{"title":"Inflammatory suppression and immunity regulation benefits of honokiol in a rat model of acute peritonitis via the regulation of NLRP3 inflammasome and Sirt1/autophagy axis.","authors":"Ximing Pan, Zhou Hua, Guocai Fan, Qinglong Feng","doi":"10.14670/HH-18-688","DOIUrl":"10.14670/HH-18-688","url":null,"abstract":"<p><strong>Background: </strong>NLRP3 inflammasome and Sirt1/autophagy axis are potential targets for advancing acute peritonitis (AP). Honokiol (HNK), a bioactive substance, has the potential to improve AP.</p><p><strong>Materials and methods: </strong>The AP model rats were established by cecal ligation and puncture (CLP). Rats were randomized into the Sham, Sham+HNK, CLP, and CLP+HNK groups. The therapeutic effects of HNK on organ infection, inflammation and immunity were observed in AP rats. The inflammation of RAW 264.7 cells was induced by lipopolysaccharide (LPS) and divided into the Control, HNK, LPS, and LPS+HNK groups. The effects of HNK on immunity and inflammation were observed. Moreover, the inflammatory cell model was further transfected with NLRP3 overexpressing plasmid, and the regulatory effect of HNK on NLRP3 in AP cells was detected.</p><p><strong>Results: </strong>HNK treatment improved survival, biochemical indexes, and lung and kidney injury and inhibited inflammatory cytokine release and bacterial infection in CLP rats. In CLP rats and RAW 264.7 cells, HNK treatment improved the release of the CD4+ and CD8+ T cells, decreased the associated proteins' levels of the NLRP3 inflammasome, and activated the expression of proteins in the Sirt1/autophagy axis. It improved viability and reduced apoptosis and the degrees of TNF-α, IL-1β, and IL-6 mRNA in RAW 264.7 cells. In addition, HNK treatment antagonized the effect of NLRP3-overexpressed on inflammation and immunity.</p><p><strong>Conclusions: </strong>HNK improved AP by inhibiting NLRP3 inflammasome and activating the Sirt1 autophagy axis <i>in vivo</i> and <i>in vitro</i>.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138800883","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Selenomethionine suppresses head and neck squamous cell carcinoma progression through TopBP1/ATR and TCAB1 signaling.","authors":"Bo Zhang, Xiaodong Wei, Jiwu Li","doi":"10.14670/HH-18-665","DOIUrl":"10.14670/HH-18-665","url":null,"abstract":"<p><strong>Objective: </strong>Head and neck squamous cell carcinoma (HNSCC) is a histological type of cancer originating from the head and neck. Selenium complexes have been considered as a potential treatment for HNSCC. Therefore, the present work focused on probing the mechanism of L-selenomethionine (SeMet) in HNSCC treatment.</p><p><strong>Methods: </strong>MTT and colony formation assays were carried out to analyze the survival rate and proliferation of HNSCC cells, respectively. TUNEL staining was performed to examine apoptosis of HNSCC cells. Additionally, qRT-PCR and Western blotting assays were performed to measure mRNA and protein levels, separately.</p><p><strong>Results: </strong>SeMet treatment significantly hindered the survival and promoted the apoptosis of HNSCC cells in a dose- and time-dependently. SeMet administration promoted expression of TopBP1, ATR, H2AX, p-ATR and γ-H2AX, and suppressed that of TCAB1. Importantly, SeMet treatment suppressed the proliferation and facilitated the apoptosis of HNSCC cells, which were partly reversed by down-regulation of TopBP1 or up-regulation of TCAB1. The activation of SeMet to TopBP1/ATR signaling was rescued by TCAB1 up-regulating, and the inhibition of SeMet to TCAB1 expression was rescued by TopBP1 silencing.</p><p><strong>Conclusion: </strong>Our findings show that SeMet inhibits the proliferation of HNSCC cells and promotes their apoptosis by targeting TopBP1/ATR and TCAB1 signaling. SeMet is a potential method for HNSCC treatment.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41110780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Christiane Kuempers, Karoline Schnepf, Sebastian Marwitz, Christian Watermann, Andreas H Scheel, Rieke N Fischer, Ole Ammerpohl, Sven Perner, Daniel Drömann, Torsten Goldmann
{"title":"Upregulation and epigenetic modification of the creatine transporter SLC6A8 in non-small cell lung cancer.","authors":"Christiane Kuempers, Karoline Schnepf, Sebastian Marwitz, Christian Watermann, Andreas H Scheel, Rieke N Fischer, Ole Ammerpohl, Sven Perner, Daniel Drömann, Torsten Goldmann","doi":"10.14670/HH-18-731","DOIUrl":"10.14670/HH-18-731","url":null,"abstract":"<p><strong>Introduction: </strong>Lung cancer is a major cause of cancer-related death worldwide and effective therapies, besides surgery, are available only for a small proportion of patients. Since cellular respiration is known to be broadly altered in malignant tumors, the cellular processes of respiration can be a potential therapeutic target. One important element of cellular respiration is creatine and its transport by the creatine transporter SLC6A8. Here we describe the expression of SLC6A8 at the RNA and protein level, epigenetic modifications as well as survival analysis in NSCLC tissues and matched controls.</p><p><strong>Materials and methods: </strong>We analyzed epigenetic modifications of the <i>SLC68A</i> gene in 32 patients, of which 18 were additionally analyzed by transcriptome analysis. The expression of SLC6A8 at the protein level was assessed by immunohistochemistry using an independent cohort and correlated with clinicopathological data including survival. Kaplan-Meier analysis was performed to analyze the possible effects of the transcriptional levels of <i>SLC6A8</i> in another separate cohort (n=1925).</p><p><strong>Results: </strong><i>SLC6A8</i> loci are epigenetically modified in NSCLC compared with tumor-free controls. SLC6A8 is upregulated in NSCLC at the RNA and protein level. High mRNA expression of <i>SLC6A8</i> was associated with an overall poor prognosis in lung adenocarcinoma patients and displayed the strongest adverse prognostic effect in male smokers with adenocarcinomas. Results of transcriptome analysis were partially confirmed at the protein level.</p><p><strong>Conclusions: </strong>Our results suggest an important role of creatine and its transport via SLC6A8 in NSCLC.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Elia A Baltazar-García, Belinda Vargas-Guerrero, Luz E Gasca-Lozano, Carmen M Gurrola-Díaz
{"title":"Molecular changes underlying pulmonary emphysema and chronic bronchitis in Chronic Obstructive Pulmonary Disease: An updated review.","authors":"Elia A Baltazar-García, Belinda Vargas-Guerrero, Luz E Gasca-Lozano, Carmen M Gurrola-Díaz","doi":"10.14670/HH-18-699","DOIUrl":"10.14670/HH-18-699","url":null,"abstract":"<p><p>The aim of this review is to update and synthesize the molecular mechanisms that lead to the heterogeneous effect on tissue remodeling observed in the two most important clinical phenotypes of chronic obstructive pulmonary disease (COPD), pulmonary emphysema (PE) and chronic bronchitis (CB). Clinical and experimental evidence suggests that this heterogeneous response to promote PE, CB, or both, is related to differentiated genetic, epigenetic, and molecular conditions. Specifically, a tendency toward PE could be related to a variant in the <i>DSP</i> gene, SIRT1 downregulation, macrophage polarization to M1, as well as the involvement of the noncanonical Wnt5A signaling pathway, among other alterations. Additionally, in advanced stages of COPD, PE development is potentiated by dysregulations in autophagy, which promotes senescence and subsequently cell apoptosis, through exacerbated inflammasome activation and release of caspases. On the other hand, CB or the pro-fibrotic phenotype could be potentiated by the downregulated activity of HDAC2, the activation of the TGF-β/Smad or Wnt/β-catenin signaling pathways, macrophage polarization to M2, upregulation of TIMP-1, and/or the presence of the epithelial-mesenchymal transition (EMT) mechanism. Interestingly, the upregulated activity of MMPs, especially MMP-9, is widely involved in the development of both phenotypes. Furthermore, MMP-9 and MMP-12 enhance the severity, perpetuation, and exacerbation of COPD, as well as the development of autoimmunity in this disease.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139472292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Tsubasa Betsuyaku, Yuko Ito, Nicholas Peake, Abdul Alim Al-Bari, Gehan El-Akabawy, Nabil Eid
{"title":"Enhanced autophagy and phagocytosis of apoptotic lymphocytes in splenic macrophages of acute ethanol-treated rats: Light and electron microscopic studies.","authors":"Tsubasa Betsuyaku, Yuko Ito, Nicholas Peake, Abdul Alim Al-Bari, Gehan El-Akabawy, Nabil Eid","doi":"10.14670/HH-18-729","DOIUrl":"10.14670/HH-18-729","url":null,"abstract":"<p><p>Autophagy is a prosurvival mechanism for the clearance of damaged cellular components, specifically upon exposure to various stressors. In lymphoid organs, excessive ethanol consumption increases lymphocyte apoptosis, resulting in immunosuppression. However, ethanol-induced autophagy and related phagocytosis of apoptotic lymphocytes in the spleen have not been studied yet. Adult male Wistar rats were injected intraperitoneally either with 5 g/kg ethanol or phosphate-buffered saline (as a control group) and then sacrificed 0, 3, 6, and 24 hours after injection. Light and transmission electron microscopy (TEM) findings indicated enhanced T cell apoptosis in the white pulps of ethanol-treated rats (ETRs) compared with the control group, which peaked at 6 h and was associated with the accumulation of tingible body macrophages (TBMs). These macrophages exhibited an upregulated autophagic response, as evidenced by enhanced LC3-II (a specific marker of autophagosomes) expression, which peaked at 24h. In addition, double labeling immunofluorescence of LC3-II with lysosomal markers revealed the enhanced formation of autolysosomes in TBMs of ETRs, which was associated with suppression of p62 immunostaining, indicating the enhanced autophagic flux. Interestingly, this elevated autophagic response in ETR TBMs was accompanied by evidence of LC3-associated phagocytosis (LAP) of apoptotic splenocytes. This is based on TUNEL/LC3-II double labeling and TEM observations of phagosomes containing apoptotic bodies, enclosed within phagosomal membranes adjacent to the autophagic vacuoles. It can be concluded that enhanced prosurvival autophagy in splenic TBMs of ETRs and clearing of apoptotic lymphocytes via LAP may contribute to preventing secondary necrosis and autoimmune diseases.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140093812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Notoginsenoside Fc alleviates oxidized low-density lipoprotein-induced endothelial cell dysfunction and upregulates PPAR-γ <i>in vitro</i>.","authors":"Yuanhao Zhang, Kui Liu, Dile Wang","doi":"10.14670/HH-18-694","DOIUrl":"10.14670/HH-18-694","url":null,"abstract":"<p><strong>Background: </strong>Deep venous thrombosis (DVT) is a prevalent vascular disease and a major cause of morbidity and mortality worldwide. Notoginsenoside Fc (NFc) is a protopanaxadiol-type saponin that has been shown to have beneficial effects on several disorders. However, its function in DVT is unclear.</p><p><strong>Methods: </strong>Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to mimic DVT <i>in vitro</i> and treated with NFc to investigate its functions. CCK-8 assay was utilized for measuring cell viability. Western blotting was used for detecting protein levels of proinflammatory cytokines, apoptosis-related markers, and peroxisome proliferator-activated receptor-γ (PPAR-γ). Flow cytometry was performed for cell apoptosis detection. Levels of oxidative stress-related markers were examined by the DCFH-DA method and ELISA. RT-qPCR was utilized for the measurement of PPAR-γ mRNA level.</p><p><strong>Results: </strong>NFc increased the viability and suppressed inflammation, apoptosis, and oxidative stress in ox-LDL-treated HUVECs. NFc treatment induced upregulation of PPAR-γ in HUVECs.</p><p><strong>Conclusion: </strong>NFc mitigates ox-LDL-induced dysfunction of HUVECs.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139402590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"NRF3 suppresses the metastasis of triple-negative breast cancer cells by inhibiting ERK activation in a ROS-dependent manner.","authors":"Chenhui Zheng, Yue Pan, Bangyi Lin, Jin Li, Qi Chen, Zhibao Zheng","doi":"10.14670/HH-18-786","DOIUrl":"https://doi.org/10.14670/HH-18-786","url":null,"abstract":"<p><strong>Purpose: </strong>Our previous study demonstrated that NRF3 (NFE2L3, Nuclear Factor-erythroid 2-related factor 3) could suppress cell metastasis and proliferation in breast cancer. In this study, we investigated the mechanisms underlying its function in breast cancer.</p><p><strong>Methods: </strong>In the present study, NRF3 expression and its clinical characteristics in breast cancer were analyzed using public datasets and clinical specimens. After breast cancer cells were overexpressed NRF3, FACS was used to detect the intracellular ROS levels. The migration and invasion activities of NRF3-ectopic expressed breast cancer cells were determined by transwell assay. To validate the role of ROS/ERK axis in the inhibitory effect of NRF3 in cell metastasis, ROS scavenger NAC was also included.</p><p><strong>Results: </strong>We found that NRF3 mRNA was highly expressed, while NRF3 protein was extremely lowly expressed in breast cancer tissues compared with their normal counterparts, and low level NRF3 was associated with poorer prognosis in patients with triple negative breast cancer (TNBC). More interestingly, overexpression of NRF3 protein significantly increased cellular ROS production and dramatically decreased p-ERK level and cell migration in TNBC cells. Mechanistically, NRF3 protein was found to be mutually regulated by valosin-containing protein (VCP). Strikingly, VCP-knockdown dramatically increased NRF3 protein expression, but NRF3-knockin also decreased VCP expression in return. Moreover, antioxidant NAC treatment effectively increased the level of p-ERK and VCP expression, as well as cell migration and invasion abilities of TNBC cells.</p><p><strong>Conclusion: </strong>NRF3, a tumor suppressor downregulated by VCP, could attenuate cell metastasis in TNBC cells by increasing cellular ROS accumulation and subsequently inhibiting the ERK phosphorylation.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lingyan Jin, Hye-Yeong Jin, Younghoon Kim, Nam-Yun Cho, Jeong Mo Bae, Jung Ho Kim, Sae-Won Han, Tae-You Kim, Gyeong Hoon Kang
{"title":"Clinicopathological and molecular features of genome-stable colorectal cancers.","authors":"Lingyan Jin, Hye-Yeong Jin, Younghoon Kim, Nam-Yun Cho, Jeong Mo Bae, Jung Ho Kim, Sae-Won Han, Tae-You Kim, Gyeong Hoon Kang","doi":"10.14670/HH-18-785","DOIUrl":"https://doi.org/10.14670/HH-18-785","url":null,"abstract":"<p><p>Colorectal cancers (CRCs) are traditionally divided into those with either chromosomal instability (CIN) or microsatellite instability (MSI). By utilizing TCGA data, the Laird team found a subset of CRCs, namely, genome-stable CRCs (GS CRCs), which lack both CIN and MSI. Although the molecular features of GS CRCs have been described in detail, the clinicopathological features are not well defined. A total of 437 CRCs were analyzed for copy number variation (CNV) statuses in eight genes (<i>ARID1A, EGFR, FGFR1, KDM5B, MYBL2, MYC, SALL4,</i> and <i>SETDB1</i>) using droplet-digital PCR. CRCs that showed CNV in ≤ one gene and no MSI were defined as GS-like CRCs. Clinicopathological and molecular features of GS-like CRCs were compared with those of CIN-like CRCs. GS-like CRCs comprised 4.6% of CRCs and showed a predilection toward the proximal colon, lower nuclear optical density, <i>KRAS</i> mutation, <i>PIK3CA</i> mutation, and aberrant expression of KRT7. Survival analysis showed no significant difference between the three subgroups. Through our study, the GS-like subtype was found to comprise a minor proportion of CRCs and have proclivity toward a proximal bowel location, hypochromatic tumor nuclei, aberrant KRT7 expression, and a high frequency of <i>KRAS</i> and <i>PIK3CA</i> mutations.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141590198","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Glucan-rich polysaccharides obtained from split gill mushroom [<i>Schizophyllum commune (Fr.)</i>] ameliorate hyperglycemia by enhancing insulin and GLUT2 pancreas in type 2 diabetic rats.","authors":"Udomlak Matsathit, Decha Sermwittayawong, Manaras Komolgriengkrai, Wipapan Khimmaktong","doi":"10.14670/HH-18-784","DOIUrl":"https://doi.org/10.14670/HH-18-784","url":null,"abstract":"<p><strong>Aims: </strong>Abnormalities in the secretion of insulin are the cause of pathology and complications in diabetic patients. The aim of this study was to investigate the anti-diabetic effect of polysaccharide extracts from the split gill mushroom in type 2 diabetes rats administered a low dose of streptozotocin (STZ) in combination with a high-fat diet.</p><p><strong>Methods: </strong>The rats were divided into 6 groups: the control group (ND), the control group fed with polysaccharide extract from split gill (ND240), the diabetes group (HFD+DM), the diabetic group fed 120 (HFD+S120) and 240 mg/kg BW polysaccharide extract (HFD+S240), and the diabetic group receiving metformin (HFD+Met). Subsequently, the Islets of Langerhans of pancreatic tissue were studied using a light microscope and transmission electron microscopy (TEM). Immunofluorescence for the detection of insulin and glucose transporter 2 (GLUT2) proteins, and malondialdehyde (MDA) were also detected in pancreatic tissue.</p><p><strong>Results: </strong>In the diabetic and HFD+120 groups, the tissues harbored various pathologies. The HFD+S240 and HFD+Met groups were found to have lower blood sugar levels. The levels of insulin and GLUT2 increased compared with the diabetic group. Additionally, the levels of MDA were reduced.</p><p><strong>Conclusions: </strong>The use of polysaccharide extract from split gill mushrooms (240 mg/kg BW) is an alternative to treating various pathologies in the relief or treatment of diabetes mellitus.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141579483","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"GATA4 is diagnostically useful for distinguishing primary ovarian mucinous carcinomas from metastatic colorectal adenocarcinomas to the ovary.","authors":"Kunio Mochizuki, Ippei Tahara, Tomohiro Inoue, Kazunari Kasai, Tetsuo Kondo","doi":"10.14670/HH-18-783","DOIUrl":"https://doi.org/10.14670/HH-18-783","url":null,"abstract":"<p><strong>Aim: </strong>Determining the primary origin of an ovarian mucin-producing carcinoma can be challenging at times because some metastases of primary colorectal origin may exhibit gross, microscopic, and/or immunohistochemical features that overlap with those of primary ovarian mucinous carcinomas (OMCs). We hypothesized that GATA binding protein 4 (GATA4) might be a novel, useful marker for differentiating primary OMCs from metastatic colorectal adenocarcinomas to the ovary.</p><p><strong>Methodology: </strong>For comparison with the usefulness of other markers (special AT-rich sequence-binding protein 2 (SATB2) and caudal type homeobox 2 (CDX2)), we elucidated the expression profiles of GATA4 in OMCs, colorectal non-mucinous adenocarcinomas (CNMACs), and colorectal mucinous adenocarcinomas (CMACs) using immunohistochemistry.</p><p><strong>Results: </strong>We confirmed GATA4 expression (H-score ≥50 points) in 93%, SATB2 in 0%, and CDX2 in 64% of 14 OMCs. GATA4 was expressed in 13%, SATB2 in 90%, and CDX2 in 93% of 30 CNMACs. GATA4 was expressed in 20%, SATB2 in 73%, and CDX2 in 100% of 30 CMACs.</p><p><strong>Conclusion: </strong>The expression of GATA4 in a mucus-producing ovarian tumor strongly supports it being a primary OMC rather than a metastatic colorectal carcinoma: GATA4 expression indicates OMC and SATB2 expression indicates colorectal adenocarcinoma. However, three cases of colorectal adenocarcinoma were GATA4-positive and SATB2-negative, so the GATA4/SATB2 marker combination is not absolute for determining the primary site. Further research for more markers is necessary to find the ideal combination.</p>","PeriodicalId":13164,"journal":{"name":"Histology and histopathology","volume":null,"pages":null},"PeriodicalIF":2.5,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141554645","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}