Pub Date : 2025-07-19eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_108_2024
Azaa Salim Al-Zakwani, Bushra Said Al-Hatrooshi
Kikuchi's lymphadenitis (KL) is a benign, self-limited, reactive condition with unknown etiology, usually seen in young women of Asian descent. It is most commonly seen in the cervical lymph nodes; however, in rare cases, axillary lymph nodes are involved. Cytological diagnosis is possible with adequate lymph node sampling by fine needle aspiration in the proper clinical setting. Pathologists face some difficulties in making the diagnosis of KL due to overlapping findings among other conditions, such as tuberculosis, lupus lymphadenitis, non-specific reactive conditions, and even malignant lymphoma. Diagnosis by cytopathology has the advantage of using minimally invasive interventions, which avoid the unnecessary excision of the lymph node for this benign condition. Herein, we report a case of KL in a patient who presented with right axillary lymphadenopathy.
{"title":"Fine needle aspiration diagnosis of Kikuchi's lymphadenitis in axillary lymphadenopathy: A rare condition with an unusual presentation site.","authors":"Azaa Salim Al-Zakwani, Bushra Said Al-Hatrooshi","doi":"10.25259/Cytojournal_108_2024","DOIUrl":"10.25259/Cytojournal_108_2024","url":null,"abstract":"<p><p>Kikuchi's lymphadenitis (KL) is a benign, self-limited, reactive condition with unknown etiology, usually seen in young women of Asian descent. It is most commonly seen in the cervical lymph nodes; however, in rare cases, axillary lymph nodes are involved. Cytological diagnosis is possible with adequate lymph node sampling by fine needle aspiration in the proper clinical setting. Pathologists face some difficulties in making the diagnosis of KL due to overlapping findings among other conditions, such as tuberculosis, lupus lymphadenitis, non-specific reactive conditions, and even malignant lymphoma. Diagnosis by cytopathology has the advantage of using minimally invasive interventions, which avoid the unnecessary excision of the lymph node for this benign condition. Herein, we report a case of KL in a patient who presented with right axillary lymphadenopathy.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"69"},"PeriodicalIF":3.1,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514765/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281614","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}
Pub Date : 2025-07-16eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_89_2025
Sana Ahuja, Sufian Zaheer
{"title":"Liquid-based cytology in the era of multi-omics and artificial intelligence integration.","authors":"Sana Ahuja, Sufian Zaheer","doi":"10.25259/Cytojournal_89_2025","DOIUrl":"10.25259/Cytojournal_89_2025","url":null,"abstract":"","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"68"},"PeriodicalIF":3.1,"publicationDate":"2025-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281577","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}
Pub Date : 2025-07-02eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_271_2024
Xinyue Zhang, Di Na
Objective: The immunosuppressive tumor microenvironment (TME) limits the treatment effectiveness of immunotherapy in gastric cancer (GC). This study investigated the role of membrane-spanning four domains subfamily A member 4A (MS4A4A) in the regulation of macrophage polarization and its effect on the immune response in GC, with the aim of enhancing the effectiveness of immunotherapy by addressing the immunosuppressive TME.
Material and methods: A GC ectopic tumor model was initiated in C57BL/6 mice with subcutaneous MKN-45 injection. Five groups were formed by randomly dividing the mice: model, MS4A4A recombinant protein, MS4A4A antibody, programmed cell death protein 1 (PD-1) antibody, and MS4A4A recombinant protein + PD-1 antibody groups. MKN-45 cells and bone marrow-derived macrophages (BMDMs) were cocultured with the MS4A4A protein or antibody. Macrophage polarization was analyzed through flow cytometry, gene expression through quantitative real-time polymerase chain reaction (qRT-PCR), cytokine levels through enzyme-linked immunosorbent assay, protein expression through Western blot, and tumor morphology through hematoxylin and eosin staining.
Results: In the GC mouse model, the MS4A4A recombinant protein markedly enhanced tumor growth (P < 0.001), and the MS4A4A antibody exhibited an inhibitory effect (P < 0.001). MS4A4A recombinant protein decreased the levels of inflammatory cytokine concentrations and increased those of anti-inflammatory mediator concentrations (P < 0.001). By contrast, the MS4A4A antibody treatment group displayed the opposite effect. Inhibition of MS4A4A enhanced the accumulation of macrophages, CD4+ T cells, and CD8+ T cells in the tumor (P < 0.001). Flow cytometry and qRT-PCR analyses showed that MS4A4A promoted M2 macrophage polarization, and MS4A4A antibody induced M1 polarization (P < 0.001). MS4A4A played a key role in inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells subunit p50 (p50) during M1 polarization. Furthermore, the PD-1 antibody reversed the pro-tumor effects of MS4A4A, which reestablished pro-inflammatory cytokine levels while lowering anti-inflammatory cytokines (P < 0.001).
Conclusion: This study shows that MS4A4A promotes tumor growth by inducing M2 macrophage polarization and suppressing immune responses, while MS4A4A antibody enhances anti-tumor immunity by inducing M1 polarization. PD-1 antibody reverses MS4A4A's pro-tumor effects, restoring anti-tumor immunity. MS4A4A inhibitors or their combination with PD-1 antibodies may offer a promising strategy for GC immunotherapy.
{"title":"Membrane-spanning 4 domains subfamily A member 4A inhibition promotes M1 macrophage polarization and enhances anti-gastric cancer immune response.","authors":"Xinyue Zhang, Di Na","doi":"10.25259/Cytojournal_271_2024","DOIUrl":"10.25259/Cytojournal_271_2024","url":null,"abstract":"<p><strong>Objective: </strong>The immunosuppressive tumor microenvironment (TME) limits the treatment effectiveness of immunotherapy in gastric cancer (GC). This study investigated the role of membrane-spanning four domains subfamily A member 4A (MS4A4A) in the regulation of macrophage polarization and its effect on the immune response in GC, with the aim of enhancing the effectiveness of immunotherapy by addressing the immunosuppressive TME.</p><p><strong>Material and methods: </strong>A GC ectopic tumor model was initiated in C57BL/6 mice with subcutaneous MKN-45 injection. Five groups were formed by randomly dividing the mice: model, MS4A4A recombinant protein, MS4A4A antibody, programmed cell death protein 1 (PD-1) antibody, and MS4A4A recombinant protein + PD-1 antibody groups. MKN-45 cells and bone marrow-derived macrophages (BMDMs) were cocultured with the MS4A4A protein or antibody. Macrophage polarization was analyzed through flow cytometry, gene expression through quantitative real-time polymerase chain reaction (qRT-PCR), cytokine levels through enzyme-linked immunosorbent assay, protein expression through Western blot, and tumor morphology through hematoxylin and eosin staining.</p><p><strong>Results: </strong>In the GC mouse model, the MS4A4A recombinant protein markedly enhanced tumor growth (<i>P</i> < 0.001), and the MS4A4A antibody exhibited an inhibitory effect (<i>P</i> < 0.001). MS4A4A recombinant protein decreased the levels of inflammatory cytokine concentrations and increased those of anti-inflammatory mediator concentrations (<i>P</i> < 0.001). By contrast, the MS4A4A antibody treatment group displayed the opposite effect. Inhibition of MS4A4A enhanced the accumulation of macrophages, CD4<sup>+</sup> T cells, and CD8<sup>+</sup> T cells in the tumor (<i>P</i> < 0.001). Flow cytometry and qRT-PCR analyses showed that MS4A4A promoted M2 macrophage polarization, and MS4A4A antibody induced M1 polarization (<i>P</i> < 0.001). MS4A4A played a key role in inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells subunit p50 (p50) during M1 polarization. Furthermore, the PD-1 antibody reversed the pro-tumor effects of MS4A4A, which reestablished pro-inflammatory cytokine levels while lowering anti-inflammatory cytokines (<i>P</i> < 0.001).</p><p><strong>Conclusion: </strong>This study shows that MS4A4A promotes tumor growth by inducing M2 macrophage polarization and suppressing immune responses, while MS4A4A antibody enhances anti-tumor immunity by inducing M1 polarization. PD-1 antibody reverses MS4A4A's pro-tumor effects, restoring anti-tumor immunity. MS4A4A inhibitors or their combination with PD-1 antibodies may offer a promising strategy for GC immunotherapy.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"63"},"PeriodicalIF":3.1,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514763/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281612","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}
Pub Date : 2025-07-02eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_8_2025
Jinzhe Chu, Nannan Ren, Chunli Xie
Objective: Acute cerebral infarction (ACI) has a high incidence and complex etiology, so searching for specific diagnosis and therapeutic target molecules is highly important. This paper aimed to explore the mechanism of karyopherin alpha 4 (KPNA4) in ACI.
Material and methods: Four groups were established: sham group, middle cerebral artery occlusion (MCAO) group, MCAO + small interfering negative control (si-NC group), and MCAO + si-KPNA4 group. The Zea-Longa scoring standard was adopted to assess the neurological impairment of ACI rats. The expression of KPNA4 was verified by real-time quantity polymerase chain reaction assay and Western blot. 2,3,5-Triphenyltetrazolium chloride and hematoxylin and eosin staining were used to examine the effects of KPNA4 knockdown on brain injury in ACI rats. The apoptosis of oxygen-glucose deprivation (OGD)-SH-SY5Y cells was measured by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling method. Western blot analysis was performed to analyze the expression levels of apoptosis-related proteins, inflammatory factors, and nuclear factor kappa B (NF-κB) pathway-related proteins.
Results: KPNA4 was overexpressed in the MCAO rat brain (P < 0.0001) and OGD-SH-SY5Y cells (P < 0.0001). Knocking down KPNA4 significantly reduced the degree of brain damage (P < 0.0001) and reduced the apoptosis of OGD-SH-SY5Y cells (P < 0.001). KPNA4 knockdown also significantly decreased the expression levels of inflammatory factors (interleukin [IL]-1β, tumor necrosis factor-α, and IL-18; P < 0.0001) and prohibited the phosphorylation of the inhibitor of NF-κB and p65 (P < 0.0001). However, NF-κB agonist lipopolysaccharide reversed the inhibition of KPNA4 knockdown.
Conclusion: In general, KPNA4 knockdown can mitigate ACI-induced damage by regulating the NF-kB pathway. This finding provides a new perspective on the treatment of ACI.
{"title":"Preliminary investigation on the correlation between karyopherin alpha 4 and acute cerebral infarction.","authors":"Jinzhe Chu, Nannan Ren, Chunli Xie","doi":"10.25259/Cytojournal_8_2025","DOIUrl":"10.25259/Cytojournal_8_2025","url":null,"abstract":"<p><strong>Objective: </strong>Acute cerebral infarction (ACI) has a high incidence and complex etiology, so searching for specific diagnosis and therapeutic target molecules is highly important. This paper aimed to explore the mechanism of karyopherin alpha 4 (<i>KPNA4</i>) in ACI.</p><p><strong>Material and methods: </strong>Four groups were established: sham group, middle cerebral artery occlusion (MCAO) group, MCAO + small interfering negative control (si-NC group), and MCAO + si-<i>KPNA4</i> group. The Zea-Longa scoring standard was adopted to assess the neurological impairment of ACI rats. The expression of <i>KPNA4</i> was verified by real-time quantity polymerase chain reaction assay and Western blot. 2,3,5-Triphenyltetrazolium chloride and hematoxylin and eosin staining were used to examine the effects of <i>KPNA4</i> knockdown on brain injury in ACI rats. The apoptosis of oxygen-glucose deprivation (OGD)-SH-SY5Y cells was measured by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling method. Western blot analysis was performed to analyze the expression levels of apoptosis-related proteins, inflammatory factors, and nuclear factor kappa B (NF-κB) pathway-related proteins.</p><p><strong>Results: </strong><i>KPNA4</i> was overexpressed in the MCAO rat brain (<i>P</i> < 0.0001) and OGD-SH-SY5Y cells (<i>P</i> < 0.0001). Knocking down <i>KPNA4</i> significantly reduced the degree of brain damage (<i>P</i> < 0.0001) and reduced the apoptosis of OGD-SH-SY5Y cells (<i>P</i> < 0.001). <i>KPNA4</i> knockdown also significantly decreased the expression levels of inflammatory factors (interleukin [IL]-1β, tumor necrosis factor-α, and IL-18; <i>P</i> < 0.0001) and prohibited the phosphorylation of the inhibitor of NF-κB and p65 (<i>P</i> < 0.0001). However, NF-κB agonist lipopolysaccharide reversed the inhibition of <i>KPNA4</i> knockdown.</p><p><strong>Conclusion: </strong>In general, <i>KPNA4</i> knockdown can mitigate ACI-induced damage by regulating the NF-kB pathway. This finding provides a new perspective on the treatment of ACI.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"64"},"PeriodicalIF":3.1,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514764/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281582","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}
Pub Date : 2025-07-02eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_10_2025
Zhen Wang, Yong Wang, Yan Cheng, Jingwen Zhang, Wenyang Nie, Xueqiang Liu, Hualiang Deng
Objective: Coronary microvascular dysfunction following myocardial infarction (MI) serves as a critical factor affecting cardiac repair and functional recovery. Hyperhomocysteinemia (HHcy) has been closely associated with cardiovascular diseases, particularly in terms of its detrimental effects on microvasculature post-MI. Although transcription factor SP1 plays crucial roles in various physiological and pathological processes, its specific mechanism in the reversal of HHcy-induced microvascular dysfunction after MI remains unclear. The purpose of this study was to explore the possible mechanism of SP1 on HHcy-induced microvascular dysfunction.
Material and methods: This study utilized an HHcy mouse model and an in vitro model of human coronary artery endothelial cells (HCAECs) to systematically investigate the role of SP1 in post-MI microvascular dysfunction. Cardiac microvascular perfusion was assessed using fluorescein isothiocyanate (FITC)-labeled tomato lectin. Western blot analysis was employed to examine the expression levels of signal transducer and activator of transcription 3 (STAT3), conductance calcium-activated potassium channel protein 4 (KCNN4, also known as KCa3.1), and endothelial nitric oxide synthase (eNOS). The eNOS inhibitor N(ω)-nitro-L-arginine methyl ester (L-NAME) and STAT3 inhibitor Stattic were used to validate the signaling pathway.
Results: SP1 considerably improved microvascular dysfunction and angiogenic capacity in HHcy mice after MI. It enhanced cardiac microvascular function recovery by activating the STAT3/KCa3.1/eNOS signaling pathway. The eNOS inhibitor L-NAME reversed the protective effects of SP1, which indicates the crucial role of eNOS in SP1-mediated cardiovascular protection. Furthermore, SP1 alleviated homocysteine and hypoxia-induced cytotoxicity in HCAECs through this pathway, and the inhibition of the STAT3/KCa3.1/eNOS pathway blocked SP1's protective effects.
Conclusion: This study revealed for the first time the mechanism by which SP1 reverses HHcy-induced post-MI microvascular dysfunction through the activation of the STAT3/KCa3.1/eNOS pathway. The findings not only deepen our understanding of the pathological mechanisms of post-MI microvascular dysfunction but also provide an important theoretical basis for the development of new cardiovascular disease treatment strategies. SP1, as a potential therapeutic target, may play a crucial role in future cardiovascular disease treatments.
目的:心肌梗死(MI)后冠状动脉微血管功能障碍是影响心脏修复和功能恢复的重要因素。高同型半胱氨酸血症(HHcy)与心血管疾病密切相关,特别是其对心肌梗死后微血管的有害影响。虽然转录因子SP1在各种生理和病理过程中发挥重要作用,但其在逆转hhcy诱导的心肌梗死后微血管功能障碍中的具体机制尚不清楚。本研究的目的是探讨SP1在hhcy诱导的微血管功能障碍中的可能机制。材料与方法:本研究采用HHcy小鼠模型和人冠状动脉内皮细胞(HCAECs)体外模型,系统探讨SP1在心肌梗死后微血管功能障碍中的作用。采用异硫氰酸荧光素(FITC)标记的番茄凝集素评估心脏微血管灌注。Western blot检测信号转导和转录激活因子3 (STAT3)、电导钙活化钾通道蛋白4 (KCNN4,又称KCa3.1)、内皮型一氧化氮合酶(eNOS)的表达水平。使用eNOS抑制剂N(ω)-硝基- l -精氨酸甲酯(L-NAME)和STAT3抑制剂Stattic来验证信号通路。结果:SP1通过激活STAT3/KCa3.1/eNOS信号通路,显著改善心肌梗死后HHcy小鼠微血管功能障碍和血管生成能力。eNOS抑制剂L-NAME逆转SP1的保护作用,这表明eNOS在SP1介导的心血管保护中起着至关重要的作用。此外,SP1通过该通路减轻了同型半胱氨酸和缺氧诱导的hcaec细胞毒性,抑制STAT3/KCa3.1/eNOS通路阻断了SP1的保护作用。结论:本研究首次揭示了SP1通过激活STAT3/KCa3.1/eNOS通路逆转hhcy诱导的心肌梗死后微血管功能障碍的机制。这些发现不仅加深了我们对心肌梗死后微血管功能障碍病理机制的认识,也为制定新的心血管疾病治疗策略提供了重要的理论依据。SP1作为一个潜在的治疗靶点,可能在未来的心血管疾病治疗中发挥关键作用。
{"title":"Decoding post-myocardial infarction coronary microvascular dysfunction: The SP1-driven STAT3/KCa3.1/eNOS protective mechanism.","authors":"Zhen Wang, Yong Wang, Yan Cheng, Jingwen Zhang, Wenyang Nie, Xueqiang Liu, Hualiang Deng","doi":"10.25259/Cytojournal_10_2025","DOIUrl":"10.25259/Cytojournal_10_2025","url":null,"abstract":"<p><strong>Objective: </strong>Coronary microvascular dysfunction following myocardial infarction (MI) serves as a critical factor affecting cardiac repair and functional recovery. Hyperhomocysteinemia (HHcy) has been closely associated with cardiovascular diseases, particularly in terms of its detrimental effects on microvasculature post-MI. Although transcription factor SP1 plays crucial roles in various physiological and pathological processes, its specific mechanism in the reversal of HHcy-induced microvascular dysfunction after MI remains unclear. The purpose of this study was to explore the possible mechanism of SP1 on HHcy-induced microvascular dysfunction.</p><p><strong>Material and methods: </strong>This study utilized an HHcy mouse model and an <i>in vitro</i> model of human coronary artery endothelial cells (HCAECs) to systematically investigate the role of SP1 in post-MI microvascular dysfunction. Cardiac microvascular perfusion was assessed using fluorescein isothiocyanate (FITC)-labeled tomato lectin. Western blot analysis was employed to examine the expression levels of signal transducer and activator of transcription 3 (STAT3), conductance calcium-activated potassium channel protein 4 (KCNN4, also known as KCa3.1), and endothelial nitric oxide synthase (eNOS). The eNOS inhibitor N(ω)-nitro-L-arginine methyl ester (L-NAME) and STAT3 inhibitor Stattic were used to validate the signaling pathway.</p><p><strong>Results: </strong>SP1 considerably improved microvascular dysfunction and angiogenic capacity in HHcy mice after MI. It enhanced cardiac microvascular function recovery by activating the STAT3/KCa3.1/eNOS signaling pathway. The eNOS inhibitor L-NAME reversed the protective effects of SP1, which indicates the crucial role of eNOS in SP1-mediated cardiovascular protection. Furthermore, SP1 alleviated homocysteine and hypoxia-induced cytotoxicity in HCAECs through this pathway, and the inhibition of the STAT3/KCa3.1/eNOS pathway blocked SP1's protective effects.</p><p><strong>Conclusion: </strong>This study revealed for the first time the mechanism by which SP1 reverses HHcy-induced post-MI microvascular dysfunction through the activation of the STAT3/KCa3.1/eNOS pathway. The findings not only deepen our understanding of the pathological mechanisms of post-MI microvascular dysfunction but also provide an important theoretical basis for the development of new cardiovascular disease treatment strategies. SP1, as a potential therapeutic target, may play a crucial role in future cardiovascular disease treatments.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"66"},"PeriodicalIF":3.1,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514766/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281562","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}
Pub Date : 2025-07-02eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_35_2025
Hongjun Zhao, Yongjian Ji, Chaozhao Liang
Objective: Clear-cell renal cell carcinoma (ccRCC) is the most prevalent type of renal cancer. Solute carrier family 6 member 3 (SLC6A3) is associated with ccRCC. This research aimed to investigate the function of SLC6A3 in promoting the metabolism and development of ccRCC tumor cells.
Material and methods: SLC6A3 expression levels in ccRCC cell lines were assessed through quantitative real-time polymerase chain reaction and Western blot (WB). A stable overexpression of SLC6A3 was achieved in the 786-O cell line, and stable knockdown was established in the OS-RC-2 cell line through lentiviral transfection. Cell biological behavior was evaluated through flow cytometry, terminal deoxynucleotidyl transferase media dUTP nick end labeling staining, Cell counting kit-8 assays, and Transwell assays. Extracellular acidification rate measurements, WB, and biochemical assays were performed to detect cellular glycolysis and ferroptosis following alterations in SLC6A3 expression. The effects of SLC6A3 overexpression or knockdown on ccRCC tumor were further verified in xenograft models using nude mice.
Results: SLC6A3 was elevated in various ccRCC cell lines. The 786-O cells exhibited a relatively low baseline SLC6A3 expression (P < 0.01), which prompted subsequent overexpression experiments, whereas OSRC-2 cells showed the highest baseline expression (P < 0.001), which led to subsequent knockdown studies. The overexpression of SLC6A3 in 786-O cells increased the glycolytic activity, decreased ferroptosis levels, and promoted viability and invasion of ccRCC cells. Conversely, knockdown of SLC6A3 in OS-RC-2 cells reduced glycolytic activity, increased ferroptosis levels, and inhibited viability and invasion of ccRCC cells. All results were statistically significant. These findings were corroborated by in vivo experiments.
Conclusion: SLC6A3 is markedly overexpressed in ccRCC. It influences the promotion of tumor growth by enhancing glycolysis and suppressing ferroptosis. These insights highlight the potential of SLC6A3 for innovative therapeutic strategies in ccRCC management.
目的:透明细胞肾细胞癌(ccRCC)是最常见的肾癌类型。溶质载体家族6成员3 (SLC6A3)与ccRCC相关。本研究旨在探讨SLC6A3在促进ccRCC肿瘤细胞代谢和发育中的作用。材料与方法:采用实时定量聚合酶链反应(pcr)和Western blot (WB)技术检测SLC6A3在ccRCC细胞株中的表达水平。SLC6A3在786-O细胞系中稳定过表达,并通过慢病毒转染在OS-RC-2细胞系中稳定敲低。通过流式细胞术、末端脱氧核苷酸转移酶培养基dUTP缺口末端标记染色、细胞计数试剂盒-8检测和Transwell检测评估细胞生物学行为。在SLC6A3表达改变后,采用细胞外酸化率测量、WB和生化分析检测细胞糖酵解和铁凋亡。在裸鼠异种移植模型中进一步验证SLC6A3过表达或敲低对ccRCC肿瘤的影响。结果:SLC6A3在各种ccRCC细胞系中表达升高。786-O细胞显示出相对较低的SLC6A3基线表达(P < 0.01),这促使了随后的过表达实验,而OSRC-2细胞显示出最高的基线表达(P < 0.001),这导致了随后的敲低研究。SLC6A3在786-O细胞中的过表达增加了糖酵解活性,降低了铁凋亡水平,促进了ccRCC细胞的活力和侵袭性。相反,在OS-RC-2细胞中敲低SLC6A3可降低糖酵解活性,增加铁凋亡水平,抑制ccRCC细胞的活力和侵袭。所有结果均具有统计学意义。这些发现得到了体内实验的证实。结论:SLC6A3在ccRCC中明显过表达。它通过促进糖酵解和抑制铁下垂来促进肿瘤生长。这些发现突出了SLC6A3在ccRCC治疗中创新治疗策略的潜力。
{"title":"Solute carrier family 6 member 3 promotes the development of clear cell renal cell carcinoma by enhancing glycolysis and inhibiting ferroptosis.","authors":"Hongjun Zhao, Yongjian Ji, Chaozhao Liang","doi":"10.25259/Cytojournal_35_2025","DOIUrl":"10.25259/Cytojournal_35_2025","url":null,"abstract":"<p><strong>Objective: </strong>Clear-cell renal cell carcinoma (ccRCC) is the most prevalent type of renal cancer. Solute carrier family 6 member 3 (SLC6A3) is associated with ccRCC. This research aimed to investigate the function of SLC6A3 in promoting the metabolism and development of ccRCC tumor cells.</p><p><strong>Material and methods: </strong>SLC6A3 expression levels in ccRCC cell lines were assessed through quantitative real-time polymerase chain reaction and Western blot (WB). A stable overexpression of SLC6A3 was achieved in the 786-O cell line, and stable knockdown was established in the OS-RC-2 cell line through lentiviral transfection. Cell biological behavior was evaluated through flow cytometry, terminal deoxynucleotidyl transferase media dUTP nick end labeling staining, Cell counting kit-8 assays, and Transwell assays. Extracellular acidification rate measurements, WB, and biochemical assays were performed to detect cellular glycolysis and ferroptosis following alterations in SLC6A3 expression. The effects of SLC6A3 overexpression or knockdown on ccRCC tumor were further verified in xenograft models using nude mice.</p><p><strong>Results: </strong>SLC6A3 was elevated in various ccRCC cell lines. The 786-O cells exhibited a relatively low baseline SLC6A3 expression (<i>P</i> < 0.01), which prompted subsequent overexpression experiments, whereas OSRC-2 cells showed the highest baseline expression (<i>P</i> < 0.001), which led to subsequent knockdown studies. The overexpression of SLC6A3 in 786-O cells increased the glycolytic activity, decreased ferroptosis levels, and promoted viability and invasion of ccRCC cells. Conversely, knockdown of SLC6A3 in OS-RC-2 cells reduced glycolytic activity, increased ferroptosis levels, and inhibited viability and invasion of ccRCC cells. All results were statistically significant. These findings were corroborated by in vivo experiments.</p><p><strong>Conclusion: </strong>SLC6A3 is markedly overexpressed in ccRCC. It influences the promotion of tumor growth by enhancing glycolysis and suppressing ferroptosis. These insights highlight the potential of SLC6A3 for innovative therapeutic strategies in ccRCC management.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"65"},"PeriodicalIF":3.1,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514767/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281519","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}
Pub Date : 2025-07-02eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_176_2024
Beibei Ren, Bing Zhang, Xiangnan Jiang, He Zhang, Qingxin Xia
Metaplastic breast carcinoma comprises a heterogeneous group of morphologic variants characterized by markedly aggressive pathological behavior and diverse histological subtypes. Accurate diagnosis of metaplastic breast carcinoma is essential for implementing effective and individualized treatment approaches; however, its differentiation from similar tumors may occasionally be challenging. We report two cases of breast tumors diagnosed using comprehensive tumor genome analysis.
{"title":"The application of tumor origin genetic testing in differential diagnosis of breast metaplastic carcinoma.","authors":"Beibei Ren, Bing Zhang, Xiangnan Jiang, He Zhang, Qingxin Xia","doi":"10.25259/Cytojournal_176_2024","DOIUrl":"10.25259/Cytojournal_176_2024","url":null,"abstract":"<p><p>Metaplastic breast carcinoma comprises a heterogeneous group of morphologic variants characterized by markedly aggressive pathological behavior and diverse histological subtypes. Accurate diagnosis of metaplastic breast carcinoma is essential for implementing effective and individualized treatment approaches; however, its differentiation from similar tumors may occasionally be challenging. We report two cases of breast tumors diagnosed using comprehensive tumor genome analysis.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"67"},"PeriodicalIF":3.1,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12514762/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281505","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}
Pub Date : 2025-06-14eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_21_2025
Anna M Dmitrieva, Ilayda G Kocak, Lydia Meder
Lung cancer is the leading cause of cancer-related deaths worldwide, with genetic- and protein-based diagnostics playing a crucial role in disease detection and improving patient outcomes. Glycosylation, a major post-translational modification, has recently emerged as a factor influencing cancer progression, immune evasion, and therapeutic resistance. Aberrant glycosylation patterns, particularly among receptor tyrosine kinases (RTKs), have been shown to modulate oncogenic signaling pathways and influence tumor growth. This review provides a comprehensive overview of how glycosylation alterations affect the stability, function, and therapeutic targeting of key RTKs relevant in lung adenocarcinoma: Epidermal growth factor receptor, human epidermal growth factor receptor 2, and cellular mesenchymal-epithelial transition factor, rearranged during transfection, anaplastic lymphoma kinase, and ROS proto-oncogene 1 receptor tyrosine kinase. Despite substantial advances in targeted therapies, initial and acquired resistance remain a major challenge in the treatment of lung cancer. There is growing evidence that strategies targeting glycosylation can be combined with established treatment protocols to help overcome resistance. Finally, we propose future directions for the advancement of glycosylation-based approaches to improve precision medicine.
{"title":"Aberrations in the glycosylation of receptor tyrosine kinases: A focus on lung adenocarcinoma.","authors":"Anna M Dmitrieva, Ilayda G Kocak, Lydia Meder","doi":"10.25259/Cytojournal_21_2025","DOIUrl":"10.25259/Cytojournal_21_2025","url":null,"abstract":"<p><p>Lung cancer is the leading cause of cancer-related deaths worldwide, with genetic- and protein-based diagnostics playing a crucial role in disease detection and improving patient outcomes. Glycosylation, a major post-translational modification, has recently emerged as a factor influencing cancer progression, immune evasion, and therapeutic resistance. Aberrant glycosylation patterns, particularly among receptor tyrosine kinases (RTKs), have been shown to modulate oncogenic signaling pathways and influence tumor growth. This review provides a comprehensive overview of how glycosylation alterations affect the stability, function, and therapeutic targeting of key RTKs relevant in lung adenocarcinoma: Epidermal growth factor receptor, human epidermal growth factor receptor 2, and cellular mesenchymal-epithelial transition factor, rearranged during transfection, anaplastic lymphoma kinase, and ROS proto-oncogene 1 receptor tyrosine kinase. Despite substantial advances in targeted therapies, initial and acquired resistance remain a major challenge in the treatment of lung cancer. There is growing evidence that strategies targeting glycosylation can be combined with established treatment protocols to help overcome resistance. Finally, we propose future directions for the advancement of glycosylation-based approaches to improve precision medicine.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"62"},"PeriodicalIF":3.1,"publicationDate":"2025-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12289112/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144709646","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}
Pub Date : 2025-06-14eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_53_2025
Wanzhen Yang, Junyi Tu, Kaixi Dai, Kemin Jia
Objective: This study aims to explore the role of microfibrillar-associated protein-2 (MFAP2) in oral squamous cell carcinoma (OSCC).
Material and methods: Analysis of MFAP2 expression in diverse cancers and its relationship with head-and-neck squamous cell carcinoma (HNSC) prognosis. MFAP2 abundance was identified in OSCC cells and in human oral epithelial cells using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays. Knockdown and overexpression techniques were utilized to examine the mechanism by which MFAP2 and nuclear factor erythroid 2-related factor 2 (NRF2) affect OSCC malignancy. Cell viability, proliferation, and apoptosis were assessed using cell counting kit-8, colony formation, flow cytometry, wound healing, and Transwell tests. Messenger ribonucleic acid expression was detected using qRT-PCR, whereas protein level was analyzed using Western blot.
Results: MFAP2 and Kelch-like E3 ubiquitin ligase (ECH)-associated protein 1 (KEAP1) had high expression levels in numerous tumors, including OSCC, and the high expression level of MFAP2 was associated with unfavorable HNSC outcomes. MFAP2 was abundantly expressed in five OSCC cell lines, with the peak expression observed in squamous cell carcinoma (SCC)-15 and SCC-9 cells, making them suitable for subsequent studies. MFAP2 knockdown hindered the proliferative and mobile capacity of OSCC cells, yet it supported cell apoptosis. MFAP2 silencing led to a notable drop in KEAP1 and NRF2 expression levels in OSCC cells. NRF2 overexpression could counteract the effects of MFAP2 knockout, which included diminished proliferation and movement and heightened apoptosis in OSCC cells.
Conclusion: The results of this study indicated that MFAP2 facilitated the malignant progression of OSCC and provided insights into the downstream regulatory mechanism of the KEAP1/NRF2 axis, highlighting the potential application of MFAP2 in OSCC management.
{"title":"Microfibrillar-associated protein-2 facilitates aggressive progression of oral squamous cell carcinoma cells through Kelch-like E3 ubiquitin ligase-associated protein 1/nuclear factor erythroid 2-related factor 2 signaling pathway.","authors":"Wanzhen Yang, Junyi Tu, Kaixi Dai, Kemin Jia","doi":"10.25259/Cytojournal_53_2025","DOIUrl":"10.25259/Cytojournal_53_2025","url":null,"abstract":"<p><strong>Objective: </strong>This study aims to explore the role of microfibrillar-associated protein-2 (MFAP2) in oral squamous cell carcinoma (OSCC).</p><p><strong>Material and methods: </strong>Analysis of MFAP2 expression in diverse cancers and its relationship with head-and-neck squamous cell carcinoma (HNSC) prognosis. MFAP2 abundance was identified in OSCC cells and in human oral epithelial cells using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays. Knockdown and overexpression techniques were utilized to examine the mechanism by which MFAP2 and nuclear factor erythroid 2-related factor 2 (NRF2) affect OSCC malignancy. Cell viability, proliferation, and apoptosis were assessed using cell counting kit-8, colony formation, flow cytometry, wound healing, and Transwell tests. Messenger ribonucleic acid expression was detected using qRT-PCR, whereas protein level was analyzed using Western blot.</p><p><strong>Results: </strong>MFAP2 and Kelch-like E3 ubiquitin ligase (ECH)-associated protein 1 (KEAP1) had high expression levels in numerous tumors, including OSCC, and the high expression level of MFAP2 was associated with unfavorable HNSC outcomes. MFAP2 was abundantly expressed in five OSCC cell lines, with the peak expression observed in squamous cell carcinoma (SCC)-15 and SCC-9 cells, making them suitable for subsequent studies. MFAP2 knockdown hindered the proliferative and mobile capacity of OSCC cells, yet it supported cell apoptosis. MFAP2 silencing led to a notable drop in KEAP1 and NRF2 expression levels in OSCC cells. NRF2 overexpression could counteract the effects of MFAP2 knockout, which included diminished proliferation and movement and heightened apoptosis in OSCC cells.</p><p><strong>Conclusion: </strong>The results of this study indicated that MFAP2 facilitated the malignant progression of OSCC and provided insights into the downstream regulatory mechanism of the KEAP1/NRF2 axis, highlighting the potential application of MFAP2 in OSCC management.</p>","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"61"},"PeriodicalIF":3.1,"publicationDate":"2025-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12289114/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144709650","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}
Pub Date : 2025-06-13eCollection Date: 2025-01-01DOI: 10.25259/Cytojournal_57_2025
Suwen Chang
<p><strong>Objective: </strong>This study aims to explore the modulatory effect of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) protein in the Notch1 signaling pathway in cervical cancer (CC) cells and assess how this modulation affects the proliferation and migration of CC cells. Moreover, this study offers fresh perspectives on the molecular mechanisms underlying CC by thoroughly analyzing the relationship between 15-PGDH and the Notch1 signaling pathway, and investigates the therapeutic potential of 15-PGDH.</p><p><strong>Material and methods: </strong>Human normal cervical epithelial cells and CC cell lines (human CC cell line [HeLa], human cervical squamous carcinoma cell line [Caski], and human cervical epidermoid carcinoma cells [ME180]) were selected as experimental models. Western blotting (WB) and quantitative reverse transcription polymerase chain reaction were performed to evaluate the protein and messenger RNA levels of 15-PGDH and Notch receptor 1 (Notch1) signaling pathway-related proteins (Jagged canonical Notch ligand 1 [Jagged1] and Hes family bHLH transcription factor 1 [Hes1]). Results suggested that the HeLa and Caski cells exhibited significant expression of 15-PGDH and Notch1 signaling-related proteins. A series of experiments, including WB, cell counting kit-8 assay, Transwell migration assay, and 5-ethynyl-2'-deoxyuridine assay, was conducted in the HeLa and Caski cells to obtain an extensive understanding of how 15-PGDH influences Notch1 signaling regulation. This study also utilized the 15-PGDH inhibitor SW033291 and a Notch1 overexpression vector to evaluate the effect of 15-PGDH on CC cell growth, motility, and Notch1 signaling pathway modulation.</p><p><strong>Results: </strong>Results demonstrated that in the normal human cervical epithelial cells, 15-PGDH was highly expressed, while the Notch1 signaling pathway-related proteins exhibited low expression quantities. However, in HeLa and Caski CC cells, 15-PGDH expression was significantly downregulated (<i>P</i> < 0.001 or <i>P</i> < 0.01), whereas the Notch1 signaling pathway was activated. Further studies revealed that 15-PGDH or its inhibitor influenced the stimulation of the Notch1 signaling pathway in the HeLa and Caski cells. Specifically, the 15-PGDH inhibitor SW033291 reduced 15-PGDH expression (<i>P</i> < 0.001 or <i>P</i> < 0.01) and promoted Notch signaling activation. Meanwhile, 15-PGDH upregulation suppressed Notch signaling activation. Furthermore, 15-PGDH successfully prevented the proliferation and migration of CC cells induced by Notch1 overexpression and reduced the activation of the Notch signaling pathway, as shown by the downregulation of Notch1 and its downstream effectors, Jagged1 and Hes1.</p><p><strong>Conclusion: </strong>This study highlights the role of 15-PGDH in regulating the Notch1 signaling pathway in CC cells, focusing on its effect on cell proliferation and migration. The results demonstrate that 15-PGDH suppresses CC cell prolif
{"title":"Mechanism of 15-hydroxyprostaglandin dehydrogenase protein inhibiting cervical cancer cell proliferation through downregulation of the notch1 signaling pathway.","authors":"Suwen Chang","doi":"10.25259/Cytojournal_57_2025","DOIUrl":"10.25259/Cytojournal_57_2025","url":null,"abstract":"<p><strong>Objective: </strong>This study aims to explore the modulatory effect of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) protein in the Notch1 signaling pathway in cervical cancer (CC) cells and assess how this modulation affects the proliferation and migration of CC cells. Moreover, this study offers fresh perspectives on the molecular mechanisms underlying CC by thoroughly analyzing the relationship between 15-PGDH and the Notch1 signaling pathway, and investigates the therapeutic potential of 15-PGDH.</p><p><strong>Material and methods: </strong>Human normal cervical epithelial cells and CC cell lines (human CC cell line [HeLa], human cervical squamous carcinoma cell line [Caski], and human cervical epidermoid carcinoma cells [ME180]) were selected as experimental models. Western blotting (WB) and quantitative reverse transcription polymerase chain reaction were performed to evaluate the protein and messenger RNA levels of 15-PGDH and Notch receptor 1 (Notch1) signaling pathway-related proteins (Jagged canonical Notch ligand 1 [Jagged1] and Hes family bHLH transcription factor 1 [Hes1]). Results suggested that the HeLa and Caski cells exhibited significant expression of 15-PGDH and Notch1 signaling-related proteins. A series of experiments, including WB, cell counting kit-8 assay, Transwell migration assay, and 5-ethynyl-2'-deoxyuridine assay, was conducted in the HeLa and Caski cells to obtain an extensive understanding of how 15-PGDH influences Notch1 signaling regulation. This study also utilized the 15-PGDH inhibitor SW033291 and a Notch1 overexpression vector to evaluate the effect of 15-PGDH on CC cell growth, motility, and Notch1 signaling pathway modulation.</p><p><strong>Results: </strong>Results demonstrated that in the normal human cervical epithelial cells, 15-PGDH was highly expressed, while the Notch1 signaling pathway-related proteins exhibited low expression quantities. However, in HeLa and Caski CC cells, 15-PGDH expression was significantly downregulated (<i>P</i> < 0.001 or <i>P</i> < 0.01), whereas the Notch1 signaling pathway was activated. Further studies revealed that 15-PGDH or its inhibitor influenced the stimulation of the Notch1 signaling pathway in the HeLa and Caski cells. Specifically, the 15-PGDH inhibitor SW033291 reduced 15-PGDH expression (<i>P</i> < 0.001 or <i>P</i> < 0.01) and promoted Notch signaling activation. Meanwhile, 15-PGDH upregulation suppressed Notch signaling activation. Furthermore, 15-PGDH successfully prevented the proliferation and migration of CC cells induced by Notch1 overexpression and reduced the activation of the Notch signaling pathway, as shown by the downregulation of Notch1 and its downstream effectors, Jagged1 and Hes1.</p><p><strong>Conclusion: </strong>This study highlights the role of 15-PGDH in regulating the Notch1 signaling pathway in CC cells, focusing on its effect on cell proliferation and migration. The results demonstrate that 15-PGDH suppresses CC cell prolif","PeriodicalId":49082,"journal":{"name":"Cytojournal","volume":"22 ","pages":"59"},"PeriodicalIF":3.1,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12289113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144709649","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}