Pub Date : 2024-12-01Epub Date: 2024-10-04DOI: 10.3892/ijmm.2024.5433
Xidan Zhu, Gang Tian, Jing Quan, Peng He, Jinbo Liu
Following the publication of the above article, an interested reader drew to the authors' attention that, with the 'Adjacent' row (top row) of immunohistochemical images shown in Fig. 2 on p. 646, the fourth and fifth panels along (the 'RAB11A' and 'RAB9A' data panels) contained an overlapping section of data, such that data which were intended to show the results from differently performed experiments had apparently been derived from the same original source. After consulting their original data, the authors were able to determine that the duplication of these panels had inadvertently occurred during the process of compiling Fig. 2. The revised version of Fig. 2, featuring the correct data for the 'Adjacent/RAB9A' experiment, is shown below. The authors confirm that the error associated with this figure did not have any significant impact on either the results or the conclusions reported in this study, and are grateful to the Editor of International Journal of Molecular Medicine for allowing them the opportunity to publish this Corrigendum. Furthermore, they apologize to the readership of the Journal for any inconvenience caused. [International Journal of Molecular Medicine 44: 643‑651, 2019; DOI: 10.3892/ijmm.2019.4213].
{"title":"[Corrigendum] Effects of miR‑340 overexpression and knockdown on the proliferation and metastasis of NSCLC cell lines.","authors":"Xidan Zhu, Gang Tian, Jing Quan, Peng He, Jinbo Liu","doi":"10.3892/ijmm.2024.5433","DOIUrl":"10.3892/ijmm.2024.5433","url":null,"abstract":"<p><p>Following the publication of the above article, an interested reader drew to the authors' attention that, with the 'Adjacent' row (top row) of immunohistochemical images shown in Fig. 2 on p. 646, the fourth and fifth panels along (the 'RAB11A' and 'RAB9A' data panels) contained an overlapping section of data, such that data which were intended to show the results from differently performed experiments had apparently been derived from the same original source. After consulting their original data, the authors were able to determine that the duplication of these panels had inadvertently occurred during the process of compiling Fig. 2. The revised version of Fig. 2, featuring the correct data for the 'Adjacent/RAB9A' experiment, is shown below. The authors confirm that the error associated with this figure did not have any significant impact on either the results or the conclusions reported in this study, and are grateful to the Editor of <i>International Journal of Molecular Medicine</i> for allowing them the opportunity to publish this Corrigendum. Furthermore, they apologize to the readership of the Journal for any inconvenience caused. [International Journal of Molecular Medicine 44: 643‑651, 2019; DOI: 10.3892/ijmm.2019.4213].</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448558/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142371793","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-10-04DOI: 10.3892/ijmm.2024.5434
Yong Yuan, Huang Huang, Tie Hu, Chenchao Zou, Yamei Qiao, Ming Fang, Jichun Liu, Songqing Lai
The early restoration of hemodynamics/reperfusion in acute myocardial infarction (AMI) is an effective therapeutic strategy to reduce sudden death and improve patient prognosis. However, reperfusion induces additional cardiomyocyte damage and cardiac tissue dysfunction. In this context, turmeric‑derived curcumin (Cur) has been shown to exhibit a protective effect against myocardial ischemia/reperfusion injury (I/RI). The molecular mechanism of its activity, however, remains unclear. The current study investigated the protective effect of Cur and its molecular mechanism via invitro experiments. The Cell Counting Kit‑8 and lactate dehydrogenase (LDH) assay kit were used to assess the cell viability and cytotoxicity. The contents of malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase, glutathione (GSH)/glutathione disulfide (GSSG), total iron, ferrous iron, caspase‑3 and reactive oxygen species (ROS) were measured using an appropriate kit. Western blotting was used to detect the expression of relevant proteins. The levels of apoptosis, mitochondrial permeability transition pore (MPTP) opening, and mitochondrial membrane potential (MMP) were detected by flow cytometry. The study findings indicated that anoxia/reoxygenation (A/R) injury significantly decreased cell viability, increased in LDH and caspase‑3 activities, induced ferroptosis, increased apoptosis and overactivated autophagy. However, pretreatment with Cur or ferrostatin‑1 (Fer‑1, a ferroptosis inhibitor) significantly increased A/R‑reduced cell viability, SOD, glutathione peroxidase activity, GSH/GSSH ratio and HES1 and glutathione peroxidase 4 protein expression; attenuated A/R‑induced LDH, MDA, total iron, ferrous iron, prostaglandin‑endoperoxide synthase 2 protein expression and prevented ROS overproduction and MMP loss. In addition, Cur inhibited caspase‑3 activity, upregulated the Bcl‑2/Bax ratio, reduced apoptotic cell number and inhibited MPTP over‑opening. Furthermore, Cur increased P62, LC3II/I, NDUFB8 and UQCRC2 expression and upregulated the p‑AMPK/AMPK ratio. However, erastin (a ferroptosis activator), pAD/HES1‑short hairpin RNA, rapamycin (an autophagy activator) and Compound C (an AMPK inhibitor) blocked the protective effect of Cur. In conclusion, Cur pretreatment inhibited ferroptosis, autophagy overactivation and oxidative stress; improved mitochondrial dysfunction; maintained energy homeostasis; attenuated apoptosis; and ultimately protected the myocardium from A/R injury via increased HES1 expression.
{"title":"Curcumin pretreatment attenuates myocardial ischemia/reperfusion injury by inhibiting ferroptosis, autophagy and apoptosis via HES1.","authors":"Yong Yuan, Huang Huang, Tie Hu, Chenchao Zou, Yamei Qiao, Ming Fang, Jichun Liu, Songqing Lai","doi":"10.3892/ijmm.2024.5434","DOIUrl":"10.3892/ijmm.2024.5434","url":null,"abstract":"<p><p>The early restoration of hemodynamics/reperfusion in acute myocardial infarction (AMI) is an effective therapeutic strategy to reduce sudden death and improve patient prognosis. However, reperfusion induces additional cardiomyocyte damage and cardiac tissue dysfunction. In this context, turmeric‑derived curcumin (Cur) has been shown to exhibit a protective effect against myocardial ischemia/reperfusion injury (I/RI). The molecular mechanism of its activity, however, remains unclear. The current study investigated the protective effect of Cur and its molecular mechanism via <i>in</i> <i>vitro</i> experiments. The Cell Counting Kit‑8 and lactate dehydrogenase (LDH) assay kit were used to assess the cell viability and cytotoxicity. The contents of malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase, glutathione (GSH)/glutathione disulfide (GSSG), total iron, ferrous iron, caspase‑3 and reactive oxygen species (ROS) were measured using an appropriate kit. Western blotting was used to detect the expression of relevant proteins. The levels of apoptosis, mitochondrial permeability transition pore (MPTP) opening, and mitochondrial membrane potential (MMP) were detected by flow cytometry. The study findings indicated that anoxia/reoxygenation (A/R) injury significantly decreased cell viability, increased in LDH and caspase‑3 activities, induced ferroptosis, increased apoptosis and overactivated autophagy. However, pretreatment with Cur or ferrostatin‑1 (Fer‑1, a ferroptosis inhibitor) significantly increased A/R‑reduced cell viability, SOD, glutathione peroxidase activity, GSH/GSSH ratio and HES1 and glutathione peroxidase 4 protein expression; attenuated A/R‑induced LDH, MDA, total iron, ferrous iron, prostaglandin‑endoperoxide synthase 2 protein expression and prevented ROS overproduction and MMP loss. In addition, Cur inhibited caspase‑3 activity, upregulated the Bcl‑2/Bax ratio, reduced apoptotic cell number and inhibited MPTP over‑opening. Furthermore, Cur increased P62, LC3II/I, NDUFB8 and UQCRC2 expression and upregulated the p‑AMPK/AMPK ratio. However, erastin (a ferroptosis activator), pAD/HES1‑short hairpin RNA, rapamycin (an autophagy activator) and Compound C (an AMPK inhibitor) blocked the protective effect of Cur. In conclusion, Cur pretreatment inhibited ferroptosis, autophagy overactivation and oxidative stress; improved mitochondrial dysfunction; maintained energy homeostasis; attenuated apoptosis; and ultimately protected the myocardium from A/R injury via increased HES1 expression.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11517743/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142371794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-10-04DOI: 10.3892/ijmm.2024.5432
Jae Hoon Song, Sun Jeong Kim, Soojin Kwon, Su Yeon Jeon, Sang Eon Park, Suk-Joo Choi, Soo-Young Oh, Hong Bae Jeon, Jong Wook Chang
Adipose tissue engraftment has become a promising strategy in the field of regenerative surgery; however, there are notable challenges associated with it, such as resorption of 50‑90% of the transplanted fat or cyst formation due to fat necrosis after fat transplantation. Therefore, identifying novel materials or methods to improve the engraftment efficiency is crucial. The present study investigated the effects of nervonic acid (NA), a monounsaturated very long‑chain fatty acid, on adipogenesis and fat transplantation, as well as its underlying mechanisms. To assess this, NA was used to treat cells during adipogenesis in vitro, and the expression levels of markers, including PPARγ and CEBPα, and signaling molecules were detected through reverse transcription‑quantitative PCR and western blotting. In addition, NA was mixed with fat grafts in in vivo fat transplantation, followed by analysis through Oil Red O staining, hematoxylin & eosin staining and immunohistochemistry. It was demonstrated that NA treatment accelerated adipogenesis through activation of the Akt/mTOR pathway and inhibition of Wnt signaling. NA treatment enriched the expression of Akt/mTOR signaling‑related genes, and increased the expression of genes involved in angiogenesis and fat differentiation in human mesenchymal stem cells (MSCs). Additionally, NA effectively improved the outcome of adipose tissue engraftment in mice. Treatment of grafts with NA at transplantation reduced the resorption of transplanted fat and increased the proportion of perilipin‑1+ adipocytes with a lower portion of vacuoles in mice. Moreover, the NA‑treated group exhibited a reduced pro‑inflammatory response and had more CD31+ vessel structures, which were relatively evenly distributed among viable adipocytes, facilitating successful engraftment. In conclusion, the present study demonstrated that NA may not only stimulate adipogenesis by regulating signaling pathways in human MSCs, but could improve the outcome of fat transplantation by reducing inflammation and stimulating angiogenesis. It was thus hypothesized that NA could serve as an adjuvant strategy to enhance fat engraftment in regenerative surgery.
{"title":"Nervonic acid improves fat transplantation by promoting adipogenesis and angiogenesis.","authors":"Jae Hoon Song, Sun Jeong Kim, Soojin Kwon, Su Yeon Jeon, Sang Eon Park, Suk-Joo Choi, Soo-Young Oh, Hong Bae Jeon, Jong Wook Chang","doi":"10.3892/ijmm.2024.5432","DOIUrl":"10.3892/ijmm.2024.5432","url":null,"abstract":"<p><p>Adipose tissue engraftment has become a promising strategy in the field of regenerative surgery; however, there are notable challenges associated with it, such as resorption of 50‑90% of the transplanted fat or cyst formation due to fat necrosis after fat transplantation. Therefore, identifying novel materials or methods to improve the engraftment efficiency is crucial. The present study investigated the effects of nervonic acid (NA), a monounsaturated very long‑chain fatty acid, on adipogenesis and fat transplantation, as well as its underlying mechanisms. To assess this, NA was used to treat cells during adipogenesis <i>in vitro</i>, and the expression levels of markers, including PPARγ and CEBPα, and signaling molecules were detected through reverse transcription‑quantitative PCR and western blotting. In addition, NA was mixed with fat grafts in <i>in vivo</i> fat transplantation, followed by analysis through Oil Red O staining, hematoxylin & eosin staining and immunohistochemistry. It was demonstrated that NA treatment accelerated adipogenesis through activation of the Akt/mTOR pathway and inhibition of Wnt signaling. NA treatment enriched the expression of Akt/mTOR signaling‑related genes, and increased the expression of genes involved in angiogenesis and fat differentiation in human mesenchymal stem cells (MSCs). Additionally, NA effectively improved the outcome of adipose tissue engraftment in mice. Treatment of grafts with NA at transplantation reduced the resorption of transplanted fat and increased the proportion of perilipin‑1<sup>+</sup> adipocytes with a lower portion of vacuoles in mice. Moreover, the NA‑treated group exhibited a reduced pro‑inflammatory response and had more CD31<sup>+</sup> vessel structures, which were relatively evenly distributed among viable adipocytes, facilitating successful engraftment. In conclusion, the present study demonstrated that NA may not only stimulate adipogenesis by regulating signaling pathways in human MSCs, but could improve the outcome of fat transplantation by reducing inflammation and stimulating angiogenesis. It was thus hypothesized that NA could serve as an adjuvant strategy to enhance fat engraftment in regenerative surgery.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11517738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142371796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-09-20DOI: 10.3892/ijmm.2024.5429
Qian Liu, Jiajia Li, Xin Li, Li Zhang, Shun Yao, Yongfeng Wang, Biguang Tuo, Hai Jin
Liver fibrosis is a pathophysiologic manifestation of chronic liver disease and a precursor to cirrhosis and hepatocellular carcinoma. Glycolysis provides intermediate metabolites as well as energy support for cell proliferation and phenotypic transformation in liver fibers. 6‑Phosphofructo‑2‑kinase/fructose‑2,6‑bisphosphatase 3 (PFKFB3) is a key activator of glycolysis and plays an important role in the process of glycolysis. The role of PFKFB3‑mediated glycolysis in myocardial fibrosis, renal fibrosis and pulmonary fibrosis has been demonstrated, and the role of PFKFB3 in the activation of hepatic stellate cells by aerobic glycolysis has been proven by relevant experiments. The present study reviews the research progress on the role and mechanism of action of PFKFB3‑mediated glycolysis in the progression of hepatic fibrosis to discuss the role of PFKFB3‑mediated glycolysis in hepatic fibrosis and to provide new ideas for research on PFKFB3 as a target for the treatment of hepatic fibrosis.
{"title":"Advances in the understanding of the role and mechanism of action of PFKFB3‑mediated glycolysis in liver fibrosis (Review).","authors":"Qian Liu, Jiajia Li, Xin Li, Li Zhang, Shun Yao, Yongfeng Wang, Biguang Tuo, Hai Jin","doi":"10.3892/ijmm.2024.5429","DOIUrl":"10.3892/ijmm.2024.5429","url":null,"abstract":"<p><p>Liver fibrosis is a pathophysiologic manifestation of chronic liver disease and a precursor to cirrhosis and hepatocellular carcinoma. Glycolysis provides intermediate metabolites as well as energy support for cell proliferation and phenotypic transformation in liver fibers. 6‑Phosphofructo‑2‑kinase/fructose‑2,6‑bisphosphatase 3 (PFKFB3) is a key activator of glycolysis and plays an important role in the process of glycolysis. The role of PFKFB3‑mediated glycolysis in myocardial fibrosis, renal fibrosis and pulmonary fibrosis has been demonstrated, and the role of PFKFB3 in the activation of hepatic stellate cells by aerobic glycolysis has been proven by relevant experiments. The present study reviews the research progress on the role and mechanism of action of PFKFB3‑mediated glycolysis in the progression of hepatic fibrosis to discuss the role of PFKFB3‑mediated glycolysis in hepatic fibrosis and to provide new ideas for research on PFKFB3 as a target for the treatment of hepatic fibrosis.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448561/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142286479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-10-04DOI: 10.3892/ijmm.2024.5435
Johnny Cheuk-On Tang, Dessy Chan, Po-Yee Chung, Yijiang Liu, Alfred King-Yin Lam, Simon Law, Wolin Huang, Albert Sun-Chi Chan, Kim-Hung Lam, Yuanyuan Zhou
Esophageal squamous cell carcinoma (ESCC) is a particularly aggressive form of cancer with high mortality. In the present study, a novel 8‑hydroxyquinoline derivative (91b1) was investigated for its anticancer activities in ESCC along with its associated mechanisms. The in vitro cytotoxic effect of 91b1 were evaluated across five ESCC cell lines using MTS assay, with cisplatin serving as a comparative standard. Changes in gene expression profile were identified by cDNA microarray and further validated by qualitative PCR and immunostaining. Additionally, protein levels of the most notably downregulated target in archival ESCC samples were also studied. 91b1 demonstrated comparable anticancer effect with cisplatin. Notably, chemokine ligand 5 (Ccl5) was identified as the most substantially downregulated gene, with its suppression at both mRNA and protein expression in ESCC cells, exhibiting a dose‑dependent manner. The recombinant human protein of CCL5 enhanced the invasion of ESCC cells using the Transwell assay. The upregulation of CCL5 protein was also detected in 50% of ESCC cell lines. CCL5 was also overexpressed in 76.9% of ESCC specimens. The overall results indicated that 91b1 could effectively induce anticancer effect on ESCC cells through downregulating CCL5 expression with suppression of tumor invasion. Overall, these findings suggested that 91b1 effectively inhibited ESCC cell proliferation and tumor invasion by downregulating CCL5 expression, highlighting its potential as a therapeutic agent for ESCC treatment.
{"title":"Downregulation of chemokine (C‑C motif) ligand 5 induced by a novel 8‑hydroxyquinoline derivative (91b1) suppresses tumor invasiveness in esophageal carcinoma.","authors":"Johnny Cheuk-On Tang, Dessy Chan, Po-Yee Chung, Yijiang Liu, Alfred King-Yin Lam, Simon Law, Wolin Huang, Albert Sun-Chi Chan, Kim-Hung Lam, Yuanyuan Zhou","doi":"10.3892/ijmm.2024.5435","DOIUrl":"10.3892/ijmm.2024.5435","url":null,"abstract":"<p><p>Esophageal squamous cell carcinoma (ESCC) is a particularly aggressive form of cancer with high mortality. In the present study, a novel 8‑hydroxyquinoline derivative (91b1) was investigated for its anticancer activities in ESCC along with its associated mechanisms. The <i>in vitro</i> cytotoxic effect of 91b1 were evaluated across five ESCC cell lines using MTS assay, with cisplatin serving as a comparative standard. Changes in gene expression profile were identified by cDNA microarray and further validated by qualitative PCR and immunostaining. Additionally, protein levels of the most notably downregulated target in archival ESCC samples were also studied. 91b1 demonstrated comparable anticancer effect with cisplatin. Notably, chemokine ligand 5 (Ccl5) was identified as the most substantially downregulated gene, with its suppression at both mRNA and protein expression in ESCC cells, exhibiting a dose‑dependent manner. The recombinant human protein of CCL5 enhanced the invasion of ESCC cells using the Transwell assay. The upregulation of CCL5 protein was also detected in 50% of ESCC cell lines. CCL5 was also overexpressed in 76.9% of ESCC specimens. The overall results indicated that 91b1 could effectively induce anticancer effect on ESCC cells through downregulating CCL5 expression with suppression of tumor invasion. Overall, these findings suggested that 91b1 effectively inhibited ESCC cell proliferation and tumor invasion by downregulating CCL5 expression, highlighting its potential as a therapeutic agent for ESCC treatment.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11517744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142371795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-10-18DOI: 10.3892/ijmm.2024.5441
Jianyu Wang, Yuankang Zou, Ruili Guan, Shuangshuang Tan, Lihong Su, Zaihua Zhao, Zipeng Cao, Kunyan Jiang, Tao Wang, Gang Zheng
Hypoxic ischemia is the primary cause of brain damage in newborns. Notably, copper supplementation has potential benefits in ischemic brain damage; however, the precise mechanisms underlying this protective effect remain unclear. In the present study, a hypoxic HT22 cell model was developed to examine the mechanism by which copper mitigates hypoxia‑induced oxidative stress. Cell viability was assessed using the Cell Counting Kit‑8 assay, mitochondrial structure was examined with a transmission electron microscope, intracellular ferrous ions and lipid reactive oxygen species levels in HT22 cells were measured using FerroOrange and BODIPY 581/591 C11 staining, copper content was determined using graphite furnace atomic absorption spectroscopy, and gene and protein expression were analyzed by reverse transcription‑quantitative PCR and western blotting. The present findings indicated that hypoxic exposure may lead to reduced cell viability, along with the upregulation of various markers associated with ferroptosis. Furthermore, hypoxia elevated the levels of reactive oxygen species, hydrogen peroxide and malondialdehyde, and decreased the activity of superoxide dismutase 1 (SOD1) in HT22 cells. In addition, the intracellular copper concentration exhibited a notable decrease, while supplementation with an appropriate dose of copper effectively shielded neurons from hypoxia‑induced oxidative stress and ferroptosis, and elevated cell viability in hypoxia‑exposed HT22 cells through the copper chaperone for superoxide dismutase/SOD1/glutathione peroxidase 4 axis. In conclusion, the present study identified a novel function of copper in protecting neurons from oxidative stress and ferroptosis under hypoxic conditions, providing fresh insights into the therapeutic potential of copper in mitigating hypoxia‑induced neuronal injury.
{"title":"Copper supplementation alleviates hypoxia‑induced ferroptosis and oxidative stress in neuronal cells.","authors":"Jianyu Wang, Yuankang Zou, Ruili Guan, Shuangshuang Tan, Lihong Su, Zaihua Zhao, Zipeng Cao, Kunyan Jiang, Tao Wang, Gang Zheng","doi":"10.3892/ijmm.2024.5441","DOIUrl":"10.3892/ijmm.2024.5441","url":null,"abstract":"<p><p>Hypoxic ischemia is the primary cause of brain damage in newborns. Notably, copper supplementation has potential benefits in ischemic brain damage; however, the precise mechanisms underlying this protective effect remain unclear. In the present study, a hypoxic HT22 cell model was developed to examine the mechanism by which copper mitigates hypoxia‑induced oxidative stress. Cell viability was assessed using the Cell Counting Kit‑8 assay, mitochondrial structure was examined with a transmission electron microscope, intracellular ferrous ions and lipid reactive oxygen species levels in HT22 cells were measured using FerroOrange and BODIPY 581/591 C11 staining, copper content was determined using graphite furnace atomic absorption spectroscopy, and gene and protein expression were analyzed by reverse transcription‑quantitative PCR and western blotting. The present findings indicated that hypoxic exposure may lead to reduced cell viability, along with the upregulation of various markers associated with ferroptosis. Furthermore, hypoxia elevated the levels of reactive oxygen species, hydrogen peroxide and malondialdehyde, and decreased the activity of superoxide dismutase 1 (SOD1) in HT22 cells. In addition, the intracellular copper concentration exhibited a notable decrease, while supplementation with an appropriate dose of copper effectively shielded neurons from hypoxia‑induced oxidative stress and ferroptosis, and elevated cell viability in hypoxia‑exposed HT22 cells through the copper chaperone for superoxide dismutase/SOD1/glutathione peroxidase 4 axis. In conclusion, the present study identified a novel function of copper in protecting neurons from oxidative stress and ferroptosis under hypoxic conditions, providing fresh insights into the therapeutic potential of copper in mitigating hypoxia‑induced neuronal injury.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11518577/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that, for the cell migration and invasion assay data shown in Fig. 3A, C, E and G on p. 1843, a number of overlapping data sections were identified such that data which were intended to show the results from differently performed experiments had apparently been derived from the same original sources; moreover, these overlaps were featured in different alignments relative to their matching partners. In addition, other errors had been made during the process of compiling the figures; for example, the authors had overlooked indicating that the protein data shown in Fig. 1F were for β‑catenin. In view of the number of overlapping data panels that were identified in Fig. 3, the Editor of International Journal of Molecular Medicine has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [ International Journal of Molecular Medicine 45: 1838‑1850, 2020; DOI: 10.3892/ijmm.2020.4543].
{"title":"[Retracted] Effects of cisplatin on the proliferation, invasion and apoptosis of breast cancer cells following β‑catenin silencing.","authors":"Xidan Zhu, Jia Feng, Wenguang Fu, Xiaojia Shu, Xue Wan, Jinbo Liu","doi":"10.3892/ijmm.2024.5437","DOIUrl":"10.3892/ijmm.2024.5437","url":null,"abstract":"<p><p>Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that, for the cell migration and invasion assay data shown in Fig. 3A, C, E and G on p. 1843, a number of overlapping data sections were identified such that data which were intended to show the results from differently performed experiments had apparently been derived from the same original sources; moreover, these overlaps were featured in different alignments relative to their matching partners. In addition, other errors had been made during the process of compiling the figures; for example, the authors had overlooked indicating that the protein data shown in Fig. 1F were for β‑catenin. In view of the number of overlapping data panels that were identified in Fig. 3, the Editor of <i>International Journal of Molecular Medicine</i> has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [ International Journal of Molecular Medicine 45: 1838‑1850, 2020; DOI: 10.3892/ijmm.2020.4543].</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11517735/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142400229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-01Epub Date: 2024-10-18DOI: 10.3892/ijmm.2024.5440
Zhou Li, Yao Zhang, Jianhua Lei, Yunxia Wu
Autophagy captures damaged or dysfunctional proteins and organelles through the lysosomal pathway to achieve proper cellular homeostasis. Autophagy possesses distinct characteristics and is given recognized functions in numerous physiological and pathological conditions, such as cancer. Early stage cancer development can be stopped by autophagy. After tumor cells have successfully undergone transformation and progressed to a late stage, the autophagy-mediated system of dynamic degradation and recycling will support cancer cell growth and adaptation to various cellular stress responses while preserving energy homeostasis. In the present study, the dual function that autophagy plays in various oral cancer development contexts and stages, the existing arguments for and against autophagy, and the ways in which autophagy contributes to oral cancer modifications, such as carcinogenesis, drug resistance, invasion, metastasis and self-proliferation, are reviewed. Special attention is paid to the mechanisms and functions of autophagy in oral cancer processes, and the most recent findings on the application of certain conventional drugs or natural compounds as novel agents that modulate autophagy in oral cancer are discussed. Overall, further research is needed to determine the validity and reliability of autophagy promotion and inhibition while maximizing the difficult challenge of increasing cancer suppression to improve clinical outcomes.
{"title":"Autophagy in oral cancer: Promises and challenges (Review).","authors":"Zhou Li, Yao Zhang, Jianhua Lei, Yunxia Wu","doi":"10.3892/ijmm.2024.5440","DOIUrl":"10.3892/ijmm.2024.5440","url":null,"abstract":"<p><p>Autophagy captures damaged or dysfunctional proteins and organelles through the lysosomal pathway to achieve proper cellular homeostasis. Autophagy possesses distinct characteristics and is given recognized functions in numerous physiological and pathological conditions, such as cancer. Early stage cancer development can be stopped by autophagy. After tumor cells have successfully undergone transformation and progressed to a late stage, the autophagy-mediated system of dynamic degradation and recycling will support cancer cell growth and adaptation to various cellular stress responses while preserving energy homeostasis. In the present study, the dual function that autophagy plays in various oral cancer development contexts and stages, the existing arguments for and against autophagy, and the ways in which autophagy contributes to oral cancer modifications, such as carcinogenesis, drug resistance, invasion, metastasis and self-proliferation, are reviewed. Special attention is paid to the mechanisms and functions of autophagy in oral cancer processes, and the most recent findings on the application of certain conventional drugs or natural compounds as novel agents that modulate autophagy in oral cancer are discussed. Overall, further research is needed to determine the validity and reliability of autophagy promotion and inhibition while maximizing the difficult challenge of increasing cancer suppression to improve clinical outcomes.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11518578/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142464651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-01Epub Date: 2024-09-02DOI: 10.3892/ijmm.2024.5420
Xudong Jiu, Wenjie Li, Yang Liu, Lin Liu, Hong Lu
Uveal melanoma (UM) is the most prevalent type of primary intraocular malignancy and is prone to metastasize, particularly to the liver. However, due to the poor understanding of the pathogenesis of UM, effective therapeutic approaches are lacking. As a phenolic compound extracted from grapes, piceatannol (PIC) exhibits anti‑cancer properties. To the best of our knowledge, however, the effects of PIC on UM have not been well investigated. Therefore, in the present study, considering the impact of pyroptosis on modulating cell viability, the mechanism underlying the effects of PIC on UM cell proliferation was explored. The inhibitory effect of PIC on proliferation of UM cells was detected by cell counting kit‑8 assay. Wound healing was used to investigate the effects of PIC on the migration of UM cells. Activity detecting assays were performed to test the apoptosis and oxidant level in UM cells. Western blotting and RT‑qPCR were used to detect the inflammatory and pyroptotic levels of UM cell after PIC treatment. PIC‑treated UM cells were screened by high‑throughput sequencing to detect the differential expression of RNA and differential genes. Si‑TREM2 transfection was used to verify the important role of TREM2 in the effects of PIC. Immunohistochemical staining was used to observe the expressions of TREM2 and GSDMR of tumor in nude mice after PIC administration. PIC effectively inhibited proliferation ability of C918 and Mum‑2b UM cell lines via enhancing apoptosis, as evidenced by enhanced activities of caspase 3 and caspase 9. In addition, treatment of UM cells with PIC attenuated cell migration in a dose‑dependent manner. PIC increased reactive oxygen species levels and suppressed the activity of the antioxidant enzymes superoxide dismutase, glutathione‑S‑transferase, glutathione peroxidase and catalase. PIC inhibited inflammatory responses in C918 cells. PIC treatment upregulated IL‑1β, IL‑18 and Nod‑like receptor protein 3 and downregulated gasdermin D (GSDMD). RNA sequencing results revealed the activation of an unconventional pyroptosis‑associated signaling pathway, namely caspase 3/GSDME signaling, following PIC treatment, which was mediated by triggering receptor expressed on myeloid cells 2 (TREM2) upregulation. As an agonist of TREM2, COG1410‑mediated TREM2 upregulation inhibited proliferation of C918 cells, displaying similar effects to PIC. Furthermore, PIC inhibited tumor growth via regulating the TREM2/caspase 3/GSDME pathway in a mouse model. Collectively, the present study revealed a novel mechanism underlying the inhibitory effects of PIC on UM, providing a potential treatment approach for UM in clinic.
葡萄膜黑色素瘤(UM)是眼内原发性恶性肿瘤中最常见的一种,容易发生转移,尤其是向肝脏转移。然而,由于人们对葡萄膜黑色素瘤的发病机制了解甚少,因此缺乏有效的治疗方法。作为一种从葡萄中提取的酚类化合物,皮脂单酚(PIC)具有抗癌特性。然而,据我们所知,PIC 对 UM 的影响尚未得到很好的研究。因此,在本研究中,考虑到热渗透对调节细胞活力的影响,我们探讨了 PIC 对 UM 细胞增殖的影响机制。通过细胞计数试剂盒-8 检测 PIC 对 UM 细胞增殖的抑制作用。利用伤口愈合来研究 PIC 对 UM 细胞迁移的影响。活性检测试验用于检测 UM 细胞的凋亡和氧化水平。用 Western 印迹法和 RT-qPCR 检测 PIC 处理后 UM 细胞的炎症和裂解水平。通过高通量测序筛选经 PIC 处理的 UM 细胞,以检测 RNA 和差异基因的差异表达。用 Si-TREM2 转染验证 TREM2 在 PIC 作用中的重要作用。免疫组化染色观察了 PIC 给药后裸鼠肿瘤中 TREM2 和 GSDMR 的表达。PIC 通过增强细胞凋亡,有效抑制了 C918 和 Mum-2b UM 细胞株的增殖能力,表现为 caspase 3 和 caspase 9 活性的增强。此外,用 PIC 处理 UM 细胞还能以剂量依赖的方式减少细胞迁移。PIC 增加了活性氧水平,抑制了超氧化物歧化酶、谷胱甘肽-S-转移酶、谷胱甘肽过氧化物酶和过氧化氢酶等抗氧化酶的活性。PIC 可抑制 C918 细胞的炎症反应。PIC 处理可上调 IL-1β、IL-18 和 Nod 样受体蛋白 3,下调 gasdermin D (GSDMD)。RNA 测序结果显示,PIC 处理后激活了一个非常规的与化脓相关的信号通路,即 caspase 3/GSDME 信号通路,该通路由触发髓系细胞上表达的受体 2(TREM2)上调介导。作为 TREM2 的激动剂,COG1410 介导的 TREM2 上调抑制了 C918 细胞的增殖,其效果与 PIC 相似。此外,在小鼠模型中,PIC 通过调节 TREM2/caspase 3/GSDME 通路抑制肿瘤生长。总之,本研究揭示了 PIC 抑制 UM 的新机制,为临床治疗 UM 提供了一种潜在的方法。
{"title":"TREM2, a critical activator of pyroptosis, mediates the anti‑tumor effects of piceatannol in uveal melanoma cells via caspase 3/GSDME pathway","authors":"Xudong Jiu, Wenjie Li, Yang Liu, Lin Liu, Hong Lu","doi":"10.3892/ijmm.2024.5420","DOIUrl":"10.3892/ijmm.2024.5420","url":null,"abstract":"<p><p>Uveal melanoma (UM) is the most prevalent type of primary intraocular malignancy and is prone to metastasize, particularly to the liver. However, due to the poor understanding of the pathogenesis of UM, effective therapeutic approaches are lacking. As a phenolic compound extracted from grapes, piceatannol (PIC) exhibits anti‑cancer properties. To the best of our knowledge, however, the effects of PIC on UM have not been well investigated. Therefore, in the present study, considering the impact of pyroptosis on modulating cell viability, the mechanism underlying the effects of PIC on UM cell proliferation was explored. The inhibitory effect of PIC on proliferation of UM cells was detected by cell counting kit‑8 assay. Wound healing was used to investigate the effects of PIC on the migration of UM cells. Activity detecting assays were performed to test the apoptosis and oxidant level in UM cells. Western blotting and RT‑qPCR were used to detect the inflammatory and pyroptotic levels of UM cell after PIC treatment. PIC‑treated UM cells were screened by high‑throughput sequencing to detect the differential expression of RNA and differential genes. Si‑TREM2 transfection was used to verify the important role of TREM2 in the effects of PIC. Immunohistochemical staining was used to observe the expressions of TREM2 and GSDMR of tumor in nude mice after PIC administration. PIC effectively inhibited proliferation ability of C918 and Mum‑2b UM cell lines via enhancing apoptosis, as evidenced by enhanced activities of caspase 3 and caspase 9. In addition, treatment of UM cells with PIC attenuated cell migration in a dose‑dependent manner. PIC increased reactive oxygen species levels and suppressed the activity of the antioxidant enzymes superoxide dismutase, glutathione‑S‑transferase, glutathione peroxidase and catalase. PIC inhibited inflammatory responses in C918 cells. PIC treatment upregulated IL‑1β, IL‑18 and Nod‑like receptor protein 3 and downregulated gasdermin D (GSDMD). RNA sequencing results revealed the activation of an unconventional pyroptosis‑associated signaling pathway, namely caspase 3/GSDME signaling, following PIC treatment, which was mediated by triggering receptor expressed on myeloid cells 2 (TREM2) upregulation. As an agonist of TREM2, COG1410‑mediated TREM2 upregulation inhibited proliferation of C918 cells, displaying similar effects to PIC. Furthermore, PIC inhibited tumor growth via regulating the TREM2/caspase 3/GSDME pathway in a mouse model. Collectively, the present study revealed a novel mechanism underlying the inhibitory effects of PIC on UM, providing a potential treatment approach for UM in clinic.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11410308/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142107178","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-01Epub Date: 2024-09-02DOI: 10.3892/ijmm.2024.5419
Divya Sankar, Iyyappan Ramalakshmi Oviya
Anaemia is a common health problem worldwide that disproportionately affects vulnerable groups, such as children and expectant mothers. It has a variety of underlying causes, some of which are genetic. A comprehensive strategy combining physical examination, laboratory testing (for example, a complete blood count), and molecular tools for accurate identification is required for diagnosis. With nearly 400 varieties of anaemia, accurate diagnosis remains a challenging task. Red blood cell abnormalities are largely caused by genetic factors, which means that a thorough understanding requires interpretation at the molecular level. As a result, precision medicine has become a key paradigm, utilising artificial intelligence (AI) techniques, such as deep learning and machine learning, to improve prognostic evaluation, treatment prediction, and diagnostic accuracy. Furthermore, exploring the immunomodulatory role of vitamin D along with biomarker‑based molecular techniques offers promising avenues for insight into anaemia's pathophysiology. The intricacy of aplastic anaemia makes it particularly noteworthy as a topic deserving of concentrated molecular research. Given the complexity of anaemia, an integrated strategy integrating clinical, laboratory, molecular, and AI techniques shows a great deal of promise. Such an approach holds promise for enhancing global anaemia management options in addition to advancing our understanding of the illness.
贫血是全球常见的健康问题,对儿童和孕妇等弱势群体的影响尤为严重。造成贫血的原因多种多样,其中有些是遗传因素。诊断时需要采取综合策略,结合体格检查、实验室检测(如全血细胞计数)和分子工具进行准确鉴定。贫血症有近 400 种,准确诊断仍是一项具有挑战性的任务。红细胞异常在很大程度上是由遗传因素引起的,这意味着要想彻底了解,就必须从分子层面进行解读。因此,利用深度学习和机器学习等人工智能(AI)技术来改善预后评估、治疗预测和诊断准确性已成为精准医疗的重要范式。此外,探索维生素 D 的免疫调节作用以及基于生物标志物的分子技术,为深入了解贫血的病理生理学提供了前景广阔的途径。再生障碍性贫血的复杂性使其成为一个值得集中进行分子研究的课题。鉴于贫血症的复杂性,将临床、实验室、分子和人工智能技术相结合的综合战略大有可为。除了增进我们对这种疾病的了解之外,这种方法还有望改善全球贫血症的管理方案。
{"title":"Multidisciplinary approaches to study anaemia with special mention on aplastic anaemia (Review).","authors":"Divya Sankar, Iyyappan Ramalakshmi Oviya","doi":"10.3892/ijmm.2024.5419","DOIUrl":"10.3892/ijmm.2024.5419","url":null,"abstract":"<p><p>Anaemia is a common health problem worldwide that disproportionately affects vulnerable groups, such as children and expectant mothers. It has a variety of underlying causes, some of which are genetic. A comprehensive strategy combining physical examination, laboratory testing (for example, a complete blood count), and molecular tools for accurate identification is required for diagnosis. With nearly 400 varieties of anaemia, accurate diagnosis remains a challenging task. Red blood cell abnormalities are largely caused by genetic factors, which means that a thorough understanding requires interpretation at the molecular level. As a result, precision medicine has become a key paradigm, utilising artificial intelligence (AI) techniques, such as deep learning and machine learning, to improve prognostic evaluation, treatment prediction, and diagnostic accuracy. Furthermore, exploring the immunomodulatory role of vitamin D along with biomarker‑based molecular techniques offers promising avenues for insight into anaemia's pathophysiology. The intricacy of aplastic anaemia makes it particularly noteworthy as a topic deserving of concentrated molecular research. Given the complexity of anaemia, an integrated strategy integrating clinical, laboratory, molecular, and AI techniques shows a great deal of promise. Such an approach holds promise for enhancing global anaemia management options in addition to advancing our understanding of the illness.</p>","PeriodicalId":14086,"journal":{"name":"International journal of molecular medicine","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11410310/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142107176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}