Cytojournal最新文献

筛选
英文 中文
A new perspective: Acyl-CoA synthetase long-chain family member 4 inhibits ubiquitin-specific protease 7-induced epithelial ovarian cancer progression by inducing ferroptosis and M1 macrophage polarization. 新视角:酰基辅酶A合成酶长链家族成员4通过诱导铁下垂和M1巨噬细胞极化抑制泛素特异性蛋白酶7诱导的上皮性卵巢癌进展。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-03-05 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_241_2024
Yazhou Qi, Qianwen Li, Limin Chen, Shuimiao Zhao, Jiaoran Nie, Gaoyuan Liu
{"title":"A new perspective: Acyl-CoA synthetase long-chain family member 4 inhibits ubiquitin-specific protease 7-induced epithelial ovarian cancer progression by inducing ferroptosis and M1 macrophage polarization.","authors":"Yazhou Qi, Qianwen Li, Limin Chen, Shuimiao Zhao, Jiaoran Nie, Gaoyuan Liu","doi":"10.25259/Cytojournal_241_2024","DOIUrl":"https://doi.org/10.25259/Cytojournal_241_2024","url":null,"abstract":"<p><strong>Objective: </strong>Epithelial ovarian cancer (EOC) is the most common and lethal type of ovarian cancer, and the cross-talk between tumor cell ferroptosis and macrophages is essential to cancer progression. This study aims to investigate the roles of ubiquitin-specific protease 7 (USP7) and acyl-CoA synthetase long-chain family member 4 (ACSL4) in the pathogenesis of EOC.</p><p><strong>Material and methods: </strong>The expression patterns of USP7 and ACSL4 in EOC cell lines were first determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot. ACSL4 recombinant protein was applied alone or in conjunction with a USP7 overexpression plasmid in EOC cells, and the effects of USP7 and ACSL4 on EOC cell proliferation and apoptosis were assessed using colony formation assays and terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling staining. The effects of USP7 and ACSL4 on ferroptosis in EOC cells were evaluated by measuring reactive oxygen species (ROS) fluorescence intensity, malondialdehyde (MDA), glutathione (GSH) levels, and glutathione peroxidase 4 (GPX4) messenger RNA (mRNA) levels. Co-culture of EOC cell-conditioned medium treated with ACSL4 recombinant protein or USP7 overexpression plasmid was performed with Human Acute Monocytic Leukemia Cell Line (THP-1) macrophages, and the expression levels of cluster of differentiation 86 and cluster of differentiation 206 were analyzed by flow cytometry. The expression levels of M1 polarization markers and M2 markers in macrophages were measured by qRT-PCR.</p><p><strong>Results: </strong>ACSL4 was expressed at low levels in the EOC cell lines, whereas USP7 was expressed at high levels. Treatment with ACSL4 recombinant protein reduced colony formation and increased apoptotic cell levels in the EOC cells (<i>P</i> < 0.001). In addition, ACSL4 treatment increased ROS fluorescence intensity and MDA levels while decreasing GSH levels and GPX4 expression (<i>P</i> < 0.001). Furthermore, ACSL4 treatment promoted the polarization of THP-1 macrophages toward M1, increasing the expression of M1 markers (<i>P</i> < 0.001). USP7 overexpression exerted the opposite effect (<i>P</i> < 0.001).</p><p><strong>Conclusion: </strong>This study reveals the critical role of USP7 in the progression of EOC. ACSL4 inhibits EOC growth and anti-apoptosis by inhibiting USP7-induced antiferroptosis and anti-M1 macrophage polarization, highlighting this mechanism as a potential therapeutic target in EOC.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"28"},"PeriodicalIF":2.5,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010880/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144051596","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metallothionein 2A enhances the yes-associated protein 1 signaling pathway to promote small-cell lung cancer metastasis. 金属硫蛋白2A增强yes相关蛋白1信号通路促进小细胞肺癌转移
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-03-03 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_201_2024
Hong Wu, Yangyang Gu, Lidong Xu
{"title":"Metallothionein 2A enhances the yes-associated protein 1 signaling pathway to promote small-cell lung cancer metastasis.","authors":"Hong Wu, Yangyang Gu, Lidong Xu","doi":"10.25259/Cytojournal_201_2024","DOIUrl":"https://doi.org/10.25259/Cytojournal_201_2024","url":null,"abstract":"<p><strong>Objective: </strong>Small-cell lung cancer (SCLC) remains challenging to treat due to its high invasiveness and propensity for drug resistance. Evidence suggests that the regulatory relationship between metallothionein 2A (MT2A) and the yes-associated protein 1 (YAP1) signaling pathway may influence the development of SCLC. Therefore, this study aims to explore the potential mechanisms affecting SCLC progression based on the regulatory interaction between YAP1 and MT2A.</p><p><strong>Material and methods: </strong>This study utilized reverse transcription quantitative polymerase chain reaction and Western blot analysis to analyze MT2A expression in cells. SCLC cell models with MT2A silencing and overexpression, as well as cotransfected cell models with YAP1 silencing and MT2A overexpression, were constructed. The effect of MT2A/YAP1 on cell growth, migration, and invasion was evaluated through a series of experiments, including cell viability assessment using cell counting kit-8 assay, colony formation examination, 5-ethynyl-2'-deoxyuridine staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining, and Transwell analysis. In addition, Western blot analysis was conducted to investigate alterations in crucial proteins associated with the YAP1 pathway and the epithelial-mesenchymal transition ( EMT) markers influenced by MT2A/YAP1. Lung metastasis and Ki67 expression were analyzed through hematoxylin and eosin staining and immunofluorescence analysis <i>in vivo</i>.</p><p><strong>Results: </strong>In the SCLC cell line ( NCI-H69 cells), MT2A exhibits increased expression, facilitating cell growth, migration, and invasion. YAP1 expression decreases when MT2A is depleted. In addition, our findings validate that MT2A facilitates EMT progression and SCLC invasion and metastasis by upregulating YAP1 expression. <i>In vitro</i>, silencing MT2A inhibits lung metastasis and Ki67 expression.</p><p><strong>Conclusion: </strong>MT2A facilitates the migration and invasion of SCLC cells by influencing the YAP1 signaling cascade. This investigation offers a fresh avenue for delving deeply into the potential mechanisms involved in the progression of SCLC.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"25"},"PeriodicalIF":2.5,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010882/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144040138","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nobiletin alleviates brain injury in uremic mice and inhibits indoxyl sulfate-induced neurotoxicity in HT22 cells through the phosphatidylinositol 3-kinase/protein kinase B signaling pathway. 诺比林素通过磷脂酰肌醇3-激酶/蛋白激酶B信号通路,减轻尿毒症小鼠脑损伤,抑制硫酸吲哚酚诱导的HT22细胞神经毒性。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-03-03 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_233_2024
Liangshi Xu, Ruyi Zhang
{"title":"Nobiletin alleviates brain injury in uremic mice and inhibits indoxyl sulfate-induced neurotoxicity in HT22 cells through the phosphatidylinositol 3-kinase/protein kinase B signaling pathway.","authors":"Liangshi Xu, Ruyi Zhang","doi":"10.25259/Cytojournal_233_2024","DOIUrl":"https://doi.org/10.25259/Cytojournal_233_2024","url":null,"abstract":"<p><strong>Objective: </strong>Uremic encephalopathy presents as central nervous system symptoms in acute and chronic renal failure. Nobiletin (NOB), an extract from chenpi, has demonstrated anti-inflammatory bioactivity and potential neuroprotective effects without remarkable toxicity. This study aims to evaluate the pharmacological effects of NOB on treating uremic brain injury and elucidate its underlying mechanisms.</p><p><strong>Material and methods: </strong>A uremic encephalopathy mouse model was established by inducing renal failure with cisplatin (DDP). The therapeutic effects of NOB were investigated by assessing its effect on brain damage and neuronal viability. HT22 murine hippocampal neurons were also treated with DDP to induce neurotoxicity, and the effects of NOB on cell viability, apoptosis, and the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway were examined. The PI3K inhibitor LY294002 was used to further investigate the involvement of the PI3K/Akt pathway in the neuroprotective effects of NOB.</p><p><strong>Results: </strong>NOB alleviated uremia-induced brain damage in mice, and this function was associated with the activation of the PI3K/Akt signaling pathway. <i>In vitro</i>, NOB improved the DPP-suppressed cell viability in HT22 neurons and restored apoptosis. NOB treatment also restored the phosphorylation levels of PI3K, Akt, and Pyruvate dehydrogenase kinase 1. These effects were partially blocked by the PI3K inhibitor LY294002.</p><p><strong>Conclusion: </strong>NOB exerts potent neuroprotective effects by activating the PI3K/Akt pathway, mitigating uremia-induced brain injury and preventing DDP-induced neurotoxicity. These findings support the potential therapeutic application of NOB for uremic encephalopathy and provide insights into its underlying mechanisms.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"27"},"PeriodicalIF":2.5,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010812/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144041556","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Scavenger receptor class B member 1 promotes lung cancer growth and metastasis through enhanced twist family BHLH transcription factor 1 signaling in vitro and in vivo: Exploration of RPPNs as a therapeutic Strategy. 清道夫受体B类成员1通过增强twist家族BHLH转录因子1信号传导在体外和体内促进肺癌的生长和转移:RPPNs作为治疗策略的探索。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-03-03 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_222_2024
Xin Liu, Hong Bian, Yan Shi, Tongxin Du
{"title":"Scavenger receptor class B member 1 promotes lung cancer growth and metastasis through enhanced twist family BHLH transcription factor 1 signaling <i>in vitro</i> and <i>in vivo</i>: Exploration of RPPNs as a therapeutic Strategy.","authors":"Xin Liu, Hong Bian, Yan Shi, Tongxin Du","doi":"10.25259/Cytojournal_222_2024","DOIUrl":"10.25259/Cytojournal_222_2024","url":null,"abstract":"<p><strong>Objective: </strong>Lung cancer remains a leading cause of cancer-related mortality worldwide. Although scavenger receptor class B member 1 (SCARB1), a crucial cell surface receptor, plays a vital role in various cancers, its function in lung cancer remains incompletely elucidated. This study aims to investigate the role and molecular mechanisms of SCARB1 in lung cancer progression and develop a novel SCARB1-targeted nanoparticle drug delivery system.</p><p><strong>Material and methods: </strong>We analyzed SCARB1 expression levels in lung cancer tissues and their correlation with patient prognosis using the Cancer Genome Atlas database. <i>In vitro</i> experiments, including quantitative real-time polymerase chain reaction, Western blot, 5-ethynyl-2'-deoxyuridine, colony formation, and Transwell analyses, were conducted to study the effects of SCARB1 on lung cancer cell proliferation, invasion, and migration. A lung metastasis model was established through tail vein injection to evaluate the role of SCARB1 in promoting lung cancer metastasis <i>in vivo</i>. We also developed red blood cell membrane-coated poly (lactic-co-glycolic acid) nanocarriers loaded with paclitaxel (RPPNs) and assessed their effect on SCARB1 expression and lung cancer progression.</p><p><strong>Results: </strong>SCARB1 was overexpressed in human lung cancer tissues and significantly associated with poor patient prognosis. <i>In vitro</i> experiments confirmed that silencing SCARB1 inhibited lung cancer cell growth, invasion, and migration. SCARB1 overexpression promoted lung cancer cell proliferation, migration, and epithelial-mesenchymal transition through twist family BHLH transcription factor 1 (Twist1) activation. <i>In vivo</i> experiments further validated the crucial role of SCARB1 in promoting lung cancer metastasis. The developed RPPNs effectively suppressed SCARB1 expression in lung cancer and demonstrated superior inhibitory effects compared with traditional RPPNs. However, SCARB1 overexpression partially antagonized the antimetastatic effects of RPPNs.</p><p><strong>Conclusion: </strong>This work elucidates, for the 1<sup>st</sup> time, the molecular mechanism by which SCARB1 promotes lung cancer growth and metastasis through the activation of the Twist1 signaling pathway and develops a novel SCARB1-targeted nanoparticle drug delivery system, namely RPPNs. The findings of this work not only deepen our understanding of the molecular mechanisms underlying lung cancer progression but also provide new strategies for lung cancer diagnosis and treatment.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"23"},"PeriodicalIF":2.5,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010815/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144027772","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Emerging regulators of gastric cancer angiogenesis: Synergistic effects of regulator of G protein signaling 4 and midkine. 胃癌血管生成的新兴调节因子:G蛋白信号传导4和midkine调节因子的协同作用。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-03-03 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_212_2024
Yanxin He, Hao Li, Kang Li, HaiPing Song
{"title":"Emerging regulators of gastric cancer angiogenesis: Synergistic effects of regulator of G protein signaling 4 and midkine.","authors":"Yanxin He, Hao Li, Kang Li, HaiPing Song","doi":"10.25259/Cytojournal_212_2024","DOIUrl":"https://doi.org/10.25259/Cytojournal_212_2024","url":null,"abstract":"<p><strong>Objective: </strong>Globally, gastric cancer (GC) is among the most prevalent cancers. The development and spread of stomach cancer are significantly influenced by angiogenesis. However, the molecular mechanisms underlying this process remain unclear. This study aimed to investigate the role of the regulator of G protein signaling 4 (RGS4) in GC angiogenesis and its potential mechanisms.</p><p><strong>Material and methods: </strong>Through <i>in vitro</i> and <i>in vivo</i> experiments, including tube formation assays and xenograft models in nude mice, we evaluated the effects of RGS4 on GC angiogenesis and metastasis. In addition, we employed techniques such as immunoprecipitation and immunofluorescence double staining to explore the interaction between RGS4 and midkine (MDK). Survival analysis was also performed to evaluate the association between the prognosis of patients with GC and the expression levels of RGS4 and MDK.</p><p><strong>Results: </strong>Our findings revealed that RGS4 is a crucial factor in GC metastasis, significantly inducing angiogenesis. Further studies indicated that RGS4 directly interacts with MDK and upregulates its expression. By upregulating MDK, RGS4 stimulates the angiogenesis and metastasis of GC. Furthermore, a poor prognosis for patients with GC is directly linked to high expression of RGS4 and MDK.</p><p><strong>Conclusion: </strong>This work is the first to clarify the molecular mechanism by which RGS4 upregulates MDK expression to increase GC angiogenesis. These findings not only enhance our understanding of the mechanisms underlying GC progression but also provide potential targets for developing new anti-angiogenic and antimetastatic therapies. RGS4 and MDK could serve as effective biomarkers for predicting the prognosis of patients with GC and offer new insights into personalized treatment approaches.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"26"},"PeriodicalIF":2.5,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010818/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144055242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Interactions of tumor necrosis factor receptor-associated factor 4 and pyruvate kinase muscle isoform 2 promote malignant behavior and aerobic glycolysis in colorectal cancer cells. 肿瘤坏死因子受体相关因子4和丙酮酸激酶肌异构体2的相互作用促进结直肠癌细胞的恶性行为和有氧糖酵解。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-03-03 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_167_2024
Tongming Liu, Shihong Zhu, Jiawei Sun, Yuanyuan Ma
{"title":"Interactions of tumor necrosis factor receptor-associated factor 4 and pyruvate kinase muscle isoform 2 promote malignant behavior and aerobic glycolysis in colorectal cancer cells.","authors":"Tongming Liu, Shihong Zhu, Jiawei Sun, Yuanyuan Ma","doi":"10.25259/Cytojournal_167_2024","DOIUrl":"10.25259/Cytojournal_167_2024","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Objective: &lt;/strong&gt;Colorectal cancer (CRC) is a malignant tumor of the digestive system, and the main causes of death are metastasis and recurrence. Tumor necrosis factor receptor-associated factor 4 (TRAF4) is associated with the development of various tumors, but its role in CRC development is limited, especially glycolysis. Therefore, TRAF4's role in the regulation of cell malignant behavior and glycolysis and its specific mechanism were explored in CRC.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Material and methods: &lt;/strong&gt;The TRAF4 or pyruvate kinase muscle isoform 2 (PKM2) gene expression was inhibited or promoted by short hairpin ribonucleic acid (sh- RNA) or overexpression (oe) plasmids in Lovo cells. Transfection efficiency was detected by Western blot and real-time quantitative polymerase chain reaction. Cell growth and colony formation were assessed using 5-ethynyl-2'-deoxyuridine and clone formation assays, respectively, and cell migration and invasion ability were observed by scratch healing and Transwell assay. Glucose uptake and lactate production were measured with a kit and used in evaluating the glycolysis capacities of the cells. The levels of TRAF4, PKM2, and glycolytic-related and wingless-type (Wnt)/beta (β)-catenin pathway-related proteins were detected by Western blot, and co-immunoprecipitation (Co-IP) verified TRAF4 and PKM2 interaction in CRC cells.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;TRAF4 expression increased in CRC cell lines (&lt;i&gt;P &lt;&lt;/i&gt; 0.05, &lt;i&gt;P &lt;&lt;/i&gt; 0.001, &lt;i&gt;P &lt;&lt;/i&gt; 0.0001). After sh-TRAF4, oeTRAF4, or oe-PKM2 transfection, TRAF4 or PKM2 expression levels in the Lovo cells decreased or increased (&lt;i&gt;P &lt;&lt;/i&gt; 0.05, &lt;i&gt;P &lt;&lt;/i&gt; 0.01, &lt;i&gt;P &lt;&lt;/i&gt; 0.001, and &lt;i&gt;P &lt;&lt;/i&gt; 0.0001). TRAF4 knockdown inhibited cell malignant behavior, glucose uptake, lactate production, and glucose transporter type 1 (GLUT1), hexokinase 2 (HK2), PKM2, and lactate dehydrogenase A (LDHA) protein expression levels in CRC cells (&lt;i&gt;P &lt;&lt;/i&gt; 0.01, &lt;i&gt;P &lt;&lt;/i&gt; 0.001, &lt;i&gt;P &lt;&lt;/i&gt; 0.0001). Co-IP experiment showed that TRAF4 was bound to PKM2. PKM2 protein level decreased after TRAF4 knockdown (&lt;i&gt;P &lt;&lt;/i&gt; 0.0001), and PKM2 protein expression increased when TRAF4 was overexpressed (&lt;i&gt;P &lt;&lt;/i&gt; 0.001). PKM2 overexpression offset the effect of TRAF4 knockdown on cell malignant behavior and aerobic glycolysis (&lt;i&gt;P &lt;&lt;/i&gt; 0.05, &lt;i&gt;P &lt;&lt;/i&gt; 0.01, &lt;i&gt;P &lt;&lt;/i&gt; 0.001, and &lt;i&gt;P &lt;&lt;/i&gt; 0.0001). Moreover, Wnt/β-catenin pathway proteins were inhibited after TRAF4 knockdown and were restored by PKM2 overexpression (&lt;i&gt;P &lt;&lt;/i&gt; 0.01 and &lt;i&gt;P &lt;&lt;/i&gt; 0.0001). Notably, the effects of TRAF4 or PKM2 overexpression on cell malignant behavior, glucose uptake, lactate production, and GLUT1, PKM2, HK2, and LDHA protein expression levels were partially offset by the Wnt/β-catenin signaling suppressor XAV939 (&lt;i&gt;P &lt;&lt;/i&gt; 0.05, &lt;i&gt;P &lt;&lt;/i&gt; 0.01, &lt;i&gt;P &lt;&lt;/i&gt; 0.001, and &lt;i&gt;P &lt;&lt;/i&gt; 0.0001).&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;TRAF4 and PKM2 are associated with CRC development. TRAF4 binds to PKM2 and promotes ","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"24"},"PeriodicalIF":2.5,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12010814/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144051602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lysine acetyltransferase 5 contributes to diabetic retinopathy by modulating autophagy through epigenetically regulating autophagy-related gene 7. 赖氨酸乙酰转移酶5通过表观遗传调控自噬相关基因7调控自噬参与糖尿病视网膜病变。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-02-18 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_187_2024
Qi Gao, Yanjun Lai, Shuai He, Yanhua Wang, Guochao Zhang, Xinyu Zhu, Shifang Zhuang
{"title":"Lysine acetyltransferase 5 contributes to diabetic retinopathy by modulating autophagy through epigenetically regulating autophagy-related gene 7.","authors":"Qi Gao, Yanjun Lai, Shuai He, Yanhua Wang, Guochao Zhang, Xinyu Zhu, Shifang Zhuang","doi":"10.25259/Cytojournal_187_2024","DOIUrl":"10.25259/Cytojournal_187_2024","url":null,"abstract":"<p><strong>Objective: </strong>Diabetic retinopathy (DR) is a prevalent and serious complication among individuals with diabetes, significantly compromising their visual acuity and overall quality of life. Lysine acetyltransferase 5 (KAT5), an essential catalytic subunit of the nucleosome acetyltransferase of the H4 complex, is implicated in the development of various diseases, including neurological disorders, breast cancer, and lung cancer. However, the function of KAT5 in DR remains poorly understood. This study aims to investigate the influence of KAT5 on autophagy (Atg) during DR.</p><p><strong>Material and methods: </strong>Experiments were conducted using streptozotocin (STZ)-treated rats to induce diabetes and observe changes in KAT5 expression and its effect on Atg. Retinal tissues and RF/6A cells were utilized to analyze the expression levels of various proteins and their involvement in Atg and apoptosis. KAT5 depletion and Atg7 knockdown were performed to further understand their roles in the process.</p><p><strong>Results: </strong>The eyeballs of STZ-treated rats showed increased expression of KAT5. Depletion of KAT5 attenuated STZ-induced DR injury in rats. The retinal tissues of STZ-treated rats exhibited reduced expression of B-cell lymphoma-2 (Bcl-2) and increased levels of BCL-2-associated X protein and cleaved caspase 3, which could be reversed by KAT5 depletion. STZ treatment induced expression of Beclin-1 and microtubule-associated protein 1 light chain 3B in retinal tissues, and KAT5 knockdown blocked this effect. In monkey retinal choroidal endothelial ( RF/6A) cells, high glucose (HG) treatment decreased 5-ethynyl-2'-deoxyuridine-positivecells and increased terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells, which were reversed by KAT5 depletion. KAT5 depletion also attenuated HG-induced apoptosis and Atg in RF/6A cells. Mechanistically, KAT5 depletion reduced histone H3 lysine 27 acetylation and ribonucleic acid ( RNA) polymerase II enrichment on the Atg7 promoter, leading to a decrease in the messenger RNA ( mRNA) and protein expression of Atg7. Atg7 knockdown suppressed Atg in RF/6A cells under HG conditions and reversed the effect of KAT5 depletion on cell apoptosis and Atg.</p><p><strong>Conclusion: </strong>The findings suggest that KAT5 contributes to DR by modulating Atg through epigenetic regulation of Atg7. KAT5 emerges as a valuable target for DR treatment, providing a fresh perspective on the disease's pathogenesis and laying the foundation for the development of potential therapeutic strategies.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"22"},"PeriodicalIF":2.5,"publicationDate":"2025-02-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11932963/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143711850","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prominin 2 knockdown inhibits the growth, migration, and invasion of non-small cell lung cancer cells by repressing phosphatidylinositol 3 kinase/protein kinase B pathway. pronin2敲低通过抑制磷脂酰肌醇3激酶/蛋白激酶B通路抑制非小细胞肺癌细胞的生长、迁移和侵袭。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-02-17 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_118_2024
Biao He, Ze Chen, Liang Zhong, Xiaoyong Pang
{"title":"Prominin 2 knockdown inhibits the growth, migration, and invasion of non-small cell lung cancer cells by repressing phosphatidylinositol 3 kinase/protein kinase B pathway.","authors":"Biao He, Ze Chen, Liang Zhong, Xiaoyong Pang","doi":"10.25259/Cytojournal_118_2024","DOIUrl":"10.25259/Cytojournal_118_2024","url":null,"abstract":"<p><strong>Objective: </strong>The prognosis of patients with non-small cell lung cancer (NSCLC) is poor, and this malignancy represents a grievous danger to human health due to its high rates of recurrence and metastasis. Previous studies have linked prominin 2 (PROM2) to certain cancers. However, the impact of PROM2 on the biological behavior of NSCLC cells and regulatory pathways has rarely been explored. Therefore, the study aims to elucidate the roles and regulatory mechanisms of PROM2 in the cell function of NSCLC by interfering with PROM2.</p><p><strong>Material and methods: </strong>PROM2 messenger ribonucleic acid (mRNA) and protein expression levels in NSCLC cells were analyzed by applying quantitative real-time polymerase chain reaction and Western blot analysis. Phosphatidylinositol 3 kinase (PI3K), protein kinase B (AKT), and phosphorylated-AKT (p-AKT) protein levels were evaluated through Western blot analysis. Cell counting kit-8 and Transwell assays were used to evaluate NSCLC cell proliferation, migration, and invasion.</p><p><strong>Results: </strong>PROM2 mRNA protein levels drastically increased in NSCLC tissues and cells. High PROM2 mRNA level was related to the poor prognosis of patients with NSCLC. PROM2 silencing remarkably repressed NCI-H520 and A549 cell proliferation, migration, and invasion. Furthermore, PI3K and p-AKT protein levels clearly decreased after PROM2 silencing. Importantly, rescue experiments elucidated that PI3K/AKT pathway activation could reverse the inhibitory effect of PROM2 silencing on the proliferation, migration, and invasion of NCI-H520 and A549 cells.</p><p><strong>Conclusion: </strong>This study verified that PROM2 knockdown inhibits the growth, migration, and invasion of NSCLC by repressing the PI3K/AKT pathway.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"21"},"PeriodicalIF":2.5,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11932975/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143711851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploration of immunocytochemical biomarkers related to central lymph node metastasis in papillary thyroid microcarcinoma. 甲状腺乳头状微癌中央淋巴结转移相关免疫细胞化学生物标志物的探讨。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-02-14 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_162_2024
Lulu Rong, Jie Wang, Qian Wang, Yanli Zhu, Wenhao Ren
{"title":"Exploration of immunocytochemical biomarkers related to central lymph node metastasis in papillary thyroid microcarcinoma.","authors":"Lulu Rong, Jie Wang, Qian Wang, Yanli Zhu, Wenhao Ren","doi":"10.25259/Cytojournal_162_2024","DOIUrl":"10.25259/Cytojournal_162_2024","url":null,"abstract":"<p><strong>Objective: </strong>The presence of central lymph node metastasis (CLNM) represents a critical determinant in ascertaining the necessity for surgical intervention in patients with papillary thyroid microcarcinoma (PTMC). However, the predominant current methodologies for confirming the central lymph node status in clinical practice are hampered by the low predictive accuracy of preoperative ultrasound examination and the high risk of preoperative fine needle aspiration (FNA). Consequently, the objective of this study is to investigate and identify specific immunocytochemical biomarkers for predicting CLNM in PTMC patients based on preoperative thyroid FNA samples.</p><p><strong>Material and methods: </strong>In this study, the messenger ribonucleic acid sequencing data of pathological tumor stage 1 (pT1) papillary thyroid carcinoma (PTC) accompanied by pathological node stage information were initially retrieved from The Cancer Genome Atlas database. The differential expression genes (DEGs) between the pT1N1-PTC group and the pT1N0-PTC group were ascertained through bioinformatics methodology. Subsequently, these DEGs were imported into Cytoscape software to identify hub genes. Ultimately, immunohistochemical and immunocytochemical staining were employed to validate whether the biomarkers corresponding to the main hub genes demonstrated statistical significance in predicting CLNM within propensity score-matched PTMC samples.</p><p><strong>Results: </strong>In this study, a total of 292 DEGs and 10 hub genes were successfully identified. Subsequently, immunohistochemical and immunocytochemical staining were conducted on 208 PTMC cases selected through propensity score matching. Among these 208 cases, the biomarkers (Cytokeratin 5/6 [CK5/6], Chromogranin A [CgA], and Pair box gene 2 [Pax-2]) corresponding to the main hub genes (Cytokeratin 5 [KRT5], Cytokeratin 6 [KRT6A], Chromogranin A [CHGA], and PAX2) were subjected to immunohistochemical staining in postoperative thyroidectomy specimens, the immunohistochemical staining results revealed a statistically significant difference in CK5/6 expression between PTMCs with and without CLNM (<i>P</i> = 0.002). Subsequently, CK5/6 immunocytochemical staining performed on preoperative thyroid FNA liquid-based samples further corroborated that CK5/6 expression was more prone to being positive in PTMCs with CLNM (<i>P</i> = 0.010).</p><p><strong>Conclusion: </strong>CK5/6 is a valuable immunocytochemical biomarker capable of predicting the occurrence of CLNM in PTMC patients prior to surgery.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"18"},"PeriodicalIF":2.5,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11932946/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143711847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring miR-3148's impact on Krüppel-like factor 6-driven mitophagy and apoptosis in myocardial ischemic injury. 探讨miR-3148对心肌缺血损伤中kr<s:1> ppel样因子6驱动的线粒体自噬和细胞凋亡的影响。
IF 2.5 4区 医学
Cytojournal Pub Date : 2025-02-14 eCollection Date: 2025-01-01 DOI: 10.25259/Cytojournal_209_2024
Chusheng Huang, Lipeng Li, Hailong Deng, Jincheng Su, Qingjun Wei, Ying He, Lei Xian
{"title":"Exploring miR-3148's impact on Krüppel-like factor 6-driven mitophagy and apoptosis in myocardial ischemic injury.","authors":"Chusheng Huang, Lipeng Li, Hailong Deng, Jincheng Su, Qingjun Wei, Ying He, Lei Xian","doi":"10.25259/Cytojournal_209_2024","DOIUrl":"10.25259/Cytojournal_209_2024","url":null,"abstract":"<p><strong>Objective: </strong>Myocardial infarction (MI) is a leading cause of death worldwide, accounting for millions of fatalities annually. The injury and repair of cardiomyocytes are closely associated with the changes in gene expression. MicroRNAs could serve as a potential target for MI treatment. This work aims to investigate the role of miR-3148 in mitochondrial dynamics during acute MI (AMI) with a specific focus on its regulatory mechanisms in mitophagy and apoptosis, which could reveal potential therapeutic targets for AMI treatment.</p><p><strong>Material and methods: </strong>MiR-3148 levels in patients with AMI and experimental models were measured to assess the effects of miR-3148 on cardiomyocyte viability under oxygen and glucose deprivation (OGD). The present investigation involved monitoring mitophagy markers, including PTEN-induced kinase 1 (PINK1), parkin RBR E3 ubiquitin-protein ligase (Parkin), Beclin1, and microtubule-associated protein 1A/1B light chain 3 II/I (LC3 II/I) ratio, as well as apoptotic markers such as cysteine-aspartic acid protease (Caspase) 9, Caspase 3, and cytochrome C (Cyt C). In addition, Krüppel-like factor 6 (KLF6) was examined as a target of miR-3148.</p><p><strong>Results: </strong>MiR-3148 was significantly elevated in patients with AMI and models. MiR-3148 overexpression reduced cardiomyocyte viability, whereas miR-3148 knockdown protected against OGD injury. The inhibition of miR-3148 activated mitophagy, as shown by the increased PINK1, Parkin, Beclin1 levels, and LC3 II/I ratios, and reduced sequestosome 1 (p62), and apoptotic markers levels. MiR-3148 directly targeted KLF6, reducing its expression. The suppression of KLF6 aggravated OGD injury by disrupting PINK1/Parkin-mediated mitophagy and enhancing apoptosis. Attenuating KLF6 expression reversed the protective effects of miR-3148 inhibition, indicating reciprocal regulation.</p><p><strong>Conclusion: </strong>In myocardial ischemic injury, miR-3148 modulates PINK1/Parkin-mediated mitophagy and apoptosis through KLF6 regulation. This finding highlights miR-3148 as a key factor in the pathogenesis of AMI and as a potential therapeutic target.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"19"},"PeriodicalIF":2.5,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11932976/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143711848","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信