Gastric-type adenocarcinoma (GAS) of the uterine cervix is a rare and aggressive subtype of cervical adenocarcinoma characterized by intrinsic resistance to chemotherapy and poor clinical outcomes due to the lack of effective treatment options. To address this critical unmet need, we established a novel GAS-derived cell line, KGAS, from ascitic fluid collected from a patient with recurrent GAS. Short tandem repeat (STR) analysis confirmed the genetic identity between the primary tumor and the cell line. Upon transplantation into immunocompromised mice, KGAS cells formed tumors that expressed Claudin-18 and MUC6, clinically recognized markers of GAS. Furthermore, KGAS cells exhibited marked resistance to paclitaxel and carboplatin, showing significantly reduced growth inhibition compared to HeLa cells. We also established a paclitaxel- and carboplatin-resistant subline, rKGAS, and performed microRNA (miRNA) sequencing to explore the molecular basis of acquired chemoresistance. Seventeen differentially expressed miRNAs were identified between KGAS and rKGAS cells. Upregulated miRNAs in rKGAS were predicted to target oncogenes such as BCL2, MET, SIRT1, and VEGFA, whereas downregulated miRNAs were associated with tumor suppressor genes, including IGF1R, TNFAIP3, and MTOR. The KGAS and rKGAS cell lines represent valuable preclinical models for elucidating the molecular mechanisms of chemoresistance and malignant progression in cervical GAS, and may contribute to the development of novel therapeutic strategies for this challenging cancer subtype.
{"title":"Establishment of KGAS, a cell line derived from gastric-type adenocarcinoma of the uterine cervix.","authors":"Hiroaki Yamada, Akira Yokoi, Eri Asano-Inami, Masami Kitagawa, Kosuke Yoshida, Kazuhiro Suzuki, Shin Nishio, Hiroaki Kajiyama, Naotake Tsuda","doi":"10.1007/s13577-025-01286-9","DOIUrl":"10.1007/s13577-025-01286-9","url":null,"abstract":"<p><p>Gastric-type adenocarcinoma (GAS) of the uterine cervix is a rare and aggressive subtype of cervical adenocarcinoma characterized by intrinsic resistance to chemotherapy and poor clinical outcomes due to the lack of effective treatment options. To address this critical unmet need, we established a novel GAS-derived cell line, KGAS, from ascitic fluid collected from a patient with recurrent GAS. Short tandem repeat (STR) analysis confirmed the genetic identity between the primary tumor and the cell line. Upon transplantation into immunocompromised mice, KGAS cells formed tumors that expressed Claudin-18 and MUC6, clinically recognized markers of GAS. Furthermore, KGAS cells exhibited marked resistance to paclitaxel and carboplatin, showing significantly reduced growth inhibition compared to HeLa cells. We also established a paclitaxel- and carboplatin-resistant subline, rKGAS, and performed microRNA (miRNA) sequencing to explore the molecular basis of acquired chemoresistance. Seventeen differentially expressed miRNAs were identified between KGAS and rKGAS cells. Upregulated miRNAs in rKGAS were predicted to target oncogenes such as BCL2, MET, SIRT1, and VEGFA, whereas downregulated miRNAs were associated with tumor suppressor genes, including IGF1R, TNFAIP3, and MTOR. The KGAS and rKGAS cell lines represent valuable preclinical models for elucidating the molecular mechanisms of chemoresistance and malignant progression in cervical GAS, and may contribute to the development of novel therapeutic strategies for this challenging cancer subtype.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 6","pages":"159"},"PeriodicalIF":3.1,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12436518/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145066149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The Switch/Sucrose Nonfermentable (SWI/SNF) complexes are chromatin remodeling factors that consist of multiple protein subunits. Each subunit plays a distinct role in gene regulation and is aberrantly expressed in tumors, such as neuroendocrine neoplasms (NENs). BRG1-associated factor 53B (BAF53B), which is also known as ACTL6B, is a neuron-specific subunit that acts as a regulator during neurogenesis. Because the BAF53B expression pattern in tumors is unknown, the present study investigated the expression in cell lines and tissues. Publicly available transcriptome data indicated that BAF53B mRNA was highly expressed in NEN-derived cell lines. We performed immunohistochemical staining on tissue microarrays of different types of NENs with neuroendocrine (NE) marker expression (n = 117) (small cell lung carcinoma (SCLC)lung carcinoid (LC), gastroenteropancreatic-NEN (GEP-NEN), esophageal neuroendocrine carcinoma (ENEC), medullary thyroid carcinoma (MTC), neuroblastoma (NB), and pheochromocytoma (PHEO)) and non-NENs (n = 178). While few positive cells were observed in many cases of non-NENs (e.g., lung adenocarcinoma), positive expression was found in cases of NENs (SCLC (14/19, 73.7%), LC (12/16, 75.0%), GEP-NEN (4/9, 44.4%), ENEC (1/2, 50.0%), MTC (24/27, 88.9%), NB (18/20, 90.0%), and PHEO (16/24, 66.7%)). In NCI-H889 cells, BAF53B knockdown did not affect the cellular viability, and its effect on NE marker expression was only marginal. However, a gene expression microarray analysis suggested that BAF53B-regulated genes were associated with the development and progression of NENs. Our analysis revealed that BAF53B was an immunohistochemical marker for specific NENs, indicating its potentially important role in the pathogenesis.
{"title":"The SWI/SNF chromatin remodeling complex subunit BAF53B as an immunohistochemical marker for neuroendocrine neoplasms.","authors":"Kouhei Sakurai, Mako Ochiai, Kanata Kojima, Kento Kato, Tatsuya Ando, Taku Kato, Hiroyasu Ito","doi":"10.1007/s13577-025-01289-6","DOIUrl":"10.1007/s13577-025-01289-6","url":null,"abstract":"<p><p>The Switch/Sucrose Nonfermentable (SWI/SNF) complexes are chromatin remodeling factors that consist of multiple protein subunits. Each subunit plays a distinct role in gene regulation and is aberrantly expressed in tumors, such as neuroendocrine neoplasms (NENs). BRG1-associated factor 53B (BAF53B), which is also known as ACTL6B, is a neuron-specific subunit that acts as a regulator during neurogenesis. Because the BAF53B expression pattern in tumors is unknown, the present study investigated the expression in cell lines and tissues. Publicly available transcriptome data indicated that BAF53B mRNA was highly expressed in NEN-derived cell lines. We performed immunohistochemical staining on tissue microarrays of different types of NENs with neuroendocrine (NE) marker expression (n = 117) (small cell lung carcinoma (SCLC)lung carcinoid (LC), gastroenteropancreatic-NEN (GEP-NEN), esophageal neuroendocrine carcinoma (ENEC), medullary thyroid carcinoma (MTC), neuroblastoma (NB), and pheochromocytoma (PHEO)) and non-NENs (n = 178). While few positive cells were observed in many cases of non-NENs (e.g., lung adenocarcinoma), positive expression was found in cases of NENs (SCLC (14/19, 73.7%), LC (12/16, 75.0%), GEP-NEN (4/9, 44.4%), ENEC (1/2, 50.0%), MTC (24/27, 88.9%), NB (18/20, 90.0%), and PHEO (16/24, 66.7%)). In NCI-H889 cells, BAF53B knockdown did not affect the cellular viability, and its effect on NE marker expression was only marginal. However, a gene expression microarray analysis suggested that BAF53B-regulated genes were associated with the development and progression of NENs. Our analysis revealed that BAF53B was an immunohistochemical marker for specific NENs, indicating its potentially important role in the pathogenesis.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 6","pages":"158"},"PeriodicalIF":3.1,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058420","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Myocardial fibrosis is a complex pathological process that often leads to myocardial dysfunction, heart failure, and ultimately, death. A critical contributor to the development of cardiac fibrosis is the endothelial-to-mesenchymal transition (EndMT). Apigenin, a natural compound derived from Matricaria chamomilla, has shown potential anti-fibrotic effects, although its precise mechanism of action is not fully understood. This study investigated the effects of apigenin (API) on EndMT and myocardial fibrosis using an in vitro human coronary artery endothelial cell EndMT model and an in vivo animal model of fibrosis. At appropriate concentrations, apigenin significantly inhibited TGF-β1-induced EndMT and myocardial fibrosis without affecting cell viability. Mechanistically, we found that apigenin suppressed ribosome biogenesis in coronary endothelial cells. Through differential gene screening, GTP-binding protein 4 (GTPBP4) was identified as a key target gene regulating ribosome biogenesis during the progression of myocardial fibrosis. Our results indicate that GTPBP4 plays a pivotal role in the apigenin-mediated inhibition of both ribosome biogenesis and EndMT in these cells. By downregulating GTPBP4 expression, apigenin suppressed EndMT, alleviated myocardial fibrosis, improved cardiac function, and reduced biomarkers of myocardial injury. These findings demonstrate for the first time that apigenin mitigates myocardial fibrosis and EndMT by inhibiting GTPBP4 expression, positioning apigenin as a promising therapeutic candidate for the prevention and treatment of myocardial fibrosis.
{"title":"Apigenin inhibits endothelial-to-mesenchymal transition of coronary artery endothelial cells and myocardial fibrosis by regulating ribosome biogenesis through GTPBP4 modulation.","authors":"Xiaoyong Hu, Zhaoying Yang, Rui Tang, Zhongying Lv, Ting Zou, Yuanyuan Dou, Hongjian Li","doi":"10.1007/s13577-025-01291-y","DOIUrl":"10.1007/s13577-025-01291-y","url":null,"abstract":"<p><p>Myocardial fibrosis is a complex pathological process that often leads to myocardial dysfunction, heart failure, and ultimately, death. A critical contributor to the development of cardiac fibrosis is the endothelial-to-mesenchymal transition (EndMT). Apigenin, a natural compound derived from Matricaria chamomilla, has shown potential anti-fibrotic effects, although its precise mechanism of action is not fully understood. This study investigated the effects of apigenin (API) on EndMT and myocardial fibrosis using an in vitro human coronary artery endothelial cell EndMT model and an in vivo animal model of fibrosis. At appropriate concentrations, apigenin significantly inhibited TGF-β1-induced EndMT and myocardial fibrosis without affecting cell viability. Mechanistically, we found that apigenin suppressed ribosome biogenesis in coronary endothelial cells. Through differential gene screening, GTP-binding protein 4 (GTPBP4) was identified as a key target gene regulating ribosome biogenesis during the progression of myocardial fibrosis. Our results indicate that GTPBP4 plays a pivotal role in the apigenin-mediated inhibition of both ribosome biogenesis and EndMT in these cells. By downregulating GTPBP4 expression, apigenin suppressed EndMT, alleviated myocardial fibrosis, improved cardiac function, and reduced biomarkers of myocardial injury. These findings demonstrate for the first time that apigenin mitigates myocardial fibrosis and EndMT by inhibiting GTPBP4 expression, positioning apigenin as a promising therapeutic candidate for the prevention and treatment of myocardial fibrosis.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 6","pages":"157"},"PeriodicalIF":3.1,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145042047","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-07DOI: 10.1007/s13577-025-01285-w
Huirui Liu, Yuting Ouyang, Hongyan Ge
Age-related eye diseases (AREDs) are the leading cause of visual impairment in the elderly, affecting the structure of the anterior and posterior segments of the eye, significantly reducing the quality of life of patients, and even leading to irreversible blindness. Typical AREDs include age-related cataract (ARC), dry eye disease (DED), age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), the global prevalence of which continues to rise, becoming a serious public health concern. SIRT1 is an NAD + dependent deacetylase, which plays an important physiological regulatory role in ocular tissues, mainly affecting gene expression and various cellular processes by regulating the acetylation status of substrate proteins. Studies have shown that SIRT1 plays a key role in oxidative stress, inflammation, autophagy, apoptosis and metabolism, and its expression or activity decreases can accelerate cell senescence and promote the occurrence and development of AREDs. In addition, SIRT1 expression levels and changes in its activity have been shown to be strongly associated with AREDs, making it a potential target for disease intervention and therapy. Therefore, this review systematically summarizes the biological role and regulatory mechanism of SIRT1 in AREDs, and explored its potential value as a therapeutic target, providing theoretical basis for future drug development and clinical transformation.
{"title":"SIRT1 as a potential target for age-related eye diseases: mechanisms and therapeutic strategies.","authors":"Huirui Liu, Yuting Ouyang, Hongyan Ge","doi":"10.1007/s13577-025-01285-w","DOIUrl":"https://doi.org/10.1007/s13577-025-01285-w","url":null,"abstract":"<p><p>Age-related eye diseases (AREDs) are the leading cause of visual impairment in the elderly, affecting the structure of the anterior and posterior segments of the eye, significantly reducing the quality of life of patients, and even leading to irreversible blindness. Typical AREDs include age-related cataract (ARC), dry eye disease (DED), age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), the global prevalence of which continues to rise, becoming a serious public health concern. SIRT1 is an NAD + dependent deacetylase, which plays an important physiological regulatory role in ocular tissues, mainly affecting gene expression and various cellular processes by regulating the acetylation status of substrate proteins. Studies have shown that SIRT1 plays a key role in oxidative stress, inflammation, autophagy, apoptosis and metabolism, and its expression or activity decreases can accelerate cell senescence and promote the occurrence and development of AREDs. In addition, SIRT1 expression levels and changes in its activity have been shown to be strongly associated with AREDs, making it a potential target for disease intervention and therapy. Therefore, this review systematically summarizes the biological role and regulatory mechanism of SIRT1 in AREDs, and explored its potential value as a therapeutic target, providing theoretical basis for future drug development and clinical transformation.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 6","pages":"155"},"PeriodicalIF":3.1,"publicationDate":"2025-09-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008544","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-31DOI: 10.1007/s13577-025-01284-x
Zhaoyu Liu, Xiang Song, Zhuang Liu, Xiaoyu He, Hong Gao, Xunru Liu
B-cell non-Hodgkin lymphoma (B-NHL) is a diverse group of aggressive lymphoid malignancies characterized by its molecular complexity. This study investigated the role of the upstream stimulatory factor 1 (USF1)-ribosomal protein S6 kinase B2 (RPS6KB2) axis in B-NHL progression through the AKT/HDM2 (also known as MDM2)/p53 signaling pathway. Using data from the GEO database, RPS6KB2 was identified to be overexpressed in B-NHL, which was confirmed by RT-qPCR, immunohistochemistry, and western blotting in both B-NHL tissues and cell lines. Functional studies revealed that RPS6KB2 knockdown reduced cell proliferation, migration, and tumor growth, while promoting apoptosis, effects that could be reversed by the AKT activator SC79. Bioinformatics analysis showed that USF1 activated the transcription of RPS6KB2 by directly binding to its promoter region. USF1 downregulation inhibited B-NHL progression, which was rescued by RPS6KB2 overexpression. These findings suggest that the USF1-RPS6KB2 axis contributes to B-NHL progression by activating the AKT/HDM2/p53 pathway.
{"title":"USF1-induced RPS6KB2 activation influences aggressive phenotype in B-cell non-Hodgkin lymphoma.","authors":"Zhaoyu Liu, Xiang Song, Zhuang Liu, Xiaoyu He, Hong Gao, Xunru Liu","doi":"10.1007/s13577-025-01284-x","DOIUrl":"10.1007/s13577-025-01284-x","url":null,"abstract":"<p><p>B-cell non-Hodgkin lymphoma (B-NHL) is a diverse group of aggressive lymphoid malignancies characterized by its molecular complexity. This study investigated the role of the upstream stimulatory factor 1 (USF1)-ribosomal protein S6 kinase B2 (RPS6KB2) axis in B-NHL progression through the AKT/HDM2 (also known as MDM2)/p53 signaling pathway. Using data from the GEO database, RPS6KB2 was identified to be overexpressed in B-NHL, which was confirmed by RT-qPCR, immunohistochemistry, and western blotting in both B-NHL tissues and cell lines. Functional studies revealed that RPS6KB2 knockdown reduced cell proliferation, migration, and tumor growth, while promoting apoptosis, effects that could be reversed by the AKT activator SC79. Bioinformatics analysis showed that USF1 activated the transcription of RPS6KB2 by directly binding to its promoter region. USF1 downregulation inhibited B-NHL progression, which was rescued by RPS6KB2 overexpression. These findings suggest that the USF1-RPS6KB2 axis contributes to B-NHL progression by activating the AKT/HDM2/p53 pathway.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 5","pages":"154"},"PeriodicalIF":3.1,"publicationDate":"2025-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144975999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-27DOI: 10.1007/s13577-025-01273-0
Yong Li, Yang Yue, Chaoqun Cen, Bo Liu
miR-487a-3p shows the potential of modulating adipose-derived stem cells (ADSCs) differentiation. This study aimed to investigate the mechanism of miR-487a-3p on the osteogenic differentiation of ADSCs. In this work, ADSCs were induced to differentiate into osteoblasts. miR-487a-3p were regulated by miRNA mimics or inhibitors in ADSCs. Wnt family member 5A (WNT5A) siRNA was used to reverse miR-487a-3p inhibitor-induced effects on WNT5A expression in ADSCs. Fat mass and obesity-associated protein (FTO) in ADSCs were altered by shRNAs or overexpression vectors. Calcium nodule, ALP activity, and biomarkers of osteogenic differentiation (OD) were investigated. Rats received ovariectomy (OVX) to construct osteoporosis (OP) model, followed by ADSCs transplantation. Histopathological changes, bone histomorphometry, and detection of OD biomarkers were performed. We found that osteogenesis induced a decrease in miR-487a-3p expression and an increase in FTO expression. miR-487a-3p upregulation inhibited the OD of ADSCs, including decreases in calcium nodule formation, ALP activity, and OD biomarkers. miR-487a-3p downregulation showed the opposite role in OD. miR-487a-3p negatively regulated WNT5A in ADSCs. WNT5A silence reversed the effect of miR-487a-3p downregulation on OD. FTO silence caused the increase in m6A of pri-miR-487a. FTO overexpression inhibited DGCR8 recruitment in pri-miR-487a, and reversed the effect of miR-487a-3p upregulation in OD. ADSCs transplantation improved OP symptom in rats, including improvement of femur tissue, increase in percent bone volume and trabecular number, and upregulation in OD biomarkers. miR-487a-3p downregulation enhanced the therapeutic role of ADSC in rats with OP. Collectively, FTO regulated pri-miR-487a maturation via m6A-dependent pathway, which altered the WNT5A-mediated osteogenesis of ADSCs.
{"title":"FTO inhibited miR-487a-3p biosynthesis via N6-methyladenosine-dependent pathway to promote WNT5A-mediated osteogenic differentiation of adipose-derived stem cells.","authors":"Yong Li, Yang Yue, Chaoqun Cen, Bo Liu","doi":"10.1007/s13577-025-01273-0","DOIUrl":"10.1007/s13577-025-01273-0","url":null,"abstract":"<p><p>miR-487a-3p shows the potential of modulating adipose-derived stem cells (ADSCs) differentiation. This study aimed to investigate the mechanism of miR-487a-3p on the osteogenic differentiation of ADSCs. In this work, ADSCs were induced to differentiate into osteoblasts. miR-487a-3p were regulated by miRNA mimics or inhibitors in ADSCs. Wnt family member 5A (WNT5A) siRNA was used to reverse miR-487a-3p inhibitor-induced effects on WNT5A expression in ADSCs. Fat mass and obesity-associated protein (FTO) in ADSCs were altered by shRNAs or overexpression vectors. Calcium nodule, ALP activity, and biomarkers of osteogenic differentiation (OD) were investigated. Rats received ovariectomy (OVX) to construct osteoporosis (OP) model, followed by ADSCs transplantation. Histopathological changes, bone histomorphometry, and detection of OD biomarkers were performed. We found that osteogenesis induced a decrease in miR-487a-3p expression and an increase in FTO expression. miR-487a-3p upregulation inhibited the OD of ADSCs, including decreases in calcium nodule formation, ALP activity, and OD biomarkers. miR-487a-3p downregulation showed the opposite role in OD. miR-487a-3p negatively regulated WNT5A in ADSCs. WNT5A silence reversed the effect of miR-487a-3p downregulation on OD. FTO silence caused the increase in m6A of pri-miR-487a. FTO overexpression inhibited DGCR8 recruitment in pri-miR-487a, and reversed the effect of miR-487a-3p upregulation in OD. ADSCs transplantation improved OP symptom in rats, including improvement of femur tissue, increase in percent bone volume and trabecular number, and upregulation in OD biomarkers. miR-487a-3p downregulation enhanced the therapeutic role of ADSC in rats with OP. Collectively, FTO regulated pri-miR-487a maturation via m6A-dependent pathway, which altered the WNT5A-mediated osteogenesis of ADSCs.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 5","pages":"152"},"PeriodicalIF":3.1,"publicationDate":"2025-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144975912","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Intestinal failure-associated liver disease (IFALD) is a life-threatening complication of short bowel syndrome (SBS), characterized by cholestasis, hepatic steatosis, and hepatic fibrosis. Fish oil-based lipid emulsion (FO) has been demonstrated to ameliorate IFALD compared to soybean oil lipid emulsion (SO). However, the mechanisms underlying the beneficial effects of FO remain elusive. This study investigated the effects of FO on the activation of hepatic stellate cells (HSCs) that are primarily responsible for liver fibrosis by differentiating into fibroblasts in a transforming growth factor-β1 (TGF-β1) dependent manner. The human HSCs line LX-2 cells were stimulated with TGF-β1 in the presence of FO and SO. FO, but not SO, inhibited the degradation of lipid droplets induced by TGF-β1, suggesting that FO maintains HSCs in a quiescent state. Furthermore, FO suppressed LX-2 cell proliferation and partially abolished the autocrine regulation of TGF-β1 and subsequent activation of HSCs, as evidenced by the reduced expression of alpha-1 type I collagen (Col1a1) mRNA. These effects were specific to LX-2 and were not observed in the human hepatocellular carcinoma cell line HuH-7. The unique fatty acid composition of FO, characterized by high levels of long-chain polyunsaturated fatty acids with carbon chains of 20 or more, may contribute to its antifibrotic properties. These findings suggest that FO suppresses excessive HSCs activation while maintaining physiological functions, providing novel insights into the antifibrotic mechanisms of FO.
{"title":"100% Fish oil-based lipid emulsion inhibits hepatic stellate cell activation via suppression of the TGF-β1 autocrine signaling.","authors":"Ryo Watanabe, Mayako Morii, Kenki Yamagata, Takashi Ebihara, Takehiko Sasaki, Yoshihiro Mezaki, Masaru Mizuno","doi":"10.1007/s13577-025-01283-y","DOIUrl":"10.1007/s13577-025-01283-y","url":null,"abstract":"<p><p>Intestinal failure-associated liver disease (IFALD) is a life-threatening complication of short bowel syndrome (SBS), characterized by cholestasis, hepatic steatosis, and hepatic fibrosis. Fish oil-based lipid emulsion (FO) has been demonstrated to ameliorate IFALD compared to soybean oil lipid emulsion (SO). However, the mechanisms underlying the beneficial effects of FO remain elusive. This study investigated the effects of FO on the activation of hepatic stellate cells (HSCs) that are primarily responsible for liver fibrosis by differentiating into fibroblasts in a transforming growth factor-β1 (TGF-β1) dependent manner. The human HSCs line LX-2 cells were stimulated with TGF-β1 in the presence of FO and SO. FO, but not SO, inhibited the degradation of lipid droplets induced by TGF-β1, suggesting that FO maintains HSCs in a quiescent state. Furthermore, FO suppressed LX-2 cell proliferation and partially abolished the autocrine regulation of TGF-β1 and subsequent activation of HSCs, as evidenced by the reduced expression of alpha-1 type I collagen (Col1a1) mRNA. These effects were specific to LX-2 and were not observed in the human hepatocellular carcinoma cell line HuH-7. The unique fatty acid composition of FO, characterized by high levels of long-chain polyunsaturated fatty acids with carbon chains of 20 or more, may contribute to its antifibrotic properties. These findings suggest that FO suppresses excessive HSCs activation while maintaining physiological functions, providing novel insights into the antifibrotic mechanisms of FO.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 5","pages":"153"},"PeriodicalIF":3.1,"publicationDate":"2025-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144975941","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-08-25DOI: 10.1007/s13577-025-01280-1
Keywan Mortezaee
Regulatory T cell (Treg) immunome profile and targets in antiprogrammed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) is a subject of extensive research, but there are still complexities in the area due to the nature of tumor microenvironment (TME). TME of solid tumors contains factors exerting a range of effects on Tregs including development, recruitment, expansion, stability and their immunosuppressive activity. Anti-PD-1 secondarily causes replenishment of intratumoral Tregs, which further intensify tumor immunosuppression. Besides, Treg depletion may also compromise immune checkpoint inhibitor (ICI) efficacy in addition to its other adversarial effects. Thus, a desired ICI booster is to use agents preferentially acting on intratumoral Tregs. Modulation of hypoxia, and regulation of Treg-related cytokines, chemokines, receptors and chromatin modifying factors in tumor TME provide supplementary approaches to anti-PD-(L)1. Factors acting on Tregs have diverse or even dual functions in TME. Treg expansion inhibitory and immunosuppressive tumor-associated macrophage (TAM) recruitment stimulatory effects of stimulator of interferon genes (STING), effector suppressor Treg activating and tumor-specific CD8+ T cell stimulatory effects of interleukin type 2 receptor alpha (IL-2Rα, also called CD25), cell type-dependent dual activities of CXCR3 and inducible T cell costimulatory (ICOS), exposure time-dependent dual effects of glucocorticoid-induced TNFR-related protein (GITR) and CD70 on Tregs and T cells, and exposure level-dependent dual activities of IFN-γ on Tregs are examples require consideration in designing Treg-based strategies. The main direction of this review is to provide updated information about targeting modulators of intratumoral Tregs with agents/compounds aiming to expand anti-PD-(L)1 efficacy and durability in solid tumors.
{"title":"Regulatory T cells in cancer anti-PD-(L)1 therapy.","authors":"Keywan Mortezaee","doi":"10.1007/s13577-025-01280-1","DOIUrl":"10.1007/s13577-025-01280-1","url":null,"abstract":"<p><p>Regulatory T cell (Treg) immunome profile and targets in antiprogrammed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) is a subject of extensive research, but there are still complexities in the area due to the nature of tumor microenvironment (TME). TME of solid tumors contains factors exerting a range of effects on Tregs including development, recruitment, expansion, stability and their immunosuppressive activity. Anti-PD-1 secondarily causes replenishment of intratumoral Tregs, which further intensify tumor immunosuppression. Besides, Treg depletion may also compromise immune checkpoint inhibitor (ICI) efficacy in addition to its other adversarial effects. Thus, a desired ICI booster is to use agents preferentially acting on intratumoral Tregs. Modulation of hypoxia, and regulation of Treg-related cytokines, chemokines, receptors and chromatin modifying factors in tumor TME provide supplementary approaches to anti-PD-(L)1. Factors acting on Tregs have diverse or even dual functions in TME. Treg expansion inhibitory and immunosuppressive tumor-associated macrophage (TAM) recruitment stimulatory effects of stimulator of interferon genes (STING), effector suppressor Treg activating and tumor-specific CD8<sup>+</sup> T cell stimulatory effects of interleukin type 2 receptor alpha (IL-2Rα, also called CD25), cell type-dependent dual activities of CXCR3 and inducible T cell costimulatory (ICOS), exposure time-dependent dual effects of glucocorticoid-induced TNFR-related protein (GITR) and CD70 on Tregs and T cells, and exposure level-dependent dual activities of IFN-γ on Tregs are examples require consideration in designing Treg-based strategies. The main direction of this review is to provide updated information about targeting modulators of intratumoral Tregs with agents/compounds aiming to expand anti-PD-(L)1 efficacy and durability in solid tumors.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 5","pages":"150"},"PeriodicalIF":3.1,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144975860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nicotinamide N-methyltransferase (NNMT) is an S-adenosyl-l-methionine (SAM)-dependent cytosolic enzyme, and a growing body of evidence suggest that it plays an essential role in cancer progression. Recently, NNMT has a role in methylation metabolism and tumorigenesis and was associated with a poor prognosis against numerous cancers. In addition, it has been reported that NNMT has been overexpressed in the stroma of advanced high-grade serous carcinoma and may contribute to decreased survival. This study aimed to identify novel biomarkers to predict resistance and investigate their clinicopathologic significance in paclitaxel-resistant advanced or recurrent ovarian clear cell carcinoma (OCCC). Fluorescence-labeled two-dimensional differential gel electrophoresis (2D-DIGE), immunohistochemical, and MASCOT analyses allowed us to identify the cytoplasmic metabolic enzyme NNMT. In cultured cell studies, NNMT protein expression was higher in paclitaxel-resistant OVMANA and OVTOKO cells than in paclitaxel-sensitive KK and ES-2 cells. Furthermore, although analysis of clinical tissue samples showed no association with poor prognosis in 7 individuals with low NNMT expression in the cytoplasm of OCCC cells, high expression of NNMT in the cytoplasm of OCCC cells may be associated with low sensitivity to paclitaxel in OCCC and may have prognostic implications. Therefore, targeting therapy to reduce cytoplasmic NNMT expression levels may increase the sensitivity of OCCC to paclitaxel.
{"title":"Nicotinamide N-methyltransferase enhances paclitaxel resistance in ovarian clear cell carcinoma.","authors":"Ryoko Kikuchi-Koike, Masaru Sakamoto, Yuko Sasajima, Yuko Miyagawa, Hiroshi Uozaki, Kenji Umayahara, Kei Hashimoto, Yuko Takahashi, Kazuki Takasaki, Chikara Kihira, Haruka Nishida, Takayuki Ichinose, Mana Hirano, Haruko Hiraike, Kazunori Nagasaka","doi":"10.1007/s13577-025-01282-z","DOIUrl":"10.1007/s13577-025-01282-z","url":null,"abstract":"<p><p>Nicotinamide N-methyltransferase (NNMT) is an S-adenosyl-l-methionine (SAM)-dependent cytosolic enzyme, and a growing body of evidence suggest that it plays an essential role in cancer progression. Recently, NNMT has a role in methylation metabolism and tumorigenesis and was associated with a poor prognosis against numerous cancers. In addition, it has been reported that NNMT has been overexpressed in the stroma of advanced high-grade serous carcinoma and may contribute to decreased survival. This study aimed to identify novel biomarkers to predict resistance and investigate their clinicopathologic significance in paclitaxel-resistant advanced or recurrent ovarian clear cell carcinoma (OCCC). Fluorescence-labeled two-dimensional differential gel electrophoresis (2D-DIGE), immunohistochemical, and MASCOT analyses allowed us to identify the cytoplasmic metabolic enzyme NNMT. In cultured cell studies, NNMT protein expression was higher in paclitaxel-resistant OVMANA and OVTOKO cells than in paclitaxel-sensitive KK and ES-2 cells. Furthermore, although analysis of clinical tissue samples showed no association with poor prognosis in 7 individuals with low NNMT expression in the cytoplasm of OCCC cells, high expression of NNMT in the cytoplasm of OCCC cells may be associated with low sensitivity to paclitaxel in OCCC and may have prognostic implications. Therefore, targeting therapy to reduce cytoplasmic NNMT expression levels may increase the sensitivity of OCCC to paclitaxel.</p>","PeriodicalId":49194,"journal":{"name":"Human Cell","volume":"38 5","pages":"151"},"PeriodicalIF":3.1,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12378295/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144975878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}