38MAPK, p38MAPK, p-JNK MAPK, JNK MAPK, p-NF-Κb, NF-Κb, and NOTCH protein levels in LenR Huh7 cells after incubation with different groups. (G1: corn oil, G2: DSF, G3: CuO NPs, G4: Len@CuO, and G5: DSF@CuO). Data are presented as mean ± SD and are representative of three independent experiments. *** <span class="html-italic">p</span> &lt; 0.001, ns, not significant.</p>
Full article ">Figure 6
<p>DSF@CuO Gel could inhibit tumor growth in LenR mice models. (<b>A</b>) Schematic illustrating treatment procedure. (<b>B</b>) Quantitative evaluation of sustained release effects of DSF, DSF@CuO, and DSF@CuO Gel in mouse models (n = 3). (<b>C</b>) IVIS results for evaluating retention behavior of DSF, DSF@CuO, and DSF@CuO Gel hydrogel after intratumor injection. (<b>D</b>) Photographs of dissected mouse tumor tissues after completion of treatment (n = 5). (<b>E</b>) In vivo tumor volume curve of each group of mice during treatment process. (<b>F</b>) Survival analysis of mice across different treatment groups. (G1: sodium alginate, G2: DSF, G3: CuO NPs, G4: DSF@CuO, and G5: DSF@CuO Gel). Data are presented as mean ± SD and are representative of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>DSF@CuO Gels could inhibit tumors in an in situ mice model of HCC. (<b>A</b>) A schematic illustrating the treatment procedure. (<b>B</b>,<b>C</b>) IVIS for monitoring the size of in situ HCC in mice. (<b>D</b>) Immunofluorescence images of cuproptosis proteins in mouse tumor tissues after treatment across different groups; cell nuclei were stained with DAPI (blue), the DLAT were stained with FITC (green), and the LIAS were stained with cy3 (green) (scale bars = 20 μm). (<b>E</b>) Survival analysis of mice during the treatment period across different treatment groups. (<b>F</b>,<b>G</b>) WB detection of HIF-1α expression. (G1: sodium alginate, G2: DSF, G3: CuO NPs, G4: DSF@CuO, and G5: DSF@CuO Gel). Data are presented as mean ± SD and are representative of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Scheme 1
<p>Schematic illustration of injection of DSF@CuO Gel hydrogel for treatment of LenR HCC.</p>
Full article ">
Back to TopTop