Pub Date : 2025-10-14DOI: 10.1007/s10735-025-10634-w
Khaled Abdul-Aziz Ahmed, Khalid M. Alqaisi, Noralhuda Ayad Ibrahim, Najat Jabbar Ahmed, Qosay A. Al-Balas, Ahmed Aj. Jabbar, Muna Horabi, Hanan Ibrahim Althagbi, Goran Noori Saleh, Ahmed Hameed Al-Dabhawi, Rawaz Rizgar Hassan, Talal Salem Al-Qaisi
Gastric ulcers are one of the major public health burdens in the modern era, with increased complications that could be a result of alcohol abuse or H. pylori infection. In this study, we investigated the therapeutic potential and acute toxicity effects of Pterostilbene (PSB) in ethanol-mediated gastropathy, as well as its underlying molecular mechanism, in rats. Gastric ulcers are provoked by absolute ethanol (5 mL/kg, i.g.) in male Sprague–Dawley rats after receiving oral treatments: physiological saline (negative, 5 mL/kg), omeprazole (positive control, 20 mg/kg), and PSB (30 and 60 mg/kg). PSB pretreatment significantly alleviated clinical signs, reduced the macroscopic ulcer index, and improved gastric mucosal morphology, including gastric defense barriers (mucus and glycoprotein production). PSB Pretreatment improved ethanol-mediated microscopical alterations, as indicated by reduced submucosal oedema, decreased hemorrhagic/lesion areas, and restoration of mucosal integrity. PSB down-regulated apoptotic actions (reduced P53 and increased Bcl-2 protein expression), lowered inflammatory conditions (decreased TNF-α, IL-6, and increased IL-10), and limited oxidative stress tissue injuries (up-regulated SOD, CAT, and PGE2 while lowering MDA). The PSB gastroprotection may be linked to a strengthened gastric defense and anti-oxidative/anti-inflammatory pathways, ultimately curbing apoptotic actions by modulating death signals, P53, and Bcl-2 proteins. The outcomes present PSB as a viable nutraceutical and biopharmaceutical product for managing stomach disorders, including gastric ulcers.
{"title":"Pterostilbene (grape flavonoid) mitigates gastric ulcer events in vivo: biochemical and histopathological approaches","authors":"Khaled Abdul-Aziz Ahmed, Khalid M. Alqaisi, Noralhuda Ayad Ibrahim, Najat Jabbar Ahmed, Qosay A. Al-Balas, Ahmed Aj. Jabbar, Muna Horabi, Hanan Ibrahim Althagbi, Goran Noori Saleh, Ahmed Hameed Al-Dabhawi, Rawaz Rizgar Hassan, Talal Salem Al-Qaisi","doi":"10.1007/s10735-025-10634-w","DOIUrl":"10.1007/s10735-025-10634-w","url":null,"abstract":"<div><p>Gastric ulcers are one of the major public health burdens in the modern era, with increased complications that could be a result of alcohol abuse or <i>H. pylori</i> infection. In this study, we investigated the therapeutic potential and acute toxicity effects of Pterostilbene (PSB) in ethanol-mediated gastropathy, as well as its underlying molecular mechanism, in rats. Gastric ulcers are provoked by absolute ethanol (5 mL/kg, i.g.) in male Sprague–Dawley rats after receiving oral treatments: physiological saline (negative, 5 mL/kg), omeprazole (positive control, 20 mg/kg), and PSB (30 and 60 mg/kg). PSB pretreatment significantly alleviated clinical signs, reduced the macroscopic ulcer index, and improved gastric mucosal morphology, including gastric defense barriers (mucus and glycoprotein production). PSB Pretreatment improved ethanol-mediated microscopical alterations, as indicated by reduced submucosal oedema, decreased hemorrhagic/lesion areas, and restoration of mucosal integrity. PSB down-regulated apoptotic actions (reduced P53 and increased Bcl-2 protein expression), lowered inflammatory conditions (decreased TNF-α, IL-6, and increased IL-10), and limited oxidative stress tissue injuries (up-regulated SOD, CAT, and PGE2 while lowering MDA). The PSB gastroprotection may be linked to a strengthened gastric defense and anti-oxidative/anti-inflammatory pathways, ultimately curbing apoptotic actions by modulating death signals, P53, and Bcl-2 proteins. The outcomes present PSB as a viable nutraceutical and biopharmaceutical product for managing stomach disorders, including gastric ulcers.</p></div>","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 6","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145284465","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}
Pub Date : 2025-10-14DOI: 10.1007/s10735-025-10623-z
Xudong Yao, Tao Peng
The impaired osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) contributes significantly to osteoporosis (OP) pathogenesis. While Forkhead box p1 (FOXP1) is known to regulate stem cell differentiation, its specific role in BMSCs osteogenic differentiation during OP remains unclear. Here, BMSCs were cultured in osteogenic medium for 14 d to induce osteogenic differentiation. We found that FOXP1 was upregulated in BMSCs treated with osteogenic medium, and FOXP1 overexpression promoted BMSC osteogenic differentiation, whereas FOXP1 knockdown inhibited BMSCs osteogenic differentiation. Mechanistically, METTL14 mediated m6A methylation of FOXP1 mRNA, which was recognized by YTHDF1/YTHDF3 to enhance its mRNA stability. Notably, METTL14 overexpression promoted osteogenic differentiation of BMSCs, this effect was abolished by FOXP1 knockdown. The stabilized FOXP1 protein activated the Wnt/β-catenin signaling pathway to drive BMSC osteogenesis. In vivo, administration of FOXP1-overexpressing lentivirus in ovariectomized (OVX) mice significantly attenuated osteoporosis progression. Collectively, our findings reveal that METTL14-dependent m6A modification and YTHDF1/YTHDF3-mediated stabilization of FOXP1 alleviate osteoporosis in OVX mice through Wnt/β-catenin activation, positioning FOXP1 as a promising therapeutic target for postmenopausal osteoporosis.
{"title":"Methyltransferase-like 14 mediated FOXP1 m6A modification alleviates osteoporosis by regulating the Wnt/β-catenin pathway","authors":"Xudong Yao, Tao Peng","doi":"10.1007/s10735-025-10623-z","DOIUrl":"10.1007/s10735-025-10623-z","url":null,"abstract":"<div><p>The impaired osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) contributes significantly to osteoporosis (OP) pathogenesis. While Forkhead box p1 (FOXP1) is known to regulate stem cell differentiation, its specific role in BMSCs osteogenic differentiation during OP remains unclear. Here, BMSCs were cultured in osteogenic medium for 14 d to induce osteogenic differentiation. We found that FOXP1 was upregulated in BMSCs treated with osteogenic medium, and FOXP1 overexpression promoted BMSC osteogenic differentiation, whereas FOXP1 knockdown inhibited BMSCs osteogenic differentiation. Mechanistically, METTL14 mediated m6A methylation of FOXP1 mRNA, which was recognized by YTHDF1/YTHDF3 to enhance its mRNA stability. Notably, METTL14 overexpression promoted osteogenic differentiation of BMSCs, this effect was abolished by FOXP1 knockdown. The stabilized FOXP1 protein activated the Wnt/β-catenin signaling pathway to drive BMSC osteogenesis. In vivo, administration of FOXP1-overexpressing lentivirus in ovariectomized (OVX) mice significantly attenuated osteoporosis progression. Collectively, our findings reveal that METTL14-dependent m6A modification and YTHDF1/YTHDF3-mediated stabilization of FOXP1 alleviate osteoporosis in OVX mice through Wnt/β-catenin activation, positioning FOXP1 as a promising therapeutic target for postmenopausal osteoporosis.</p></div>","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 6","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145284478","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}
Pub Date : 2025-10-14DOI: 10.1007/s10735-025-10622-0
Tong Wang, Xiaobo Zhang, Tao Shen, Zubing Zhou
<div><p>This study aims to investigate the lipid-lowering, anti-inflammatory, and antioxidant effects of Zhuyu Pill in non-alcoholic fatty liver disease (NAFLD), and to determine whether its therapeutic action is mediated through modulation of the IL-22/JAK1/STAT3 signaling pathway. In animal experiments, C57BL/6 mice were fed a high-fat diet for 17 weeks to induce a NAFLD model. They were randomly divided into a control group, a Zhuyu Pill treatment group, and a positive control group, and the intervention lasted for 6 weeks. Mouse liver tissue and serum were collected for biochemical analysis of blood lipids and liver enzyme activities. Liver sections were processed for HE staining and Oil Red O staining to evaluate histopathological changes and lipid accumulation. ELISA was employed to quantify inflammatory factor levels, while DCFH-DA and MitoSOX staining used to detect reactive oxygen species (ROS) in the liver. In cell experiments, mouse liver cells AML-12 were used as a model, and cells were treated with free fatty acids to simulate the NAFLD microenvironment. ELISA was used to detect changes in the expression of inflammatory factors and detecting the levels of cellular ROS and mitochondrial ROS. In addition, siRNA technology was used to silence IL-22 and activate the JAK/STAT signaling pathway with Colivelin. The changes in lipid deposition and ROS signaling in cells were observed, and the expression changes of IL-22/JAK1/STAT3 signaling pathway related proteins were detected by WB method. In animal experiments, the detection results of serum biochemical indicators in mice showed that Zhuyu Pill can effectively reduce the levels of ALT, AST, TC, TG, LDL-C, and HDL-C. HE and Oil Red O staining revealed that Zhuyu Pill markedly alleviated hepatic lipid accumulation and inflammatory infiltration. The results of inflammatory factors showed that treatment with Zhuyu Pill significantly reduced the levels of pro-inflammatory factors IL-6, TNF-α, IL-1 β, and MCP-1 in mouse serum, while increasing the expression of IL-22. The results of DCFH-DA and MitoSOX staining methods showed that Zhuyu Pill could significantly reduce the level of ROS in the liver tissue of NAFLD mice. In vitro experiments showed that treatment with Zhuyu Pill significantly reduced the levels of pro-inflammatory cytokines IL-6, TNF-α, IL-1 β, and MCP-1 in NAFLD cell models, increased the expression of IL-22, significantly reduced ROS levels, increased the expression of p-JAK1 protein, p-STAT3 protein, BCL-2 protein, and reduced the expression of BAX protein. SiRNA-mediated IL-22 silencing markedly attenuated the therapeutic effects of Zhuyu Pill on reducing lipid deposition and enhancing antioxidant defense, and also diminished its ability to activate the IL-22/JAK1/STAT3 signaling pathway. Meanwhile, STAT3 activator Colivelin was able to partially reverse the effects of IL-22 silencing. Zhuyu Pill can regulate lipid metabolism, inhibit inflammatory response, alleviate oxidati
{"title":"Zhuyu pill mitigates NAFLD through activation of IL22/JAK1/STAT3 signaling pathway","authors":"Tong Wang, Xiaobo Zhang, Tao Shen, Zubing Zhou","doi":"10.1007/s10735-025-10622-0","DOIUrl":"10.1007/s10735-025-10622-0","url":null,"abstract":"<div><p>This study aims to investigate the lipid-lowering, anti-inflammatory, and antioxidant effects of Zhuyu Pill in non-alcoholic fatty liver disease (NAFLD), and to determine whether its therapeutic action is mediated through modulation of the IL-22/JAK1/STAT3 signaling pathway. In animal experiments, C57BL/6 mice were fed a high-fat diet for 17 weeks to induce a NAFLD model. They were randomly divided into a control group, a Zhuyu Pill treatment group, and a positive control group, and the intervention lasted for 6 weeks. Mouse liver tissue and serum were collected for biochemical analysis of blood lipids and liver enzyme activities. Liver sections were processed for HE staining and Oil Red O staining to evaluate histopathological changes and lipid accumulation. ELISA was employed to quantify inflammatory factor levels, while DCFH-DA and MitoSOX staining used to detect reactive oxygen species (ROS) in the liver. In cell experiments, mouse liver cells AML-12 were used as a model, and cells were treated with free fatty acids to simulate the NAFLD microenvironment. ELISA was used to detect changes in the expression of inflammatory factors and detecting the levels of cellular ROS and mitochondrial ROS. In addition, siRNA technology was used to silence IL-22 and activate the JAK/STAT signaling pathway with Colivelin. The changes in lipid deposition and ROS signaling in cells were observed, and the expression changes of IL-22/JAK1/STAT3 signaling pathway related proteins were detected by WB method. In animal experiments, the detection results of serum biochemical indicators in mice showed that Zhuyu Pill can effectively reduce the levels of ALT, AST, TC, TG, LDL-C, and HDL-C. HE and Oil Red O staining revealed that Zhuyu Pill markedly alleviated hepatic lipid accumulation and inflammatory infiltration. The results of inflammatory factors showed that treatment with Zhuyu Pill significantly reduced the levels of pro-inflammatory factors IL-6, TNF-α, IL-1 β, and MCP-1 in mouse serum, while increasing the expression of IL-22. The results of DCFH-DA and MitoSOX staining methods showed that Zhuyu Pill could significantly reduce the level of ROS in the liver tissue of NAFLD mice. In vitro experiments showed that treatment with Zhuyu Pill significantly reduced the levels of pro-inflammatory cytokines IL-6, TNF-α, IL-1 β, and MCP-1 in NAFLD cell models, increased the expression of IL-22, significantly reduced ROS levels, increased the expression of p-JAK1 protein, p-STAT3 protein, BCL-2 protein, and reduced the expression of BAX protein. SiRNA-mediated IL-22 silencing markedly attenuated the therapeutic effects of Zhuyu Pill on reducing lipid deposition and enhancing antioxidant defense, and also diminished its ability to activate the IL-22/JAK1/STAT3 signaling pathway. Meanwhile, STAT3 activator Colivelin was able to partially reverse the effects of IL-22 silencing. Zhuyu Pill can regulate lipid metabolism, inhibit inflammatory response, alleviate oxidati","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 6","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145284490","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}
Pub Date : 2025-10-11DOI: 10.1007/s10735-025-10627-9
Jun-qi Li, Rui-ze Sun
Corneal alkali burns trigger the infiltration of inflammation-associated macrophages, which can potentially lead to vision impairment. Although Sinomenine (SIN) is recognized for its anti-inflammatory properties, its role on the polarization of macrophages and its effects on corneal alkali injury remain inadequately understood. In this study, corneal alkali damage was induced using NaOH. HE staining and immunohistochemistry were conducted to assess macrophage infiltration in corneal tissues. Macrophages were isolated via flow cytometry assay. Quantitative real-time PCR (qRT-PCR) was employed to examine the expression of genes associated with macrophage polarization. The results indicated an increase in macrophage infiltration and M1 polarization in mice subjected to corneal alkali injury. Sinomenine treatment effectively inhibited the polarization of M1 macrophages in the cornea. In mice treated with Sinomenine post-injury, there was a reduction in the expression of the m6A methylation writer protein METTL3 in macrophages, which led to an upregulation of IRAKM expression. The elevated IRAKM expression subsequently inhibited the TLR4 inflammatory pathway and reduced corneal inflammatory cell infiltration, ultimately ameliorating corneal alkali burns. Sinomenine attenuates M1 macrophage polarization through upregulating IRAKM by inhibiting the expression of the m6A methylation writer protein METTL3 in macrophages, leading to enhanced outcomes in corneal alkali injuries.
{"title":"Sinomenine enhances recovery from corneal alkali burns by attenuating macrophage M1 polarization through modulating the METTL3/IRAKM axis","authors":"Jun-qi Li, Rui-ze Sun","doi":"10.1007/s10735-025-10627-9","DOIUrl":"10.1007/s10735-025-10627-9","url":null,"abstract":"<div><p>Corneal alkali burns trigger the infiltration of inflammation-associated macrophages, which can potentially lead to vision impairment. Although Sinomenine (SIN) is recognized for its anti-inflammatory properties, its role on the polarization of macrophages and its effects on corneal alkali injury remain inadequately understood. In this study, corneal alkali damage was induced using NaOH. HE staining and immunohistochemistry were conducted to assess macrophage infiltration in corneal tissues. Macrophages were isolated via flow cytometry assay. Quantitative real-time PCR (qRT-PCR) was employed to examine the expression of genes associated with macrophage polarization. The results indicated an increase in macrophage infiltration and M1 polarization in mice subjected to corneal alkali injury. Sinomenine treatment effectively inhibited the polarization of M1 macrophages in the cornea. In mice treated with Sinomenine post-injury, there was a reduction in the expression of the m6A methylation writer protein METTL3 in macrophages, which led to an upregulation of IRAKM expression. The elevated IRAKM expression subsequently inhibited the TLR4 inflammatory pathway and reduced corneal inflammatory cell infiltration, ultimately ameliorating corneal alkali burns. Sinomenine attenuates M1 macrophage polarization through upregulating IRAKM by inhibiting the expression of the m6A methylation writer protein METTL3 in macrophages, leading to enhanced outcomes in corneal alkali injuries.</p></div>","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 5","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145256785","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}
Exosomes, nanosized extracellular vesicles ranging from 30 to 150 nm, have gained increasing attention as mediators of cell-to-cell communication. Within the islet microenvironment, exosomes mediate crosstalk among β-cells, immune cells, and endothelial cells, helping maintain islet integrity, modulate immune responses, and influence the progression of type 1 and type 2 diabetes. Because of their intrinsic role in cellular communication, exosomes are being explored as potential therapeutic tools. Engineered exosomes can be tailored to transport bioactive molecules, including insulin, peptides, or anti-inflammatory agents, directly to pancreatic cells. Such targeted delivery may enhance glycemic control while limiting immune-mediated β-cell destruction. Beyond therapy, exosomes are also being investigated as biomarkers, as their molecular cargo reflects disease-specific alterations, offering opportunities for early diagnosis and timely intervention. This review further examines the scope of exosome-based diagnostics and therapeutics, including advances in exosome engineering and stem cell–derived exosomal applications. Compared with conventional systems, exosomes offer superior targeting, fewer off-target effects, and low immunogenicity due to their natural biocompatibility. These attributes position exosomal therapy as a promising avenue for the development of personalized strategies in diabetes management. In addition, novel findings on exosomal microRNAs, proteins, and lipid components involved in β-cell survival, insulin signaling pathways, and islet inflammation are summarized. Together, these insights highlight the emerging relevance of exosome biology in understanding diabetes pathogenesis and shaping innovative therapeutic approaches.
{"title":"Exosomes in pancreatic islet biology and diabetes: Mechanisms, Biomarkers, and potential therapeutic perspectives","authors":"Venkatesan Karthick, Rajkumar Thamarai, Singamoorthy Amalraj, Mani Suganya, Panneerselvam Suganya","doi":"10.1007/s10735-025-10631-z","DOIUrl":"10.1007/s10735-025-10631-z","url":null,"abstract":"<div><p>Exosomes, nanosized extracellular vesicles ranging from 30 to 150 nm, have gained increasing attention as mediators of cell-to-cell communication. Within the islet microenvironment, exosomes mediate crosstalk among β-cells, immune cells, and endothelial cells, helping maintain islet integrity, modulate immune responses, and influence the progression of type 1 and type 2 diabetes. Because of their intrinsic role in cellular communication, exosomes are being explored as potential therapeutic tools. Engineered exosomes can be tailored to transport bioactive molecules, including insulin, peptides, or anti-inflammatory agents, directly to pancreatic cells. Such targeted delivery may enhance glycemic control while limiting immune-mediated β-cell destruction. Beyond therapy, exosomes are also being investigated as biomarkers, as their molecular cargo reflects disease-specific alterations, offering opportunities for early diagnosis and timely intervention. This review further examines the scope of exosome-based diagnostics and therapeutics, including advances in exosome engineering and stem cell–derived exosomal applications. Compared with conventional systems, exosomes offer superior targeting, fewer off-target effects, and low immunogenicity due to their natural biocompatibility. These attributes position exosomal therapy as a promising avenue for the development of personalized strategies in diabetes management. In addition, novel findings on exosomal microRNAs, proteins, and lipid components involved in β-cell survival, insulin signaling pathways, and islet inflammation are summarized. Together, these insights highlight the emerging relevance of exosome biology in understanding diabetes pathogenesis and shaping innovative therapeutic approaches.</p></div>","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 5","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145256786","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}
Pub Date : 2025-10-08DOI: 10.1007/s10735-025-10619-9
Shaima M. Almasry, Rasha A. Elmansy, Amr K. Elfayomy, Samy A Algaidi
{"title":"Correction: Ultrastructure alteration of decidual natural killer cells in women with unexplained recurrent miscarriage: a possible association with impaired decidual vascular remodelling","authors":"Shaima M. Almasry, Rasha A. Elmansy, Amr K. Elfayomy, Samy A Algaidi","doi":"10.1007/s10735-025-10619-9","DOIUrl":"10.1007/s10735-025-10619-9","url":null,"abstract":"","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 5","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145249199","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}
Pub Date : 2025-10-07DOI: 10.1007/s10735-025-10607-z
Yong Xu, Yanli Wang
Acute myeloid leukemia (AML) is a highly heterogeneous malignancy originating from prolonged abnormal proliferation of immature myeloid cells. Long non-coding RNAs (lncRNAs) are important regulators of AML progression. TMEM147-AS1 has been reported to be abnormally expressed in AML. This study aimed to further explore the roles of TMEM147-AS1 in AML and the possible mechanism. The expression of TMEM147-AS1 and miR-873-3p and its role in the clinical progression of AML were investigated. The diagnostic and prognostic value of TMEM147-AS1 in AML was assessed. The impacts of TMEM147-AS1 on AML cell function were examined. And the targeting relationship among TMEM147-AS1, miR-873-3p, and ZFX was predicted by databases and verified by dual-luciferase reporter assay. TMEM147-AS1 was upregulated in AML tissues and cell lines. High TMEM147-AS1 expression was significantly related to adverse karyotype, shorter overall survival time, and worse prognosis in AML patients. And TMEM147-AS1 can distinguish AML patients from healthy individuals with relatively high sensitivity and specificity. Furthermore, TMEM147-AS1 targeted miR-873-3p, which further targeted ZFX. TMEM147-AS1 promoted proliferation and inhibited apoptosis of AML cells through downregulating the inhibitory effect of miR-873-3p on ZFX expression. TMEM147-AS1 promotes AML progression via regulating the miR-873-3p/ZFX axis. TMEM147-AS1 is a promising diagnostic and prognostic indicator in AML.
{"title":"lncRNA TMEM147-AS1 promotes acute myeloid leukemia development by regulating miR-873-3p/ZFX axis","authors":"Yong Xu, Yanli Wang","doi":"10.1007/s10735-025-10607-z","DOIUrl":"10.1007/s10735-025-10607-z","url":null,"abstract":"<div><p>Acute myeloid leukemia (AML) is a highly heterogeneous malignancy originating from prolonged abnormal proliferation of immature myeloid cells. Long non-coding RNAs (lncRNAs) are important regulators of AML progression. TMEM147-AS1 has been reported to be abnormally expressed in AML. This study aimed to further explore the roles of TMEM147-AS1 in AML and the possible mechanism. The expression of TMEM147-AS1 and miR-873-3p and its role in the clinical progression of AML were investigated. The diagnostic and prognostic value of TMEM147-AS1 in AML was assessed. The impacts of TMEM147-AS1 on AML cell function were examined. And the targeting relationship among TMEM147-AS1, miR-873-3p, and ZFX was predicted by databases and verified by dual-luciferase reporter assay. TMEM147-AS1 was upregulated in AML tissues and cell lines. High TMEM147-AS1 expression was significantly related to adverse karyotype, shorter overall survival time, and worse prognosis in AML patients. And TMEM147-AS1 can distinguish AML patients from healthy individuals with relatively high sensitivity and specificity. Furthermore, TMEM147-AS1 targeted miR-873-3p, which further targeted ZFX. TMEM147-AS1 promoted proliferation and inhibited apoptosis of AML cells through downregulating the inhibitory effect of miR-873-3p on ZFX expression. TMEM147-AS1 promotes AML progression via regulating the miR-873-3p/ZFX axis. TMEM147-AS1 is a promising diagnostic and prognostic indicator in AML.</p></div>","PeriodicalId":650,"journal":{"name":"Journal of Molecular Histology","volume":"56 5","pages":""},"PeriodicalIF":2.2,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145237652","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}